Vous êtes sur la page 1sur 800

An Imprint of Elsevier

The Curtis Center


170 S Independence Mall W 300E
Philadelphia, Pennsylvania 19106

Urologic Oncology ISBN: 0-7216-0003-4

Copyright © 2005 by Elsevier Inc. All rights reserved.


No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the publisher. Permissions may be sought directly from Elsevier’s Health Sciences
Rights Department in Philadelphia, PA, USA: phone: (+1) 215 238 7869, fax: (+1) 215 238 2239, e-mail:
healthpermissions@elsevier.com. You may also complete your request on-line via the Elsevier homepage
(http://www.elsevier.com), by selecting “Customer Support” and then “Obtaining Permissions.”

NOTICE

Urologic oncology is an ever-changing field. Standard safety precautions must be followed, but as new
research and clinical experience broaden our knowledge, changes in treatment and drug therapy may become
necessary or appropriate. Readers are advised to check the most current product information provided by the
manufacturer of each drug to be administered to verify the recommended dose, the method and duration of
administration, and contraindications. It is the responsibility of the licensed prescriber, relying on experience
and knowledge of the patient, to determine dosages and the best treatment for each individual patient.
Neither the publisher nor the author assumes any liability for any injury and/or damage to persons or prop-
erty arising from this publication.

Library of Congress Cataloging-in-Publication Data

Urologic oncology / [edited by] Jerome P. Richie, Anthony D’Amico.


p.; cm.
Includes bibliographical references.
ISBN 0-7216-0003-4 (alk. paper)
1. Genitourinary organs–Cancer. I. Richie, Jerome P. II. D’Amico, Anthony V.
[DNLM: 1. Urologic Neoplasms–therapy. 2. Urologic Neoplasms–diagnosis.
WJ 160 U7844 2005]
RC280.U74U75 2005
616.99¢461–dc22
2004045390

Acquisitions Editor: Rebecca Schmidt Gaertner


Editorial Assistant: Suzanne Flint

Printed in the United States of America

Last digit is the print number: 9 8 7 6 5 4 3 2 1


CONTRIBUTORS

SIDNEY C. ABREU, MD RICHARD BIHRLE, MD


Fellow, Section of Laparoscopic and Minimally Invasive Surgery Professor of Urology
Glickman Urological Institute Indiana University School of Medicine
Cleveland Clinic Foundation Indianapolis, Indiana
Cleveland, Ohio 38: Radical Orchiectomy and Retroperitoneal Lymph Node Dissection
13: Laparoscopic Radical and Partial Nephrectomy

ALEX F. ALTHAUSEN, MD FIONA C. BURKHARD, MD


Associate Clinical Professor (Urology), Department of Surgery Department of Urology
Harvard Medical School and Massachusetts General Hospital University of Bern
Cancer Center Bern, Switzerland
Boston, Massachusetts 24: Orthotopic Bladder Substitution in the Male and Female
22: Selective Bladder Preservation by Combined Modality Treatment
MICHAEL C. CARR, MD, PhD
GERALD L. ANDRIOLE, MD Assistant Professor of Urology
Professor, Department of Surgery
University of Pennsylvania School of Medicine
Chief, Division of Urologic Surgery
Philadelphia, Pennsylvania
Washington University School of Medicine;
Director, Prostate Study Center at Barnes-Jewish Hospital 46: Neuroblastoma
St. Louis, Missouri
43: Superficial Carcinoma of the Penis: Management and Prognosis PETER R. CARROLL, MD
Professor and Chair, Department of Urology
DARIUS J. BÄGLI, MD, CM, FRCSC, FAAP University of California, San Francisco
Associate Professor of Surgery, Division of Urology San Francisco, California
The University of Toronto Faculty of Medicine
Toronto, Ontario, Canada 23: Noncontinent and Continent Cutaneous Urinary Diversion
26A: Clinically Localized Adenocarcinoma of the Prostate: (Stage
49: Prepubertal Testicular Tumors T1a-T2c): Surgical Management and Prognosis
GLEN W. BARRISFORD, MD
Resident in Urology WILLIAM J. CATALONA, MD
Brigham and Women’s Hospital Professor of Urology
Harvard Medical School Northwestern University Feinberg School of Medicine
Boston, Massachusetts Chicago, Illinois
32: Complications of Surgical Treatment for Localized Prostatectomy 29: Anatomic Nerve-Sparing Radical Retropubic Prostatectomy
Cancer

JAY S. BELANI, MD XAVIER CATHELINEAU, MD


Resident, Division of Urologic Surgery Professor, Department of Urology
Washington University School of Medicine L’Institut Mutualiste Montsouris
St. Louis, Missouri Paris, France
43: Superficial Carcinoma of the Penis: Management and Prognosis 31: Laparoscopic Radical Prostatectomy

ARIE BELLDEGRUN, MD, FACS


Professor of Urology; SAM S. CHANG, MD
Chief, Division of Urologic Oncology Assistant Professor, Department of Urologic Surgery
David Geffen School of Medicine at UCLA Vanderbilt University School of Medicine
Los Angeles, California Nashville, Tennessee
14: Treatment of Advanced Renal Cell Carcinoma 19: Prognosis and Management of Invasive Transitional Cell Carcinoma

v
vi Contributors

RICHARD CHILDS, MD ANDREW J. DRESLIN, MD


Allogeneic Hematopoietic Cell Transplant Unit, Hematology Resident in Urology
Branch Brigham and Women’s Hospital
National Heart, Lung, and Blood Institute Boston, Massachusetts
National Institutes of Health 16: Management of Upper Urinary Tract Transitional Cell Carcinoma
Bethesda, Maryland
5: Immunotherapy: Basic Guidelines
MICHAEL J. DROLLER, MD
STEVEN J. CHMURA, MD, PhD Professor of Urology
Resident, Department of Radiation and Cellular Oncology Mount Sinai School of Medicine
The University of Chicago Hospitals New York, New York
Chicago, Illinois 17: Diagnosis and Staging of Bladder Cancer
3: Principles and Applications of Radiation Oncology

DONALD S. COFFEY, PhD VICTOR FERLISE, MD


Professor, Oncology, Pharmacology and Molecular Sciences; Instructor of Urology
Director, Research Laboratories University of Pennsylvania School of Medicine
James Buchanan Brady Urological Institution Philadelphia, Pennsylvania
The Johns Hopkins Medical Institutions 39: Retroperitoneal Tumors: Diagnosis, Staging, Surgery, Management,
Baltimore, Maryland and Prognosis
1: The Molecular and Cellular Biology of Urologic Cancers

MICHAEL S. COOKSON, MD ROBERT A. FIGLIN, MD, FACP


Associate Professor, Department of Urologic Surgery Professor of Medicine and Urology
Vanderbilt University School of Medicine David Geffen School of Medicine at UCLA
Nashville, Tennessee Los Angeles, California
19: Prognosis and Management of Invasive Transitional Cell Carcinoma 14: Treatment of Advanced Renal Cell Carcinoma

MAX J. COPPES, MD, PhD, MBA


Head, Division of Paediatric Oncology; ROBERT C. FLANIGAN, MD
Professor, Departments of Oncology and Paediatrics Albert J. Speh, Jr, and Clair R. Speh Professor and Chairperson
University of Calgary Faculty of Medicine Department of Urology
Calgary, Alberta, Canada Stritch School of Medicine
47: Wilms’ Tumor Loyola University
Maywood, Illinois
PATRICK J. CREAVEN, MBBS, PhD 40: Urethral Cancer
Research Professor
School of Medicine & Biomedical Sciences
SUNY Buffalo; RICHARD S. FOSTER, MD
Senior Investigator Professor of Urology
Roswell Park Cancer Institute Indiana University School of Medicine
Buffalo, New York Indianapolis, Indiana
4: Principles of Chemotherapy for Genitourinary Cancer 38: Radical Orchiectomy and Retroperitoneal Lymph Node Dissection

ANTHONY V. D’AMICO, MD, PhD


Professor, Department of Radiation Oncology YVES FRADET, MD, FRCSC
Harvard Medical School; Professor and Chairman, Department of Surgery
Chief, Genitourinary Radiation Oncology Faculty of Medicine
Dana-Farber Cancer Institute Université Laval
Brigham and Women’s Hospital Québec, Canada
Boston, Massachusetts 20: Transurethral Surgery of Bladder Tumors
25: Cancer of the Prostate: Detection and Staging

PHILLIPP DAHM, MD JUDSON R. GASH, MD


Associate Professor, Division of Urology Associate Professor of Radiology
Department of Surgery University of Tennessee
Duke University Medical Center Knoxville, Tennessee
Durham, North Carolina 10: Diagnosis and Staging of Renal Cell Cancer
30: Radical Perineal Prostatectomy

TRACY M. DOWNS, MD INDERBIR S. GILL, MD, MCH


Assistant Professor, Division of Urology Glickman Urological Institute
University of California, San Diego School of Medicine Cleveland Clinic Foundation
La Jolla, California Cleveland, Ohio
23: Noncontinent and Continent Cutaneous Urinary Diversion 13: Laparoscopic Radical and Partial Nephrectomy
Contributors vii

MISOP HAN, MD FREDERICK A. KLEIN, MD


Assistant Professor, Department of Urology Professor and Chairman, Division of Urology
Feinberg School of Medicine Northwestern University University of Tennessee Medical Center
Chicago, Illinois Knoxville, Tennessee
29: Anatomic Nerve-Sparing Radical Retropubic Prostatectomy 10: Diagnosis and Staging of Renal Cell Cancer

J. MATTHEW HASSAN, MD ERIC A. KLEIN, MD


Resident Glickman Urological Institute
Vanderbilt University School of Medicine The Cleveland Clinic Foundation
Nashville, Tennessee Cleveland, Ohio
19: Prognosis and Management of Invasive Transitional Cell Carcinoma 37: Nongerm Cell Tumors of the Testis

BADRINATH R. KONETY, MD, MBA


NIALL M. HENEY, MD, Assistant Professor, Department of Urology
Clinical Assistant Professor of Surgery University of Iowa
Harvard Medical School and Iowa City, Iowa
Massachusetts General Hospital 15: Transitional Cell Carcinoma of the Renal Cell Pelvis and
Boston, Massachusetts Ureter: Evaluation and Treatment
22: Selective Bladder Preservation by Combined Modality Treatment
TRACEY KRUPSKI, MD
WERNER W. HOCHREITER, MD Clinical Instructor
Department of Urology David Geffen School of Medicine at UCLA
University Hospital of Bern Los Angeles, California
Bern, Switzerland 14: Treatment of Advanced Renal Cell Carcinoma
24: Orthotopic Bladder Substitution in the Male and Female
SANJAYA KUMAR, MD
Assistant Professor, Department of Surgery
AVRUM JACOBSON, MD, CM Harvard Medical School;
Urologist Division of Urology
Lakeshore General Hospital Brigham & Women’s Hospital
Montreal, Québec, Canada Boston, Massachusetts
34: Testis Tumors: Diagnosis and Staging 41: Urethrectomy

MICHAEL A.S. JEWETT, MD, FRCSC, FACS LOUIS LACOMBE, MD, FRCSC
Professor, Department of Surgery (Urology) Assistant Professor of Urology, Department of Surgery
University of Toronto Faculty of Medicine
Toronto, Ontario, Canada Université Laval
35: Seminoma: Management and Prognosis Québec, Canada
20: Transurethral Surgery of Bladder Tumors
MICHAEL W. KATTAN, PhD, MD
Associate Professor of Public Health and Biostatistics in Urology PAUL H. LANGE, MD
Cornell University Professor and Chairman
New York, New York Department of Urology
University of Washington Medical Center
25: Cancer of the Prostate: Detection and Staging Seattle, Washington
34: Testis Tumors: Diagnosis and Staging
DONALD S. KAUFMAN, MD
Clinical Professor of Medicine W. ROBERT LEE, MD, MS
Harvard Medical School; Associate Professor and Vice-Chairman
Director, The Claire and John Bertucci Center for Department of Radiation Oncology
Genitourinary Cancers Wake Forest University School of Medicine
Massachusetts General Hospital Winston-Salem, North Carolina
Boston, Massachusetts
26B: Clinically Localized Adenocarcinoma of the Prostate:
22: Selective Bladder Preservation by Combined Modality Treatment Radiation Therapy

HYUNG KIM, MD HOWARD S. LEVIN, MD


Assistant Professor, Department of Urology Staff, Department of Anatomic Pathology
Roswell Park Cancer Center The Cleveland Clinic Foundation
Buffalo, New York Cleveland, Ohio
14: Treatment of Advanced Renal Cell Carcinoma 37: Nongerm Cell Tumors of the Testis
viii Contributors

W. MARSTON LINEHAN, MD MAXWELL V. MENG, MD


Chief, Urologic Surgery Assistant Professor, Department of Urology
National Institutes of Health University of California, San Francisco
National Cancer Institute/Urologic Oncology Branch San Francisco, California
Bethesda, Maryland 23: Noncontinent and Continent Cutaneous Urinary Diversion; 26A:
5: Immunotherapy: Basic Guidelines Clinically Localized Adenocarcinoma of the Prostate: (Stage T1a-
T2c): Surgical Management and Prognosis
MARK S. LITWIN, MD, MPH
Professor of Urology and Health Services PETER D. METCALFE, MD
David Geffen School of Medicine at UCLA Resident in Urology
Los Angeles, California Dalhousie University
6: Health-Related Quality of Life Issues in Urologic Oncology Halifax, Nova Scotia, Canada
49: Prepubertal Testicular Tumors
KEVIN R. LOUGHLIN, MD, MBA
Professor of Surgery (Urology)
Harvard Medical School;
M. DROR MICHAELSON, MD, PhD
Instructor in Medicine
Senior Surgeon
Harvard Medical School
Brigham and Women’s Hospital
Boston, Massachusetts
Boston, Massachusetts
22: Selective Bladder Preservation by Combined Modality Treatment
42: Squamous Cell Carcinoma of the Penis: Diagnosis and Staging

DONALD F. LYNCH, Jr, MD AARON J. MILBANK, MD


Professor and Chairman, Department of Urology Glickman Urological Institute
Eastern Virginia Medical School and Jones Institute for The Cleveland Clinic Foundation
Reproductive Medicine Cleveland, Ohio
Norfolk, Virginia 37: Nongerm Cell Tumors of the Testis
45: Penectomy and Ilioinguinal Lymphadenectomy
MICHAEL E. MITCHELL, MD
S. BRUCE MALKOWICZ, MD Professor of Urology
Professor of Urology University of Washington School of Medicine;
University of Pennsylvania School of Medicine Chief, Division of Pediatric Urology
Philadelphia, Pennsylvania Children’s Hospital & Regional Medical Center
39: Retroperitoneal Tumors: Diagnosis, Staging, Surgery, Seattle, Washington
Management, and Prognosis 46: Neuroblastoma

MURUGESAN MANOHARAN, MD, FRCS ASHRAF MOSHARAFA, MD


Assistant Professor, Department of Urology Fellow in Urology
University of Miami School of Medicine Indiana University School of Medicine
Miami, Florida Indianapolis, Indiana
18: Superficial Transitional Cell Carcinoma of the Bladder: 38: Radical Orchiectomy and Retroperitoneal Lymph Node Dissection
Management and Prognosis
ANDREW C. NOVICK, MD
FRAY F. MARSHALL, MD Professor of Surgery
Chairman of Urology Cleveland Clinic Lerner College of Medicine;
Emory University School of Medicine Chairman, Glickman Urological Institute
Atlanta, Georgia Cleveland Clinic Foundation
11: Renal Cell Carcinoma: Localized Disease Cleveland, Ohio
12: Surgery of Renal Cell Carcinoma, Including Partial Nephrectomy
MARY FRANCES MCALEER, MD, PhD
Resident, Department of Radiation Oncology WILLIAM K. OH, MD
Thomas Jefferson University Assistant Professor of Medicine
Philadelphia, Pennsylvania Harvard Medical School
27: Regionally Advanced Adenocarcinoma of the Prostate: (T3-4N + Boston, Massachusetts
M0): Management and Prognosis 28: Metastatic Adenocarcinoma of the Prostate

W. SCOTT MCDOUGAL, MD MICHAEL P. O’LEARY, MD


Walter S. Kerr, Jr. Professor of Urology Assistant Professor, Department of Surgery
Harvard Medical School; Harvard Medical School;
Chief, Urology Service Division of Urology
Massachusetts General Hospital Brigham and Women’s Hospital
Boston, Massachusetts Boston, Massachusetts
44: Invasive Carcinoma of the Penis: Management and Prognosis 32: Management of Complications of Radical Prostatectomy Surgery
Contributors ix

KENNETH OGAN, MD MARTIN G. SANDA, MD


Assistant Professor of Urology Visiting Associate Professor at Harvard Medical School,
Emory University School of Medicine Division of Urology;
Atlanta, Georgia Beth Israel Deaconess Medical Center
11: Renal Cell Carcinoma: Localized Disease Boston, Massachusetts
2: Gene Therapy for Urologic Cancer: Basic Principles, Prospects, and
RISHIKESH PANDYA, MCH, DNB, MS Limitations
Fellow, Division of Urology
University of Toronto WILLIAM U. SHIPLEY, MD
Toronto, Ontario, Canada Andres Soriano Professor of Radiation Oncology
35: Seminoma: Management and Prognosis Harvard Medical School;
Head, Genitourinary Oncology Unit
DAVID F. PAULSON, MD Department of Radiation Oncology
Professor of Urology Massachusetts General Hospital
Duke University Medical Center Boston, Massachusetts
Durham, North Carolina 22: Selective Bladder Preservation by Combined Modality Treatment
30: Radical Perineal Prostatectomy
WENDLA SILVERBERG, MD
DAVID F. PENSON, MD, MPH Resident, Department of Radiation and Cellular Oncology
Associate Professor of Urology and Preventive Medicine
The University of Chicago
Keck School of Medicine
Chicago, Illinois
University of Southern California
Los Angeles, California 3: Principles and Applications of Radiation Oncology
6: Health-Related Quality of Life Issues in Urologic Oncology
DONALD G. SKINNER, MD
DAVID I. QUINN, MD Professor and Chairman, Department of Urology
Assistant Professor of Medicine Hanson-White Chair in Medical Research
Keck School of Medicine Keck School of Medicine
University of Southern California University of Southern California
Los Angeles, California Los Angeles, California
4: Principles of Chemotherapy for Genitourinary Cancer 21: Partial and Radical Cystectomy

DEREK RAGHAVAN, MD, PhD JOSEPH A. SMITH, Jr, MD


Professor, Lerner College of Medicine; William L. Bray Professor and Chairman
Chairman and Director, Cleveland Clinic Taussig Cancer Center Department of Urologic Surgery
The Cleveland Clinic Foundation Vanderbilt University School of Medicine
Cleveland, Ohio Nashville, Tennessee
4: Principles of Chemotherapy for Genitourinary Cancer 19: Prognosis and Management of Invasive Transitional Cell Carcinoma

MICHAEL L. RITCHEY, MD HOWARD M. SNYDER, III, MD


Professor of Surgery and Pediatrics; Professor of Urology
Director, Division of Urology University of Pennsylvania School of Medicine
University of Texas Health Science Center Philadelphia, Pennsylvania
Houston Medical Center
Houston, Texas 48: Rhabdomyosarcoma of the Pelvis and Paratesticular Structures
47: Wilms’ Tumor
MARK S. SOLOWAY, MD
RONALD RODRIGUEZ, MD, PhD Professor and Chairman
Assistant Professor, Department of Urology Department of Urology
The Johns Hopkins University University of Miami School of Medicine
Baltimore, Maryland Miami, Florida
2: Gene Therapy for Urologic Cancer: Basic Principles, Prospects, 18: Superficial Transitional Cell Carcinoma of the Bladder:
and Limitations Management and Prognosis

RANDALL G. ROWLAND, MD, PHD GRAEME S. STEELE, MD


Professor and Chief, Division of Urology Assistant Professor of Surgery
University of Kentucky College of Medicine Harvard Medical School and
Lexington, Kentucky Brigham and Women’s Hospital
36: Nonseminomatous Germ Cell Tumors: Management and Boston, Massachusetts
Prognosis 16: Management of Upper Urinary Tract Transitional Cell Carcinoma
x Contributors

JOHN P. STEIN, MD PAMELA UNGER, MD


Associate Professor of Urology Associate Professor of Pathology
Keck School of Medicine Mount Sinai School of Medicine
University of Southern California New York, New York
Los Angeles, California 33: Seminal Vesicles: Diagnosis, Staging, Surgery, Management, and
21: Partial and Radial Cystectomy Prognosis

RICHARD G. STOCK, MD RICHARD K. VALICENTI, MD


Professor of Radiation Oncology Associate Professor of Radiation Oncology
Mount Sinai School of Medicine Thomas Jefferson University
New York, New York Philadelphia, Pennsylvania
33: Seminal Vesicles: Diagnosis, Staging, Surgery, Management, 27: Regionally Advanced Adenocarcinoma of the Prostate: (T3-4N +
and Prognosis M0): Management and Prognosis
NELSON N. STONE, MD
Professor of Urology and Radiation Oncology GUY VALLANCIEN, MD
Mount Sinai School of Medicine Head, Departments of Urology and Nephrology
New York, New York L’Institut Mutualiste Montsouris
33: Seminal Vesicles: Diagnosis, Staging, Surgery, Management, and Paris, France
Prognosis 31: Laparoscopic Radical Prostatectomy

URS E. STUDER, MD CELI VAROL, MD


Chairman Department of Urology
University of Bern; University Hospital of Bern
Director, Department of Urology Bern, Switzerland
University Hospital of Bern 24: Orthotopic Bladder Substitution in the Male and Female
Bern, Switzerland
24: Orthotopic Bladder Substitution in the Male and Female
E. DARRACOTT VAUGHAN, Jr, MD
AGNIESZKA SZOT BARNES, MD, MS Professor
Research Fellow in Prostate Image-Guided Therapy Program Weill Medical College of Cornell University
Department of Radiology New York, New York
Brigham and Women’s Hospital 8: Adrenal Tumors
Boston, Massachusetts
7: Image-Guided Minimally Invasive Therapy JOHANNES VIEWEG, MD
Associate Professor of Urology and Immunology
SHAHIN TABATABAEI, MD Duke University Medical Center
Instructor in Surgery Durham, North Carolina
Harvard Medical School 30: Radical Perineal Prostatectomy
Boston, Massachusetts
44: Invasive Carcinoma of the Penis: Management and Prognosis DONALD VINDIVICH, MD
Senior Researcher, James Buchanan Brady Urological Institution
MIAH-HIANG TAY, MBBS, MRCP The Johns Hopkins Medical Institutions
National Cancer Centre of Singapore Baltimore, Maryland
Singapore
1: The Molecular and Cellular Biology of Urologic Cancers
28: Metastatic Adenocarcinoma of the Prostate

CLARE M.C. TEMPANY, MB, BAO, BCh DAVID S. WANG, MD


Professor, Department of Radiology Assistant Professor of Urology
Harvard Medical School; Boston University School of Medicine
Director, Clinical Magnetic Resonance Imaging Boston, Massachusetts
Brigham and Women’s Hospital 9: Open and Laparoscopic Surgery of Adrenal Tumors
Boston, Massachusetts
7: Image-Guided Minimally Invasive Therapy PADRAIG WARDE, MB, BCh, BAO
Professor, Department of Radiation Oncology
RABI TIGUERT, MD University of Toronto;
Fellow, Urology–Oncology Associate Director, Radiation Medicine Program
Université Laval Princess Margaret Hospital
Québec, Canada Toronto, Ontario, Canada
20: Transurethral Surgery of Bladder Tumors 35: Seminoma: Management and Prognosis
Contributors xi

W. BEDFORD WATERS, MD HSI-YANG WU, MD


Professor of Urology Assistant Professor of Urology
University of Tennessee University of Pittsburgh
Knoxville, Tennessee Pittsburgh, Pennsylvania
10: Diagnosis and Staging of Renal Cell Cancer 48: Rhabdomyosarcoma of the Pelvis and Paratesticular Structures

RALPH R. WEICHSELBAUM, MD JASON B. WYNBERG, MD, FRCSC


Chairman, Department of Radiation and Cellular Oncology Urologic Oncology Fellow
The University of Chicago National Cancer Institute
Chicago, Illinois Bethesda, Maryland
3: Principles and Applications of Radiation Oncology 5: Immunotherapy: Basic Guidelines

RICHARD D. WILLIAMS, MD ANTHONY L. ZIETMAN, MD


Professor and Head, Department of Urology Professor of Radiation Oncology
Rubin H. Flocks Chair Harvard Medical School;
University of Iowa Director of Residency Training for Radiation Oncology
Iowa City, Iowa Massachusetts General Hospital
15: Transitional Cell Carcinoma of the Renal Cell Pelvis and Ureter: Boston, Massachusetts
Evaluation and Treatment 22: Selective Bladder Preservation by Combined Modality Treatment

HOWARD N. WINFIELD, MD
Professor of Urology
Director of Endourology and Minimally Invasive Surgery;
University of Iowa
Iowa City, Iowa
9: Open and Laparoscopic Surgery of Adrenal Tumors
PREFACE

Urologic cancer has become a major public health problem in the United States: an estimated 42%
of all new cancers in men and 16% of cancer deaths in men are a result of urologic cancers.
Although less common in women, bladder and kidney cancer still rank among significant causes
of morbidity and mortality. The impact of urologic cancers on the population and the far-reaching
advances in urologic oncology have provided the impetus for this new textbook, which covers all
aspects of urologic oncology in a concise yet focused fashion.
Major strides have been accomplished in the field of urologic oncology, particularly in the
area of prostate cancer, where an estimated 220,900 new cases were expected in 2003 and a
significant but decreasing number of deaths totaling 31,800. Three sentinel factors have cat-
alyzed the advances in the diagnosis and treatment of prostate cancer: the discovery and
application of serum prostate specific antigen and its isoforms for early detection of prostate
cancer, the development of effective surgical and radiotherapeutic approaches that increase
efficacy and reduce morbidity from prostate cancer, and the utilization of an increasing num-
ber of transrectal ultrasound spring loaded needle guided biopsies to more efficiently diag-
nose prostate cancer in an ambulatory setting.
Urologic oncology has matured from case studies to evidence-based medicine.
Randomized prospective studies in many arenas of urologic oncology have aided the clinician
in decision-making processes. The stratification of patients into various prognostic groups
based on pretreatment factors has allowed accurate comparisons of cancer control outcome
of different modalities of treatment. Urologic oncology has become a multidisciplinary spe-
cialty, with integration of medical oncology and radiation oncology specialists along with
urologic oncologist to provide the most comprehensive treatment options for the patient
with urologic malignancy.
This textbook provides basic principles of medical and surgical urologic oncology. Each
urologic cancer tumor type is reviewed with regard to incidence, etiology, clinical presenta-
tion, diagnosis and staging, treatment options, prognosis, and future directions. Common
surgical procedures for genitourinary cancers are discussed in terms of indications, preoper-
ative preparation, technique, efficacy, and side effects. Both adult and pediatric malignancies,
including neuroblastoma, Wilms’ tumor, testis tumors, and rhabdomyosarcoma, are reviewed
in detail.
Leading authorities in the field have contributed their knowledge and expertise to this
compendium of urologic oncology. The text is supplemented with tables and figures, as well
as a bibliography containing classic references and recent papers published in the urologic
and medical literature. We expect that this textbook will serve as a valuable resource to the
oncologist in need of pertinent information in every aspect of urologic oncology, from basic
principles to treatment to multidisciplinary approaches.

Jerome P. Richie, MD
Anthony V. D’Amico, MD, PhD

xiii
C H A P T E R

1 The Molecular and Cellular Biology


of Urologic Cancers
Donald S. Coffey, PhD, and Donald Vindivich, MD

The focus of this chapter is to provide an overview of anticipated that whatever causes the multimillions of
some of the important concepts and discoveries that have accumulated cases of prostate cancers in the history of
recently revolutionized our understanding of cancer. the world should have produced at least a dozen bul-
This will be accomplished by giving simple schematics bourethral cancers, but not one has ever been reported.
that show the complexity and elegance of the control The same is true for the epididymis. No simple compar-
mechanisms involved in controlling life and how these ative analysis of replication rates, DNA repair, acquired
processes go astray when cancer develops. This will not mutation, inherited gene, or lifestyle and environments
be an extensive review with detailed references but rather seems appropriate to explain this paradox of such a
an overview of the most important and complex medical marked tissue specificity for cancer risk. Something
problems. within the biologic and molecular contexts of the cell
type and differentiation would appear to be involved;
this nongenetic effect is termed an epigenetic event
THE ENIGMAS OF GENITOURINARY CANCERS
(Figure 1-2).
All of our present molecular concepts of the role of
inherited genetics as the sole cause of cancers appear to
EPIGENETIC EFFECTS
be challenged by the tissue specificity of the genitouri-
nary cancers. Each organ inherits the same genome, but Since the same DNA sequence in two different cell types
only certain organs, such as the prostate, bladder and can produce such drastic differences in cancer risk factors,
kidney, are highly prone to form cancers. In contrast, it is apparent that other factors besides just the DNA
other organs in close anatomic proximity are essentially sequence (genetics) may affect this carcinogenesis. This
devoid of any historic presence of a reported cancer, such epigenetic process is usually thought to be related to the
as the epididymis, vas deferens, and bulbourethral glands. maturation of the stem cell to a specific cell type. What is
All of these organs with vast differences in their cancer the molecular basis of epigenetics? The same gene
risk can reside within the same human; thus, they have sequence can produce a different messenger RNA in dif-
the same inheritance, genome, environmental exposure, ferent cell types due to alternative splicing of the RNA.
and have aged to exactly the same time (Figure 1-1). For DNA rearrangements can also alter the message. Recent
example, the bulbourethral gland and the prostate are attention has been focused on alterations in chromatin
both derived from the same developmental anlagen, the structure, which changes the way DNA is wrapped around
urogenital sinus, and they both bud off as adjacent struc- the histone cores to form a nucleosome and alter gene
tures from the developing urethra. Both organs are expression. One hundred and forty-seven base pairs (bp) of
androgen responsive and have a similar blood supply and DNA are coiled twice around each histone octamer that is
nerve stimulation and reside within the same host, shar- made up of two copies each of H2A, H2B, H3, and H4.
ing a common diet, carcinogenic intake, hormonal envi- This tight interaction of the nucleosome with DNA can be
ronments, and identical aging. One would have loosened by acetylation of the histone through the action

3
4 Part I Principles of Urologic Oncology

Figure 1-1 Within a human the prostate and bulbourethral


gland are similar, yet the risk of cancer is astronomically
different. Understanding the molecular basis of this tissue Figure 1-2 An inherited gene that causes cancer is present
specificity is one of the great riddles of cancer. in every cell, but it only produces cancer in a specific type of
cell: this specificity is an epigenetic event dictated by the
context of the cell.

of histone acetyl transferases (HAT) or the DNA nucleo-


some can be tightened by histone deacetylation (HDAC). there are about 3000 different types of these transcrip-
In addition, some of these histones can be methylated, tion factors within a human. It now appears that organ-
which usually occurs on the lysine of histone H3. Histone ism complexity, as well as cellular diversity, may arise
hypoacetylation and H3 methylation both tend to tighten from the diversity of these transcription complexes that
DNA and silent chromatin from being expressed. In addi- form large cellular machinery in a tissue-specific manner
tion, the DNA may be silenced by DNA methylation of self-assembly.1
through methylation of the cytosine in the five positions Once a protein is formed by translation of a specific
(5 mC). These three types of regulations work closely in gene, its turnover rate is regulated by a series of posttrans-
coordination, and can be transferred during propagation lational modifications, such as ubiquination and proteo-
to the daughter cell, and, therefore, can act like pseudoge- some interactions, clipping or glycosylation, etc. Certainly,
netic elements or as termed, an epigenetic event. folding of proteins into various dynamic structures is
A fourth element modifying chromatin structure is essential for their proper action. Many of these proteins
small molecular weight RNA, such as interference RNA are transported to specific cellular sites and can interact in
(RNAi). These and other small RNA molecules are not heterodimers with thousands of other proteins to form
read out into proteins but appear to have powerful regu- complex networks. Understanding these self-assembling
latory abilities in directing chromatin structure, message networks has now become a major frontier of cell biology.2
availability, cell function, and gene function. One of these
small RNAs that are not translated is termed DD3 and is
CARCINOGENS
overexpressed in prostate cancer and may be an impor-
tant control factor or diagnostic agent. What causes genitourinary cancers? Is it mostly environ-
DNA topology involves the winding and unwinding of ment (nurture) or inheritance (nature)? Obviously it is
the double helix and can change the super helical density both, but environment has been understudied. In China,
through the actions of topoisomerases and helices that prostate and breast cancer incidence is less than one-
can greatly alter chromatin structure. These factors are tenth of the incidence rate in the U.S., and this is true of
usually attached at the base of 60,000 molecular weight many other parts of Asia. When Asian populations, with
DNA loops that are attached to the fixed replication low prostate cancer rates, migrate to the U.S., there is a
complex on the nuclear matrix. The nuclear matrix is tis- dramatic increase in their incidence of prostate cancer
sue specific in its protein composition. This loop organ- that occurs by the second generation. This alteration risk
ization represents a much higher order structure of DNA is different with migration and would appear to involve a
and chromatin and is cell type specific and represents one change in environmental exposure or a change in lifestyle
of the frontiers of understanding epigenetic events. risk factors that are beyond a simple Mendelian model of
Five to ten percent of the total proteins made within genetics. It now appears that genes and environment
the cell are transcription factors, and it is estimated that interact in a system we have not resolved, but it can still
Chapter 1 The Molecular and Cellular Biology of Urologic Cancers 5

only be an effective carcinogenic event when it occurs in very common and is often not associated with any symp-
a specific type of cell within the body. It is still unclear toms, and it must be distinguished from prostatitis,
whether this migration to a higher cancer risk area which frequently can be symptomatic. The cause of this
removes a protective agent that was in the Asian environ- hidden prostate inflammation is unknown. It may be
ment or has added a carcinogenic agent in the United related to pathogens, it could also be associated with
States’ environment. autoimmunity. Nevertheless, this close proximity of
With few exceptions, it has been extremely difficult to highly replicating prostate cells in focal proliferative
induce prostate cancer in aging rats. However, if rats are atrophy juxtaposition with inflammatory cells that could
fed with burnt meat, both prostate cancer and breast can- produce high levels of reactive oxygen species (ROS)
cer appear.3 A polycyclic hydrocarbon has been identified could be highly detrimental to the DNA of the prostate
from burnt meat and appears to make adducts to the epithelial cells unless they can protect themselves against
DNA and thus produces mutations. DNA repair cuts out this oxidative onslaught. One of the common mecha-
these aberrant bases, and they appear in the urine. The nisms of protecting against this type of oxidative damage
production of these carcinogens in burnt meat is related is to induce a stress response that up-regulates glu-
to the temperature and time of cooking and might easily tathione-S-transferase pi (GSTPi) that provides a strong
explain many cultural variations in food processing that protection against carcinogens and ROS. These prostate
alter carcinogens. For example, the Chinese eat meat, but cells that are under stress near the inflammation have
they do not burn it excessively in the same way as we do indeed induced their defenses by the induction of this
in the U.S. in our barbecuing and tendencies to sear to GSTPi, however, a few isolated epithelial cells appear to
the point of burning in the preparation of many of our be unable to produce this protective effect. These cells
meats. Obviously, other cultural aspects of diet might are highly prone to DNA damage, and when replicated
also be involved, such as the absence of milk and cheese would then accumulate and be the early events in prosta-
in the diet of Asia in contrast to its heavy use in the U.S. tic interepithelial neoplasia (PIN), a precursor to
and Northern Europe, or the addition of tea and soy in prostate cancer. This model has been proposed and
the Asian diet. tested by Angelo De Marzo, and a recent review dis-
cusses details of the molecular mechanism.4 How is
GSTPi regulated? As stated, the DNA promoter region
INFLAMMATION
of GSTPi is in a CpG-rich island domain located within
Chronic inflammation has recently received tremendous the promoter region. This promoter is silenced by the
interest as an etiologic factor associated with many dis- methylation of the cytosine residues in these CpG
eases appearing during aging, including arteriosclerosis, islands, thus turning off the expression protective effect
arthritis, and now cancer. It has long been recognized of GSTPi for the replicating cells located in the reactive
that schistosomiasis infections in Egypt were associated oxygen environment of the inflammation. The final
with a high incidence of bladder cancer. Now a new prostate cancer cells that form have a hypermethylated
mechanism for inflammation in the formation of prostate CpG island region in the promoter of GSTPi, and this is
cancer is becoming evident.4 This new mechanism may the most common molecular and genome change (>90%)
combine many aspects of genetics and epigenetics that has been reported to be associated with all forms of
defined by lifestyle and risks. prostate cancer. The cellular and molecular events that
In the aging prostate, there appear to be many small can be correlated with the early pathology pathway to
foci of atrophy. Usually, atrophy indicates a dormant prostate cancer have been defined and named prolifera-
state or DNA synthesis that is often seen when andro- tive inflammatory atrophy (PIA).4 It is also of interest
gens are withdrawn. Following androgen ablation the that in animal models of prostate inflammation in the rat,
entire prostate becomes atrophied in relation to its that the inflammation can be suppressed by high levels of
epithelial cells and DNA synthesis and replication essen- soy in the diet; a possible clue to a protective factor in the
tially cease.5 The paradox was that in some prostates in Asian diet.5
humans these small foci of atrophy could be seen in jux- Two genes that have been implicated as candidates in
taposition to acini with highly stimulated luminal epithe- familial human prostate cancer studies and by microarray
lial cells that appeared to be under strong androgen studies are the macrophage scavenger receptor (MSR-1)
stimulation. The paradox of atrophy next to stimulation and RNASEL6 (Table 1-1). These genes have been shown
was resolved in part when it was observed that these atro- to increase infections in knock out animals devoid of these
phied epithelial cells seen in the small foci were actually genes. Thus, the role of pathogens might be implicated in
not dormant for DNA synthesis but were highly stimu- this process, but equal attention should be given at this
lated and were undergoing rapid DNA synthesis. This time to the possibility that this type of inflammation may
proliferative type of atrophy was often associated with be produced by autoimmune reactions. Estrogen imprint-
areas of prostate inflammation. Prostate inflammation is ing in the early neonate life of the rat can result in later
6 Part I Principles of Urologic Oncology

Table 1-1 Selected Genes Proposed to be Involved in Prostate Cancer Initiation or Progression,
or in Modifying the Risk of Prostate Cancer Development
Gene Proposed Function

Mutations causing decreased activity


MS (MSR-1) Antiinfectious, macrophage scavenger receptor
RNASEL Antiinfectious, apoptosis, RNASE
ELAC2 Metal-dependent hydrolase

Promoter hypermethylation resulting in gene silencing


GSTP1 Carcinogen detoxification
EDNRB Endothelin receptor
ER (alpha and beta) Estrogen receptor

LOH and point mutation


PTEN Cell survival and proliferation, phosphatase
TP53 (also p53) Cell survival and proliferation, genome stability
LOH and haploinsufficiency
NKX3-1 Cell differentiation and proliferation
CDKN1B (P27KlP1) Cell proliferation, brake for proliferation

Point mutations
COPEB (also KLR6) Transcription regulation
Androgen receptor (AR) Cell proliferation, survival and differentiation
Amplification
AR Cell proliferation, survival, and differentiation

Overexpressed at mRNA and protein level


HTERT (telomerase) Cell immortality
HPN (Hepsin) Transmembrane protease
FASN Fatty-acid synthesis
AMACR (racemase) Fatty-acid metabolism, branched chain
EZH2 Transcription repressor, cell proliferation
MYC Cell proliferation
BCL2 Cell survival

Polymorphisms affecting prostate cancer risks


AR (CAG and GGC repeats) Cell proliferation, survival, and differentiation
CYP17 Androgen metabolism
SRD5A2 (5 Alpha reductase) Androgen metabolism

Metastasis suppressor (down-regulated)


KAI1 CD44
NME23 KISSI
BRMS1
MAP2K4

development of marked inflammation in the adult rat’s involving single electron transfer resulting in free rad-
prostate if they are carried out in strains that have a high icals that can be highly deleterious to DNA resulting in
propensity for autoimmune disease. specific types of DNA oxidative damage. In addition,
The body’s defense against many infectious insults is nitric oxide (NO), which is formed by NO synthetase
to produce reactive oxygen, which is primarily pro- activity in the metabolism of arginine, produces NO, a
duced by macrophages. Reactive oxygen can also occur powerful oxidizing agent when combined with ROS.
from normal metabolism within the cell and has often GSTPi is a powerful defender against these types of
been suggested as part of the aging process. As hydro- oxidative agents produced by inflammation, as well as
gen from food metabolism interacts with oxygen to protecting against adduct formation on the DNA
make water, they go through a series of oxidation states caused by carcinogens. These free radicals can also be
Chapter 1 The Molecular and Cellular Biology of Urologic Cancers 7

squelched in the presence of vitamin E. Thus, vitamin mismatch DNA repair genes, termed MSH2, MLH1,
E as a free radical scavenger has received popularity as PMS1, PMS2, as well as the APC tumor suppressor gene.
a chemoprotective agent and is being tested in large Two kidney cancer genes have been identified to be
clinical trials. Lipid peroxidation is another mechanism WT1, in the Wilms’ tumor, and the VHL gene associ-
for protecting DNA, and it utilizes selenium in its ated with von Hippel-Lindau syndrome, identified by
action. Marston Linehan and his colleagues.
Inflammation may be involved in the tissue specificity It is most interesting that there is a familial tendency
of cancer. Seminal vesicles, which rarely get cancer, have in both prostate and renal cell cancers but no such famil-
no inflammation like the prostates do. Furthermore, ial tendency has yet been identified for bladder cancer.
inflammation is greatly reduced in the prostates of peo- However, only 10% of many types of cancers are inher-
ple in Asia, and this reduction can be mimicked in ani- ited through a predisposing gene or genes, but 90% of
mals fed diets high in soy. The role of estrogens in cancers are acquired through living or by environmental
inflammation is also an intriguing possibility, particularly insults; these later types are termed sporadic cancers. It is
since the soy diet contains high levels of phytoestrogens believed that if we identify the inherited genes that pre-
that could serve as antiestrogens and block these effects.5 dispose to familial cancer, these genes will be the same
Much is left to discover in these new developing fields of targets that are altered by carcinogens, aging, or biologic
carcinogenesis of the prostate, but there is little doubt damage in sporadic cancers. This is the basis for Knudson’s
that major inroads have been realized in the last 4 years hypothesis, which has been proven in retinoblastoma.
by combining genetic and epigenetic concepts.4 We now Since we inherit two genes, one each from our mother
have models that might help explain why only humans and father, we therefore have two alleles for every gene
and dogs get prostate cancer, why the seminal vesicles are that can be slightly different in sequence (polymorphism)
not at risk, and why the risks are so different in Asia and or in methylation (imprinting). If both genes are required
change with migration.5 Molecular targets are being to be knocked out to produce cancer, which is the case
identified by new microarray techniques, and clinical when a suppressor gene is eliminated, then inheriting the
prevention trails are now underway in many centers. loss of one gene (loss of heterozygosity [LOH]) would
increase your chances of getting cancer, because now an
environmental insult only needs to eliminate the second
FAMILIAL CANCER
allele to inactivate the suppressor gene. This increase in
Cancer clusters in some families could be the result of probability results in the early onset of cancer, because
inherited genetic alterations or shared environmental one of the two suppressor genes had already been inacti-
factors. Single or multiple genes could be causing predis- vated at birth.
position to cancer. Sporadic occurrence suggests aggre- The similarity that approximately 10% of all colon,
gation by the adverse effects of the environment, such as breast, and prostate cancers are inherited, although the
pollutants and carcinogens, as well as socioeconomic dif- overall frequency of these genes in the population is
ferences and cultural habits, such as work, sexual, and about 0.3%, is one of the mysteries in cancer research. Is
dietary factors. It is now possible to apply statistical this similarity by chance or does it have a meaning? In
analysis to these inherited patterns, trying to correlate addition, in each case when you inherit these predispos-
these with the time of diagnosis in order to determine if ing genes you have an approximately 85% chance of get-
there is a genetic predisposition, inherited primarily ting the cancer, but 15% will not get cancer.
through genes from either the mother or the father, One of the difficulties in locating cancer-causing
termed autosomal. In this regard, Patrick C. Walsh and genes is that once you have developed cancer, it is often
his colleagues have reported that there is a hereditary accompanied by a genetic instability, which produces a
form of prostate cancer (HPC) with an early onset and a series of changes in the genome that alter the cancerous
3- to 9-fold increased incidence, depending on the num- properties of the cell. This temporal change in the can-
ber of first-degree relatives involved. Indeed, hereditary cer cell clones is called progression and produces a
prostate cancer appears to be inherited in a Mendelian marked tumor cell heterogeneity. These genetic changes
manner and is autosomal dominant, with a penetrance of that ensue because of this instability can produce a cell
approximately 85%. Intense efforts have been underway with an increased growth rate, and then this clone will
in many centers to find chromosomal linkage to this expand and dominate in the tumor. This phenomenon is
hereditary form of cancer and to identify the gene(s) termed “clonal selection,” and since it occurs with time,
involved in this increased cancer risk.7 It is hoped that it is called tumor progression. Ultimately, cells may be
these high-risk genes can be sequenced as they have been selected with not only increased growth rate but also with
in breast cancer. In breast cancer, BRCA1 and BRCA2 more aggressive properties, and alterations in many can-
have been identified as hereditary genes. Also, in colon cer genes, such as p53, appear to be related to more
cancer, 4 genes have been identified and, 3 of which are aggressive tumors. Therefore, when a tumor is removed
8 Part I Principles of Urologic Oncology

from a patient and the karyotype or DNA is examined, it have a positive effect in the induction of growth.
is possible to see tremendous changes in the chromo- These constitutive genes have a prefix like c-myc. If
somes. There are cases of chromosomal deletions, ampli- they are mutated and inserted by viruses this prefix
fications, rearrangements, and duplications, which can changes to v, like v-src.
result in changes in ploidy and abnormal amounts of 2. Suppressor genes. Loss of the function of a suppressor
DNA in the cancer cell nuclei. This was seen earlier in gene essentially removes a brake on cell growth, thus
what we call karyotyping, but this only looked at the permitting it to become up-regulated (examples are
shapes and forms of chromosomes and their banding. p53, Rb, and p16).
Later it was possible to differentially stain cancer and 3. DNA repair genes. Normal or induced errors in DNA
normal DNA using either red or green markers as probes copying, DNA damage from the environment, or
to stain and differentially compare the chromosomes. By oxidative damage must be corrected or the gene will
looking at the presence or absence of the red and green be mutated or silenced. In colon cancer, a group of
markers on cancer chromosomes, it is possible to visual- mismatch repair genes (MSH2, MLH1, PMS1, and
ize changes in their karyotype. This technique has been PMS2) have all been shown to be inherited and to
termed comparative genome hybridization (CGH). induce cancer by accumulation of DNA damage. We
Recently, all chromosomes can be painted a different know little about how telomere damage is repaired or
color; a process termed spectral karyotyping (SKY). With how repetitive DNA transposons are regulated.
this, it has been possible to find that in certain cancer 4. DNA defense genes. These genes protect the DNA
chromosomes there is a gain in areas on one arm, and a from oxidative damage or electrophiles that can form
simultaneous loss in areas on the other arm. In prostate adducts to the bases that are detrimental. There are
cancer, this occurs with loss of material in the short arm enzymes that protect the cell against ROS that form
(p) and a subsequent gain in the long arm (q) of the free radicals and produce oxidative damage to the
eighth chromosome; this is not a simple transposition. cell. As the mitochondria carry out their aerobic
Some of these changes may be causal for cancer, but oxidation, 4 electrons are required to reduce
many are just associated with the properties of the tumor molecular oxygen to water. In this process, partially
as it progresses and are simply epiphenomena. This pro- reduced intermediates of oxygen produce superoxide,
duces the complex problems that the geneticists face when hydrogen peroxide, and hydroxyl radicals that are
analyzing tumor cell chromosomes and DNA. Therefore, collectively known as ROS. ROS can also be caused
this requires the meticulous linking of the inherited chro- by ionizing radiation, UV light, or certain chemicals
mosome changes within the lineages of the families with in the environment. ROS converts guanine in DNA
the tumor types. These linkage studies are most difficult to 8-oxoguanine, which is highly mutagenic and
and usually require years of work. New molecular probes preferentially mispairs with adenine during
and information from the Human Genome Project are replication. There are enzymes, such as glutathione-
speeding this process, and certainly automated and high- S-transferase (GST), glutathione reductase, quinone
throughput systems are accelerating this search, which has reductase, superoxide dismutase, catalase, and other
been a most difficult problem for cancer research. protective enzymes that inactivate electrophiles,
Certainly, many candidate genes have been identified and carcinogens, and ROS. Carcinogens in our
are being verified or eliminated by painstaking work. environment are often in a pro-form and need to be
The problem then will be to link specific sequences to activated by type 1 enzymes or the active carcinogen
gene functions, gene control, and disease. There will be needs to be inactivated by type 2 enzymes. For
much variation and polymorphism within the population, example, procarcinogens like benzpyrene are inactive
genetic types, and races. Several different types of each and must be metabolized by epoxidases to form the
inherited cancer may exist. The genome can also change active carcinogen that reacts with DNA; this
through aging, replication errors, and failures in DNA represents a type 1 reaction. Type 2 reactions are
repair. This is a complex but critical problem in under- represented by the family of glutathione transferases,
standing genetic changes associated with cancer. Large pop- glucuronosyltransferases, and quinone reductases, all
ulations will be required in these studies to assure accuracy.7 of which can inactivate carcinogens or ROS. Types 1
and 2 enzymes can be induced or altered by
environment, diet, or inheritance, altering the rate of
CANCER GENES
cancer formation. There are several isoforms and
Cancer susceptibility is driven primarily by six types of polymorphisms; for example, GST-M is related to
genes: bladder and glutathione-S-transferase isoforms
(GST-π) methylation to prostate cancer.
1. Oncogenes. A series of over 60 genes have been 5. Viral genes. Retroviruses, polyoma, adenoma, and
identified that are activated or overexpressed and that papilloma viruses can also introduce genes into the
Chapter 1 The Molecular and Cellular Biology of Urologic Cancers 9

mammalian cell, which when expressed induce not have to occur in a specific order, although this model
malignant transformation. This includes large has not been completely confirmed. Certainly, just inher-
T-antigen, E1, E6, and oncogenes. iting one familial cancer gene seems to guarantee the rest
6. DNA methylation genes. DNA methylation is altered of the hits, since there is often an 85% chance of devel-
in many cancers and for unknown reasons. oping cancer when an inherited gene is involved, an
Hypermethylation of CpG islands in promoter effect termed penetrance.
regions can silence genes. DNA methylation can vary How do the aforementioned oncogenes and suppres-
in maternal and paternal genes, termed imprinting. sor genes function within the cell to cause cancer? They
Loss of imprinting (LOI) is a common change in appear to regulate cell replication, death, and growth.
cancers. At present, all of the above six mechanisms There are about 60 oncogenes of primarily four types:
are being studied to determine what causes urologic
cancers. At the moment, there is only definitive 1. Genes for growth factors or their receptors (e.g.,
evidence that the VHL gene is associated with von platelet-derived growth factor [PDGF], erb-B, and
Hippel-Lindau syndrome, and the WT1 gene is RET).
associated with Wilms’ tumor. The p53 gene is 2. Genes affecting cell-signaling pathways, such as ras
associated with bladder cancer but it may only be a and src.
progression marker, as it is in prostate cancer. No 3. Genes acting as transcription factors that activate
specific gene has yet been shown to be inherited in early growth genes, such as the myc oncogenes.
prostate cancer, although practically all of these 4. Genes affecting the cell cycle: Bcl-2 is an inhibitor of
tumors are associated with inactivation of one of the cell death that when overexpressed, blocks apoptosis
GST-π. As stated, this inactivation of expression is and allows cells to survive and accumulate.
accomplished through methylation of the CpG Overexpressing factors that bind to suppressors can
islands in the promoter region, which down-regulates remove the brake. For example, MDM-2 removes
the gene. This genomic change is almost universal in the suppressor brake p53 by binding to it and
both familial and sporadic prostate cancers but is not inactivating it. Many virus proteins are expressed in
believed to be the inherited gene that causes the an infected cell, such as large T, E1A, and E7, and
cancer, but we do not know what controls DNA have the ability to complex suppressor molecules,
methylation. such as p53 and Rb. In summary, turning these genes
on turns on cell growth. Suppressors are brake
Since both aging and cancer produce heterogeneity in molecules that turn growth off. Removing the brake,
the stability of various chromosomes, it is hard to elimi- of course, turns on the growth. These brakes can be
nate this form of noise in the system, without careful removed either by inheriting the loss of this gene, by
study. In addition, many normal genes have different mutating the gene and activating it, or by turning off
DNA sequences, which are called polymorphism. These the gene through regulation, which is the case when
polymorphisms are inherited and can produce different the DNA in its promoter region is methylated.
types of isozymes or genetic patterns that may or may not
have effects on how these genes function. Some of these How do the suppressor genes function as brakes?
polymorphisms are certainly going to increase tendencies Many of these genes are located in the nucleus and affect
towards malignant transformation that would enhance the cell cycle regulation. The Rb gene is present in all
the chances of acquiring cancer, which will add to the cells and codes for a master brake on the cell cycle that is
complexity. Many suppressor genes not only will be lost discussed in the following. The p53 is one of the best-
through mutation or genetic inactivation but can also be known suppressors and is abnormally regulated in most
down-regulated and turned off by nongenetic or epige- cancers. It blocks the cell cycle by inducing a series of cell
netic means, such as DNA methylation. cycle kinase inhibitors. This p53 protein is activated when
Many traits within the human body, resulting in spe- the cell detects damage, such as DNA breakage, and blocks
cific phenotypes, do require many genes operating in the cell cycle at the G1/S checkpoint to allow time for DNA
concert to produce their specific phenotype. This poly- repair. If the damage is extensive the p53 induces abnormal
genic phenomenon can be operating in some cancers. cells to undergo a suicide through apoptosis. The p53 can
Indeed, there are multiple steps involved in the evolution also affect the mechanism of mitosis; abnormalities may
of cancer. It is well known that multiple hits are required, result in mitotic dysjunction (Figure 1-3).
resulting in multiple changes, which occur with time. It MTS-1, also called p16, is another suppressor
has been estimated that 3 to 6 changes may be the mini- involved in the braking components of the cell cycle.
mum requirement to produce a clone of cells with the Other suppressor genes function in the cytoplasm, such
properties to propagate the cancer to a lethal stage. It has as APC, which is involved in colon cancer. APC may affect
been suggested that these hits are cumulative and may the cell adhesion molecule mechanisms by interacting
10 Part I Principles of Urologic Oncology

PDGF, myc, and Bcl-2. However, none of these have been


shown to be involved in cancer as inherited factors. There
are many candidate genes for familial prostate cancer, such
as RNASEL for HPC-1 on 1q25-25; ELAC2 for HPC-2;
and MSR-1 but as yet none have been confirmed to a point
of certainty. Certainly these genes play a major role in uro-
logic cancers in controlling growth and progression, but
what is the inherited gene that sets off prostate cancer?
This will soon be resolved, as many groups are rapidly
mapping in on the target of candidate genes.6

MICROARRAYS AND PROTEOMICS


DNA expressed as RNA can be reversed transcribed to
obtain a c-DNA sample that can be hybridized to the
Figure 1-3 Schematic of how cyclin-dependent kinases specific DNA of the gene that was transcribed to make
(CDKs) are activated or inactivated in the cell cycle. The the original messenger RNA. By placing from 10,000 to
active kinases are bound to variable cyclins and 40,000 small snippets of DNA from identified genes onto
phosphorylate Rb, thus releasing the Rb brake on the cell a chip or a microscope slide, it is possible to hybridize the
cycle.
c-DNA made from the messenger RNA to the specific
genes it represents. The gene targets are located on each
with catenin-like molecules. DPC4 is involved in pancre- small dot placed on the chip. With these high through-
atic cancer and interacts with the cell signaling mecha- put techniques, it is possible to analyze thousands of gene
nisms.1 NF-1 and NF-2 are suppressor genes involved in expression patterns in one sample in a quantitative man-
cell signaling pathways. ner. This is possible by coloring these gene (c-DNA)
Of great interest to the urologist is the WT1 gene, expression products with red for the control cell, which
which is involved in the Wilms’ tumor of the kidney, and can be normal, or by coloring green for the cancer cell (c-
the VHL gene, which is involved in renal cell cancer DNA) and then by combining the red and green messen-
accompanying von Hippel-Lindau syndrome. VHL can ger RNA, the appearance of a red dot would indicate a
either be lost by inheritance or inactivated by methylation gene that was expressed only in the normal cell. Likewise,
of the cytosine residues of the DNA located in the pro- the green dot would represent genes turned on only in
moter region of this gene. The VHL gene appears, at the the cancer cell. The expression of both red and green
moment, to be involved in the regulation of transcription. would form a yellow dot and would indicate expression in
Transcription is the conversion of the information of both cell types. With this type of microarray or with
DNA into RNA through the action of RNA polymerase other forms of differential displays, it has been possible to
II that forms messenger RNA (mRNA). An important implicate a series of genes that are turned off or on that
protein binds to the RNA polymerase II and controls the may be involved in prostate cancer in comparison to nor-
elongation of the mRNA. This transcription and elonga- mal as is shown in Table 1-1.6 Many of these genes have
tion factor is termed elongin or S III. It appears that in been activated or knocked out in transgenic mice, and
normal cells, VHL forms a protein that binds to the elon- their functions have been studied in many cases. For
gin and is involved in the control of transcription elonga- example, NKX3-1 is expressed in normal prostates and is
tion. When the VHL gene is missing or mutated, it loses decreased in prostate cancers. In mice that have lost one
its ability to complex to the elongin and, therefore, allows or more of these NKX3-1 genes abnormal duct develop-
elongin to interact with RNA polymerase II, deregulating ment and hyperplasia occurs. It then goes on to form
the process of mRNA elongation. VHL is the first sup- PIN. Likewise PTEN on 10q23 mutated in about one-
pressor gene that has been identified to control the level third of human prostate cancers and correlates with high
of transcriptional elongation. This raises the question: Gleason grade. PTEN is a phosphatase that inactivates
Why does the elongation result in cancer? It is believed PIP3 that is a signal of several growth factors, including
that this may increase the expression of certain genes IGF-1. PIP3 activates protein kinase AKT, which leads to
involved in growth control, such as myc or fos. inhibition of apoptosis causing increasing cell survival and
In summary, the only two genes so far identified for uro- a tumor. Increasing both PTEN and NKX3-1 increases
logic cancers that can be inherited and increase our inci- the severity of high-grade PIN in animal models.
dence of cancer are VHL and WT1, which cause renal cell The most consistent of these genomic alterations
cancers. In urologic cancers, other suppressors have been associated with cancer may be the GSTP1 gene, which is
implicated, such as Rb, p53, p16, and the oncogenes inactivated by hypermethylation of the promoter region
Chapter 1 The Molecular and Cellular Biology of Urologic Cancers 11

that occurs in over 90% of primary lesions of prostate heterodimers with kinases that are either active or inac-
cancer. The functions of the other genes in Table 1-1 are tive. This in turn regulates their state of phosphorylation
discussed in more detail in a recent review.6 of growth suppressors. A kinase is an enzyme that phos-
Certainly the increased expression of a gene in the phorylates a protein. In the cell cycle, these kinases are
form of messenger RNA does not necessarily reflect the termed cyclin-dependent kinases (CDKs); there are
amount of protein or its posttranslational modification. approximately 7 of these enzymes (CDK2, CDK4, etc.).
In this regard, rapid development of proteomic tech- They are usually at a constant level and inactive as shown
niques that give two-dimensional electrophoretic pat- in Figure 1-3. These CDKs are activated at specific
terns of protein content based on their molecular weight phases of the cell cycle by binding to a second type of
and charge is providing additional means of identifying molecule, called cyclins (termed cyclin A to H). These
proteins that change during various stages of cancer and are termed cyclins because their concentration varies
treatment. These isolated proteins can then be identified through the cycle, and it is these transient molecules that
in sequence using new techniques utilizing mass spec- regulate the cell cycle. Therefore, you can activate cyclin
trometry. Time of flight and fragmentation of proteins in kinases in a controlled manner by turning on the synthe-
mass spectrometers have been extremely useful especially sis and degradation of the cyclins.
when the proteins are first trapped by baiting them to Once the CDKs are activated by binding cyclins, they
specific binding elements on commercially developed appear to regulate the cell cycle by phosphorylating and
probes. Certainly as these techniques become defined turning off the brakes within the nucleus that prevent cell
and standardized they will add a new armamentarium to growth. One of the primary brakes or suppressors in the
the identification of new markers and targets. nucleus is Rb, which, when unphosphorylated, is a check-
point at G1/S and prevents the cycle from proceeding.
When the cyclin kinase is activated, it phosphorylates the
THE CONTROL OF THE CELL CYCLE, CELL
Rb, thus removing the brake and allowing the cell cycle
DEATH, AND TUMOR GROWTH
to initiate DNA synthesis and to continue to complete
In normal tissues, the rate of cell replication and the rate growth to the daughter cell.
of cell death are in a tightly controlled balance, but it is CDKs can also be inactivated by binding to a group of
unknown how this balance is maintained. However, when CDK inhibitors (CDKIs). Examples of this type of
an imbalance occurs, either through an increase in cell inhibitor are p16, p21, and p27. If these inhibitors are
replication or a decrease in cell death, there is an accu- induced, the cell cycle stops, growth is suppressed, and so
mulation of cells that forms the tumor. This balance a checkpoint is formed. How are these inhibitors
involves growth factors, cell signaling, and control of the induced? This occurs in part through the normal func-
cell cycle, as well as apoptosis. DNA damage, aging, and tion of p53, which acts like an inducer and can up-
senescence activate certain signals, which we believe to regulate these inhibitors. The p53 is usually turned on
be “death” genes that cause the cell to commit suicide. when cells are damaged. In this case, the cell wishes to
Signaling of the cell cycle for growth and cell death is one make the decision not to proceed through cell cycle and
of the most active areas of science. to repair itself. In summary, expression of p53 is
First we will review how the cell cycle functions. The increased during cell and/or DNA damage and induces a
cell is usually quiescent in the nongrowing phase, which braking system on the cell cycle to prevent defective cells
is termed G0. Growth factors, steroids, and hormones from being made. If the p53 is damaged, lost, or down
can stimulate the cell to grow and undergo an active regulated, this checkpoint is eliminated. This results in
phase of biochemical events, termed G1 or the gap period damaged DNA proceeding and accumulating through
that occurs before DNA synthesis. After the biochemical each cell cycle and may result in the large amount of
preparation in G1, the cell undergoes DNA synthesis, genetic instability and DNA damage that occur in cancer.
termed the S phase. Following the replication of the We have just discussed most briefly how the cell cycle
complete DNA, there is a second gap called G2, where is regulated, but how does a cell determine to undergo
the cell prepares itself for mitosis. Then the mitosis (M cell death or apoptosis? Damage to the DNA is detected
phase) ensues, in which the mitotic spindle separates the in part by a series of checkpoint including AMT, ATR,
two sets of chromosomes. Then the nucleus reorganizes KU 70, and poly-ADP ribosylation. Broken ends of the
and the cell cycle is completed. Recently, there has been DNA often bind KU 70 or have a special polymer added
a tremendous amount of research delineating the bio- to them that is made from a breakdown product of
chemical controls of the cell cycle. There are specific nicotinamide-adenine dinucleotide (NAD). The poly-
checkpoints at the interface between each of these ADP ribosylation of the ends of damaged DNA appears
phases, in which the cell stops to determine its next deci- to set off a signal that can induce cell death.
sion. These decisions in the cycle are primarily con- Other ways to induce cell death are to remove the cell
trolled by the interaction of regulatory proteins to form from its extracellular matrix (ECM) anchorage or to disturb
12 Part I Principles of Urologic Oncology

the cytoskeleton. This is termed anoikis; unknown sig- teolytic degradation of the nuclear architecture, destroy-
nals from the cell periphery and integrin disruption are ing the lamins around the nuclear periphery and the
being signaled to the nucleus to induce cell death. In internal nuclear matrix components. The cell then disin-
addition, there are large protein molecules like tumor tegrates under protease activity, and phagocytosis of the
necrosis factor (TNF), Fas-ligand and trail ligands that remaining components occurs, destroying the cell. This
act conversely to growth factors and could be termed entire event of cell death has been termed “apoptosis”
death factor. Their action is cell type specific. The TNF and is characterized by these morphologic and series of
binds to two types of cell surface receptors and the Fas- biochemical events.
ligand binds to its receptor. These complexes then As there are brakes or suppressors of growth on the
involve the activation of a series of caspases (2, 3, 6, 7, cell cycle, such as p53 and Rb, there are also brakes to
and 9) and the release of cytochrome c from the mito- stop cell death. One of the leading brakes, for example, is
chondria all in dynamic concert with either proapoptotic Bcl-2. When Bcl-2 is available, it blocks the process of
factor induction (Bid, Bad, Bax) or antiapoptotic factor cell death and therefore is termed a survival factor. How
suppression (Bc1-2, Bcl-xl). is the brake Bcl-2 removed? It can bind to a series of pro-
Sometimes growth factors can induce cell death in teins and form a heterodimer. One of these Bcl-2-binding
certain cell types. For example, tumor growth factor-β molecules is termed Bax, and now a family of these death-
(TGF-β) can activate cell death in some epithelial cells. inducing molecules has been identified. Combining the
Other growth factors appear to induce cell death when Bcl-2 with Bax removes the brake and allows cell death
they are absent; these include EGF and FGF2 and FGF7. to occur.
Of great importance to the urologist is the fact that the
absence of androgen on its receptor can induce cell death
CELL GROWTH FACTORS
in the prostate. Therefore, when the androgen with-
drawal occurs following castration, this absence of andro- There is much direct and indirect signaling that occurs
gens induces rapid cell death in the prostate epithelial between cells and organs. As shown in Figure 1-4, this
cells. This is, of course, the basis for the hormonal treat- signaling can be broken down to various types or cate-
ment of prostate cancer. gories. Growth factors (GF) are of many types, and they
In apoptosis, the fragmentation of the DNA produces bind to specific transmembrane receptors on the cell sur-
a characteristic pattern of small DNA fragments that can face, setting off kinase cascades and structural informa-
be observed on gel electrophoresis to form the multiple tion to induce cell growth or death. If the growth factor
pieces of short DNA of 170 bp surrounding the nucleo- is made and operates on the cell in which it was manu-
somes. These multiples of 170 produces a stepladder factured, it is called an autocrine factor. Usually, the
effect on DNA gel analysis. Once the DNA fragmenta- autocrine factors are secreted from the cell and then
tion occurs, it is irreversible and accompanied by the pro- bind to their specific cell surface receptors. If the growth

Figure 1-4 Examples of the types of cell signaling. (Adapted from Partin AW, Coffey DS: In
Walsh PC, Retik AB, Stamey TA, Vaughan ED Jr (eds): Campbell’s Urology, 7th edition.
Philadelphia, WB Saunders, 1997, with permission.)
Chapter 1 The Molecular and Cellular Biology of Urologic Cancers 13

factor operates within the cell, it is called an intracrine form a homopolymer with their neighbors. One of the
mechanism. If the growth factor diffuses to a neighbor- most prominent of these is E-cadherin, which is a cell
ing cell, it is termed a paracrine stimulation. If the surface CAM and extends through the membrane of the
growth factor is transported through the circulation to cell and organizes cytoskeleton components, such as
distant cells, it is termed an endocrine effect. Other spe- actin. It does that by interacting with an important mol-
cial factors can be transported by the nerves (neurocrine), ecule called catenin, which appears in several forms
or they may come from immune like cells (cytokine). called α and β. In cancers, there is aberration in the
Cells can also signal by direct communication through expression of E-cadherin, whose expression can be regu-
linkages of their structural elements. The ECM makes lated by methylation of the DNA in the promoter region
direct contact with the cell by binding to integrins, which for this gene. The linkage to actin can also be disrupted
are molecules that extend through the cellular membrane by components that can bind to the catenin, such as the
and link to the cytoskeleton within the cell. Cells can also suppressor APC, which has been delineated in colon can-
“hold hands” with their neighbors by direct linkage of cer. The cytoskeleton can also be regulated in its organi-
the cell adhesion molecules (CAMS), which form zation by binding to receptors called integrins that detect
homodimers. ECM components, such as fibronectin. Aberrations in
These direct structural linkages, which transfer infor- this linking system, which involves vinculin, tailin, and α-
mation in a vectral manner, allow the cell to sense its actinin, disrupt the organization of the cytoskeleton
neighbors. This linkage is like a telephone area code and components, such as actin.
is one of the most active areas of research in cell biology. The cytoskeleton is made up of microtubules, actin,
These combined units of structural elements form a tis- and keratins, which give the shape to the cell and a differ-
sue matrix system, as is shown in Figure 1-5. The CAMS ent structure to each cell type. The recognition of the cell

Figure 1-5 The dynamic tissue matrix system is composed of interlocking structural
components that hardwire the cell to the nucleus and DNA. The proteins of the matrix are
tissue specific.
14 Part I Principles of Urologic Oncology

structure, shape, and organization is the basis of histology. to their cell surface receptors, which span the membrane
The cytoskeleton links directly to the nuclear matrix, and activate a series of kinases on the cytoplasmic portion
which organizes the DNA into 50,000 loop domains, of the receptor, they set off a cascade of phosphorylation
termed replicons. These loops of about 60,000 bp of that proceeds to the nucleus. The kinases can either be
DNA are anchored at their base onto the nuclear matrix, tyrosine kinases, which put phosphate groups on tyro-
where DNA synthesis and DNA methylation can occur. sine, or serine kinases, which put phosphate groups on
Steroid hormone receptors bind to this nuclear matrix in the serine molecules of the target substrate. Many of
a tissue-specific manner dictated by the receptors dimer- these kinases are opposed by phosphatases. Figure 1-6
ization and interaction with coactivators on corepressors. demonstrates a few of these cascades of protein phospho-
It is this nuclear matrix protein pattern that makes up the rylations that are directed to the nucleus. Each large K
tissue specificity. The nuclear matrix protein pattern is indicates a kinase that is activated as the receptor signals
altered in cancer. This dynamic tissue matrix system down to the nucleus. A selection of five of the major
is shown in its interactive form in Figure 1-5. kinase cascades involve a series of phosphorylations of
In summary, what a cell touches determines what a cell kinase molecules that activate other kinase molecules to
does, and the disturbance in the tissue matrix system and phosphorylate, finally reaching phosphorylation of
its dynamic components cause the variation in shape that nuclear factors. These four prominent kinase pathways
we term pleomorphism that is a hallmark of cancer. Only are the jak/stat, MAP kinase, and the jnk/erk path-
the pathologist can diagnose cancer, which is done by ways and the PI-3 kinase. These phosphorylation pathways
recognizing aberrations and variations in the nuclear finally terminate in the nucleus to activate a series of
structure and in the tissues and cell structure. Cancer is a transcription factors that induce the expression of spe-
disease of cell structure. cific genes of either growth and death or survival and
senescence. The receptor tyrosine kinase receptor mole-
cule can also link itself to a series of G-protein systems to
CELL SIGNALING
activate these kinases or can act through another impor-
As mentioned, many factors can regulate cell growth and tant mechanism that converts the lipids of the membrane
cell death. They do so by interacting with specific trans- into signaling molecules. For example, phospholipase C
membrane receptors on the cell surface. Their ligands (PLC) can hydrolyze the lipid molecules of the mem-
involve growth factors, cytokines, stress signals, ECM, brane to become signals by making inositol phosphate
and death signals, such as TNF. Once these ligands bind (IP-3) or diacylglycerol (DAG). The IP-3 activates calcium

Figure 1-6 Brief example of 5 types of transmembrane receptor activating the


phosphorylation cascade to activate nuclear function. K, kinase; KK, kinase that
phosphorylates a kinase. See text for discussion.
Chapter 1 The Molecular and Cellular Biology of Urologic Cancers 15

release in the cell that is a signal. The DAG activates pro- is formed by secretion and products made from both the
tein kinase C, which is another major kinase system also stromal and epithelial cells, such as fibronectin, collagen,
signaling to the nucleus. laminin, and proteoglycans. This structural support sys-
One of the most important cell signal pathways in tem organizes the structure of the cell and polarizes it to
prostate cancer is the PI-3 kinase that activates AKT and receive growth factors and signals that come from the
cascades to block the proapoptotic factor Bax and thus stroma, epithelium, and endocrine hormones diffusing
increases cell survival. The phosphorylation of AKT is from the blood vessels.
countered by the phosphatase PTEN that is inactivated This action of growth factors steroids, and the ECM
in many prostate cancers, thus increasing Bax phospho- making the stromal–epithelial organization and cross-
rylation and increasing cell survival. This has been only a talk is shown in Figure 1-7, which is a diagram of the
brief simplified glimpse at some cell signaling pathways prostate, where the epithelium is composed of neuroen-
that are involved in cancer. docrine, secretory epithelial, and basal cells. It is believed
that the basal cells are the stem cells that differentiate to
form both secretory and neuroendocrine cells. The
STROMAL EPITHELIAL INTERACTIONS
stroma is made up of smooth muscle, fibroblast, and
All of the aforementioned signaling mechanisms and nerve cells. Threading their way through the stroma are
many more come together and are synchronized in the the capillaries, lined by endothelial cells, and the immune
cell organization forming the tissue that involves the cells, which can move in and out of the prostate. The
interaction of many stromal and epithelial signals. The capillaries bring the steroids, androgens, estrogens, and
stroma talks to the epithelium and the epithelial cells talk nutrients to the prostate. Testosterone is converted to the
to the stroma, both types are nurtured by blood vessels more active dihydrotestosterone (DHT) by 5-α-reductase
and nerves. in the stroma. In the stroma, the DHT stimulates fibrob-
The interface between the stromal and epithelial cells last growth factor-7 (KGF), which then diffuses up and
is conducted through the formation of the ECM, which activates the receptors on the epithelial cells in a

Figure 1-7 Stromal-epithelial interactions in the prostate mediated by DHT regulated growth
factors; +, stimulation; −, inhibition; NO, nitric oxide. (Adapted from Partin AW, Coffey DS: In
Walsh PC, Retik AB, Stamey TA, Vaughan ED Jr (eds): Campbell’s Urology, 7th edition.
Philadelphia, WB Saunders, 1997, with permission.)
16 Part I Principles of Urologic Oncology

paracrine manner. DHT also stimulates fibroblast units that allow the cell to replace the telomeres that are
growth factor 2 (BFGF), which both feeds back in an lost when the cell divides. In any cell in culture, telom-
autocrine effect on the stroma and has a paracrine effect erase has to be activated or the cells would not be immor-
on the epithelial cells. A similar stimulation is induced by tal. This is also the case only in the stem cells and the
DHT on the production of insulin-like growth factor II germ cells. The other cells do not have telomerase activ-
(IGF-II), which also has an autocrine and paracrine ity and are subject to cell death as the mitotic clock ticks
effect. Insulin-like growth factors are bound to a family down counting each cell cycle. In cancer cells, telomerase
of insulin-like growth factor binding proteins (IGFBP), is activated and the cells have become immortal.
which are also made by the stroma. DHT can diffuse We have reported that telomerase is one of the best
from the smooth muscle into the epithelial cells, where it markers so far in denoting prostate cancer cells from nor-
induces the synthesis of epidermal growth factor and mal and benign prostatic hypertrophy (BPH) cells. This
TGF-α. In the epithelial cells, the androgen also induces will be a new diagnostic marker when applied in an
production of IGFBP, which complexes the insulin-like appropriate manner. Telomerase is one of the most excit-
growth factors and keeps it inactive. One of the main ing frontiers in understanding senescence, immortality,
secretory proteins made by the prostate is PSA, which and how the cancer cell has broken through this aging
hydrolyzes the IGFBP to release active IGF-I and IGF- barrier to become immortal.6 Telomere shortening is one
II, which then can stimulate the growth of the epithelial of the earliest molecular lesions in cancer and can initiate
cells. PSA is a major secretory protein in the ejaculate. genetic instability by altering chromosome structure.
This diagram shows some of the cross-talk between
the stroma and epithelial cells via the testosterone and
OVERCOMING THE TUMOR CELL
DHT induction of growth factors that can function in an
HETEROGENEITY: APTAMERS AND IN VITRO
autocrine and paracrine way to these cell components. It
EVOLUTION
is also important to note that many neurotransmitters are
made in the prostate, such as NO. NO is produced by the One of the major obstacles in the treatment of urologic
endothelial, immune, and nerve cells and can have a cancers is their tremendous heterogeneity. Although
strong stimulatory effect on stromal and epithelial com- clinically appearing as a homogeneous tumor, it actually
ponents how this entire system is organized in the consists of a heterogeneous pool of cancer cell clones.
embryo, grows to adult size and then becomes dysregu- When we apply any treatment, such as chemotherapy or
lated to produce tumors with aging is the basis of much radiotherapy, we select for subclones in the tumor that
research. are resistant to our treatment. This is not related to the
response of the tumor cells to our treatment but to their
ability to use evolutionary techniques to escape any given
AGING AND TELOMERASE
therapy. There are now new technologies that will allow
Although aging is involved in cancer, we know very little us to turn therapeutic evolution on the evolution of the
about what really brings about this irreversible and dete- tumor and thus beat the cancer at its own game.
riorating effect. Certainly, accumulated damage from Aptamers can be small peptide, DNA, or RNA mole-
free radicals from reactive oxygen, as well as cross-link- cules that will bind in an antibody like fashion to any
ing and stiffening of collagen, are all key components in given target. Large pools of randomized molecules can
how we age. Importantly, there is also a biologic clock be easily made and will provide the molecular diversity to
that counts each cell division; this brings about senes- let the tumor cells select the best binding molecules to all
cence. At the end of each chromosome are repetitive the different tumor cell clones. For example, by random-
pieces of DNA called telomeres. When the cell divides izing the four bases of a 15-nucleotide RNA sequence it
each time, it loses a small amount of these telomeres, is possible to create pools with more than 415 to 109 dif-
which is caused by the inability of the DNA synthesis ferent molecules. A selection screen is set up in a way
mechanism to fully replicate the last little bit of terminal allowing us to select the best RNA molecules out of the
DNA. The loss of these repetitive pieces of DNA is random pool, that will bind with a high affinity and speci-
therefore accumulative and acts as a mitotic clock, count- ficity to our tumor cells. Bound RNA species are then
ing the cell cycles. After approximately 50 doublings, the recovered and amplified. This enriched RNA fraction is
telomeres of the cell have been reduced to a critical then subjected to a new round of selection. After 10 to 20
length, resulting in the cell’s senescence and death. Every rounds of selection, recovery, and amplification, the pool
cell is limited by this mitotic clock except cells that have will contain RNA molecules with high binding speci-
learned how to become immortal. The immortal cells ficity. Figure 1-8 gives an overview of a typical selection
stabilize their telomeres by activating the enzyme called and amplification cycle that we have involved in in vitro
telomerase. Telomerase is an enzyme that carries its own evolution to prostate cancer cells. Those aptamers can
small template made of RNA that is copied into telomere then be analyzed and produced in large quantities. They
Chapter 1 The Molecular and Cellular Biology of Urologic Cancers 17

REFERENCES

1. Levins M, Tjian R: Transcription regulation and animal


diversity. Nature 2003; 424:147.
2. Bray D: Molecular networks: the top-down view. Science
2003; 301:1864.
3. Stuart GR, Holcroft J, De Boer JG, Glickman BW:
Prostate mutations in rats induced by the suspected human
carcinogen 2-amino-1-methyl-6-phenylimidazol [4,5-6]
pyridine. Cancer; 60:266.
4. Nelson WG, De Marzo AM, Isaacs WB: Mechanisms of
disease prostate cancer. N Engl J Med 2003; 349:366.
5. Coffey DS: Similarities of prostate and breast cancer;
evolution, diet, and estrogens. Urology 2001; 57(Suppl
4A):31.
Figure 1-8 Selection cycle for enriching the binding of a 6. De Marzo AM, Nelson WG, Isaacs WB, Epstein JI:
random (109) RNA (aptamers) pool to tumor cells. PCR, Pathological and molecular aspects of prostate cancer. The
polymerase chain reaction for amplifying DNA; enriched RNA. Lancet 2003; 361:955.
The final selected aptamer binds specifically to the tumor 7. Easton DF, Schaid DJ, Whittemore AS, Isaacs WB: Where
cells.8 are the prostate cancer Genes?—A summary of eight
genome wide searches. The Prostate 2003; 57:261.
8. Lupold SE, Hicks BJ, Lin Y, Coffey DS: Identification and
can be used as highly specific cancer probes, to improve characterization of nuclease stabilized RNA molecules that
diagnosis or, when linked to a cytotoxic “warhead,” as a bind human prostate cancer cells via the prostate-specific
new therapeutic approach to treat cancer.8 membrane antigen. Cancer Res 2002; 62:4029.

ACKNOWLEDGMENTS
We wish to acknowledge the outstanding effort of Vivian
Bailey in preparing this chapter and Don Vindivich in
constructing the figures.
C H A P T E R

2 Gene Therapy for Urologic Cancer:


Basic Principles, Prospects, and
Limitations
Martin G. Sanda, MD, and Ronald Rodriguez, MD, PhD

Over 50 years of scientific research following the discovery tions. Gene transfer vectors can be generally classified
of DNA1 have led to recent insights that have set the stage as being of either viral or nonviral origin. Vector design
for preclinical development of gene therapy strategies and is guided by their desired functions: efficiency of gene
their assessment in clinical trials. The enormous volume of transfer, stability of gene expression, and safety of
scientific research that comprises the foundation for ther- clinical use.
apeutic use of molecular biology reflects the complexity of Gene transfer efficiency indicates how well a vector
linking the most fundamental unit of life, the human delivers a recombinant gene to a target cell and how
genome, to direct clinical intervention. These profound effectively the protein encoded by that gene is subse-
complexities were manifest as several unforeseen adverse quently expressed. Highly efficient gene transfer is
events in clinical gene therapy clinical trials observed con- desirable in almost all clinical strategies using gene
currently with observed effective treatment by gene ther- transfer. Two critical components determine gene
apy.2,3 Consequently, the initial exuberant enthusiasm for transfer efficiency: gene delivery by the vector and
clinical gene therapy has been tempered by an emerging activity of a vector’s expression cassette. The first deter-
respect for how the profound complexity of in vivo gene minant of efficiency, gene delivery, refers to a vector’s
transfer faces obstacles to therapeutic efficacy and carries capacity for cellular attachment, entry, and delivery of
the potential for significant clinical toxicity. the therapeutic gene (within an expression cassette) to a
Preclinical studies have identified several therapeutic site such as the target cell nucleus at which gene expres-
strategies based on gene replacement or gene transfer that, sion can occur. Delivery mechanisms of each vector sys-
in some cases, have moved to evaluation in clinical trials. tem are distinct.4–6 Cell surface density of specific
Gene therapy strategies under development include cor- receptors required for vector attachment, as well as sta-
recting tumor-specific genetic abnormalities by either bility of the vector itself in the micro-environment sur-
inhibiting the function of oncogenes (abnormal tumor rounding the target cell, affects a vector’s capacity for
genes that promote tumor cell longevity or proliferation) therapeutic gene delivery.7–9 The second principal
or restoring functional tumor suppressor genes (such as determinant of vector efficiency is the vector expression
genes that can regulate DNA repair), which are commonly cassette, which contains, in addition to the therapeutic
functionally mutated or absent in cancer. Alternatively, the gene, a promoter sequence controlling therapeutic gene
immune response of patients with cancers harboring any transcription. Some vectors (such as poxviruses) encode
of these abnormalities can be stimulated by gene therapies their own machinery for gene transcription,6 while oth-
based on use of recombinant tumor vaccines. Pivotal to all ers rely entirely on preexisting polymerases in the target
avenues of gene therapy is the gene transfer vector. cell10; however, all vectors carry a promoter region
flanking the therapeutic gene. The promoter pro-
foundly affects therapeutic gene expression and vector
GENE TRANSFER VECTORS
efficiency.11
As the vehicles for therapeutic gene delivery, gene The stability of gene expression by a specific vector
transfer vectors delegate clinical prospects and limita- system depends on the intracellular localization of the

18
Chapter 2 Gene Therapy for Urologic Cancer: Basic Principles, Prospects, and Limitations 19

therapeutic gene by the vector. Typically, episomal local- Vector Production


ization (when the therapeutic gene is not integrated in
Production of replication-deficient retroviral vectors is
the target cell chromosome) leads to transient expression
accomplished with vector packaging cell lines (Figure 2-1).
of the gene because most mammalian cells have efficient
Vector particles secreted into packaging cell supernatant
mechanisms for extruding episomal foreign DNA. In
are purified and concentrated in preparation for use for
contrast, vectors that allow integration of the transferred
gene transfer.
gene into the host cell chromosomal DNA, such as retro-
viral vectors, provide longer duration of stable expres-
sion. Although stable, durable expression may be Gene Delivery
desirable in treating hereditary disorders and chronic dis-
orders, it should be noted that transient gene expression Cells targeted for gene transfer using the purified retro-
might be sufficient for the purposes of using gene trans- viral vector incorporate the vector by endocytosis via a
fer for cancer treatment.10 specific receptor (Figure 2-2). Reverse transcriptase then
converts vector RNA into DNA, which is then integrated
into the target cell chromosomal DNA during target cell
RETROVIRUS VECTORS
proliferation. This requirement for target cell prolifera-
Retroviral vectors were used in the first clinical trials of tion as a prerequisite to transferred gene expression is
gene therapy.12,13 Although retroviral vectors provide dis- unique to retroviral vectors; retroviral vectors thus do
tinctly stable long-term expression of therapeutic genes, not readily transfer genes to quiescent cells.8,9
use of these vectors is limited by complexity of retroviral
genetic engineering and vector purification as well as by
Gene Expression
safety obstacles.3,8,9 These vectors are currently being
used predominantly in ex vivo gene transfer protocols, After integration of the retroviral vector expression cas-
although in vivo gene transfer applications are emerging. sette (the therapeutic gene flanked by sequences that

Virus envelope
Vector genome with therapeutic or protein coat
gene expression casette
Viral genome containing
therapeutic gene
expression cassette

Factors for initiation of


expression in targety cell
Transfection Assembled
recombinant
viral vector
Viral vector particles
packaging
cell line

Transfection

Vector complementary
viral gene(s)

Figure 2-1 Production of recombinant viral vectors for gene therapy. A schematic
representation of retroviral vector production is shown; analogous systems are in use for the
production of adenoviral and poxvirus vectors. For retroviral vector production, packaging
cell lines in which the therapeutic gene has replaced retroviral gag, pol, and env genes
(these genes are normally required for retrovirus particle production and packaging by
infected cells). The packaging cell line has been co-transfected with these viral genes in
trans to complement the replication defective vector genome, allowing the packaging cell
line to produce and package replication-deficient viral vector particles. (Based on Danos O,
Mulligan RC: Proc Natl Acad Sci USA 1988; 85:6460; Ghosh-Choudhury G, Haj-Ahmad Y,
Brinkley P, et al: Gene 1986; 50:161; Mulligan RC: Science 1993; 260:926.)
20 Part I Principles of Urologic Oncology

Figure 2-2 Gene transfer by retrovirus vectors. Retroviral gene transfer is mediated by
integration of the therapeutic gene into the target cell chromosomal DNA, and therefore
requires target cell DNA replication. (Based on Danos O, Mulligan RC: Proc Natl Acad Sci
USA 1988; 85:6460; Mulligan RC: Science 1993; 260:926; Crystal RG: Science 1995;
270:404; Miller N, Vile R: FASEB J 1995; 9:190.)

Attributes and Applications


promote gene expression) into the chromosome of the
target cell, the therapeutic gene is expressed by the tar- Due to limitations in the functional concentration (or
get cell’s own polymerases and other mediators of gene titer) of retroviral preparations, and rapid inactivation of
expression. Because the therapeutic gene is integrated unbound retrovirus in vivo, direct in vivo gene transfer
into the target cell genome, it is stably expressed by the using retroviral vectors has generally been inefficient,
cell long-term and is passed onto progeny should the with only a small fraction of target cells expressing the
cell continue to proliferate. Further contributing to transferred gene. Retrovirus vectors can, however, medi-
long-term stability of retroviral transferred gene ate highly efficient gene transfer ex vivo (Table 2-1).4
expression is the immunologically inert phenotype of Cancer therapy applications of retroviral vectors have
these vector constructs: immune responses targeting the therefore predominantly involved ex vivo gene transfer,
vector itself have not been an obstacle to retroviral vec- for example, to augment the immunogenicity of patient-
tor use. Although stable genomic integration, as can be derived tumor cells (by introducing into such tumor cells
achieved with retroviral vectors, may be desirable for an immunostimulatory gene) prior to use of such cells in
some therapeutic strategies (such as tumor suppressor vivo as a gene-modified tumor cell vaccine or for marker
gene replacement), stable and permanent alteration of studies.12,14,15 Because retroviral gene transfer itself nei-
the target cell genome in vivo also poses potential safety ther damages the host cell nor induces undesirable vec-
pitfalls such as potentially irreversible untoward genetic tor-specific immunity, these vectors are ideally suited for
effects in vivo. Reflecting the profound in vivo effects of gene-modified cancer cell vaccine therapies seeking to
retrovirus vector integration into chromosomal integra- elicit tumor-specific immunity. Despite early encourag-
tion, such vectors proved effective in treating severe ing results of clinical trials that used retroviral vectors to
combined immunodeficiency disease in children; how- create gene-modified tumor vaccine studies for renal
ever, cancers developed in some of these children that cancer and prostate cancer, the recent observation of vec-
are believed to have been consequent to integration of tor-induced leukemias in children undergoing retroviral
the retroviral vector.3 gene replacement therapy3 will likely reduce enthusiasm
Chapter 2 Gene Therapy for Urologic Cancer: Basic Principles, Prospects, and Limitations 21

Table 2-1 Attributes of Vectors for Therapeutic Gene Delivery


Duration of Therapeutic Efficiency of Gene
Vector Gene Expression Transfer Other

Retrovirus Stable long-term Variable Labile in vivo

Adenovirus Transient Highly efficient Immunogenic

Poxvirus Transient Highly efficient Immunogenic

Nonviral plasmid or naked Transient Inefficient Fewer safety concerns


DNA

for clinical applications to only those wherein stable Binding occurs through the interaction of the knob of the
chromosomal integration is required for the desired ther- protruding viral capsid fiber protein to its cellular recep-
apeutic effect. tor, referred to as Coxsackie and adenovirus receptor
(CAR). Internalization occurs as a result of the interac-
tion of the RGD motif of the viral penton base capsid
ADENOVIRUS VECTORS
protein with the cellular integrins (αVβ5 or αVβ3,
Interest in adenoviral vectors was prompted by their Figure 2-3).9,17,18 After internalization, the vector is
capacity for highly efficient gene transfer in vivo. The transported from the endosome to the cytoplasm, where
propensity of adenovirus vectors to induce nonspecific the adenoviral protein coat is lost as the adenoviral DNA
inflammation and a vector-specific immune response, migrates to the nucleus. In the target cell nucleus, the
however, has limited efficacy of this vector system in clin- vector remains epichromosomal and is not integrated
ical trials evaluating adenovirus vectors for gene replace- into the target cell chromosome. Replication of the tar-
ment therapy for noncancerous diseases.9,16 Moreover, an get cell is not required for therapeutic gene delivery.
inflammatory response against the adenovirus vector has
been suspected as possibly contributing to the first inci-
Gene Expression
dence of a fatal complication of gene therapy.2 Whether
the efficacy of antitumor therapies using this vector system Nuclear localization of the adenoviral vector allows the
will be attenuated or augmented by inherent immuno- target cell’s own polymerases and other mediators of
genicity of adenoviral vectors remains to be elucidated. gene expression to participate in expression of the thera-
peutic gene. However, because they remain epichromo-
somal, expression of adenoviral vector genes is transient,
Vector Production
lasting only weeks.9
Recombinant adenoviral vectors are rendered infectious
but replication defective (while retaining their capacity
Attributes and Applications
to infect cells) by deletions in an early region DNA (E1)
required for viral replication.5 The deleted E1 region Adenovirus vectors are characterized by transient
and other regions of the adenovirus genome serve as duration of gene expression in target cells, significant
sites for therapeutic genes by using shuttle plasmids and induction (by the vector) of inflammation and immunity,
homologous recombination with complementary dele- and capacity for highly efficient gene transfer in vivo
tion mutants (see Figure 2-1). A packaging cell line (293 (see Table 2-1). The transient gene expression associated
cells, a transformed human embryonic kidney cell line with adenoviral vectors is less likely to constrain the util-
containing the adenovirus E1 DNA) transfected with ity of these vectors for cancer therapy than for other
the E1 deleted adenoviral vector containing a gene of applications: gene-targeted immunotherapy, as well as
interest is used to produce the replication-deficient ade- apoptosis-inducing therapies, does not require perma-
noviral vectors.5 High concentrations of adenoviral vec- nent expression of therapeutic genes, and adenovirus
tor (titer >1011 pfu/ml) can be readily purified. vectors have been effectively applied for such therapeu-
tic strategies in preclinical models.19,20 Indeed, the tran-
sient nature of adenovirus-mediated gene transfer
Gene Delivery
circumvents the safety issue of irreversible undesirable
Adenoviruses are taken up by the target cells via a two- genetic effects such as could be encountered with retro-
step process, involving binding and internalization. viral gene transfer.
22 Part I Principles of Urologic Oncology

Figure 2-3 Gene transfer by adenovirus vectors. Adenoviral vectors do not require target
cell DNA replication for efficient gene transfer. However, nuclear localization of the vector
DNA is required since target cell nuclear factors are used for gene expression. Because
these vectors are epichromosomal, therapeutic gene expression is typically less durable
than with retroviral vectors. (Based on Ghosh-Choudhury G, Haj-Ahmad Y, Brinkley P, et al:
Gene 1986; 50:161; Crystal RG: Science 1995; 270:404; Miller N, Vile R: FASEB J 1995;
9:190.)

Adenovirus vectors are potent immunogens.16 utilizing adenoviral vectors for prostate cancer have
Although potentially desirable for gene-targeted resulted in encouraging biologic activity and is an area of
immunotherapies, the inherent immunogenicity of aden- active translational research.23–25
oviruses may also limit repeated administration of these
vectors due to sensitization-induced inflammatory toxic-
POXVIRUS VECTORS
ity.9 Moreover, adenovirus-specific immunity may con-
strain in vivo gene transfer with adenoviral vectors by Vaccinia and other poxvirus vectors are derived from
inducing humoral and cellular responses capable of elim- one of the greatest triumphs of post-classical medicine:
inating the vector and further reducing duration of target the smallpox vaccine.6 Jenner’s discovery in 1798 that a
gene expression.16 Despite these limitations, the recep- bovine poxvirus was an effective human vaccine against
tivity of many human cells to adenoviral transfection, smallpox eventually led to implementation of a con-
coupled with the relative stability and ease of production certed worldwide vaccination program by WHO,
of adenoviral vectors, led to significant tumor reduction which was implemented in 1967, and Jenner’s predic-
in several preclinical models of direct in vivo gene trans- tion of “the annihilation of the smallpox” was finally
fer and broad clinical applications.19–22 The immuno- realized in 1995, nearly 200 years after the introduction
genicity of adenoviral vectors was found to carry of poxvirus vaccines for human use. Current use of
potentially dire consequences; however, a systemic poxvirus vectors for experimental cancer therapy relate
inflammatory response, possibly manifested by unre- to the ability of these vectors to induce potent immune
stricted complement activation, was implicated in the responses in vivo.
widely publicized death of Jesse Gelsinger, a patient who
received a dose of 38 trillion adenovirus particles on a
Vector Production
phase I trial of adenoviral gene therapy for an inherited
hepatic enzyme deficiency.2 Nevertheless, early phase Because recombinant poxviruses are used as live, replica-
clinical studies using adenoviral vectors continue, and tion-competent viruses (albeit in attenuated or otherwise
Chapter 2 Gene Therapy for Urologic Cancer: Basic Principles, Prospects, and Limitations 23

nonpathogenic forms when administered in vivo), pro- cytoplasm and intracytoplasmic release of the virion
duction can be achieved by simply infecting specific host complex core (Figure 2-4).6 The virion complex core
cells that allow productive infection.6 Genetically engi- contains the vector genome, as well as RNA polymerases
neered packaging cell lines, such as those used for retro- and other enzymes required for expression of the vector
virus or adenovirus vector production, are not required. genes; the vector remains in the cytoplasm, where gene
Immunostimulatory cytokine, accessory molecule, or expression controlled by elements contained in the virion
tumor antigen genes can be inserted into vaccinia or complex core occurs.
other poxvirus vectors by flanking these genes with
poxvirus sequences in a “shuttle” plasmid and then intro-
Gene Expression
ducing this plasmid into a cell that has been infected with
whole vaccinia virus. Homologous recombination (as Poxvirus vectors are unique in their ability to express
with adenoviral vector systems) in the vaccinia and shut- therapeutic genes without requiring transport of the vec-
tle plasmid co-transfected cells then leads to insertion of tor to the target cell nucleus. In contrast to other vector
the gene of interest in a small proportion of the viral systems, which require host cell nuclear factors and
progeny (see Figure 2-1). Linking the therapeutic gene enzymes for gene expression, poxviruses carry the appara-
with an adjacent selectable marker gene allows subse- tus for synthesis of translatable RNA (including virus-
quent purification and production of exclusively recom- encoded RNA polymerases, transcription factors, capping
binant poxvirus containing the gene of interest.6 Up to enzymes, and poly(A)polymerases) either prepackaged
25,000 base pairs of foreign DNA can be accommodated within the complex virus core or encoded within the viral
by vaccinia vectors, representing the greatest size capac- genome itself.4 Expression cassettes in poxvirus vectors
ity of currently available recombinant viral vectors for thus require unique poxvirus-specific promoter regions
transferred genes.6 that can be recognized by viral transcription factors.

Gene Delivery Attributes and Applications


Infectious poxvirus virions enter the target cell via fusion Poxvirus transfection is transient and eventually toxic to
of the virion lipoprotein envelope with the target cell the target cell (see Table 2-1). The successful history of

Figure 2-4 Gene transfer by poxvirus vectors. Poxvirus vector particles contain viral RNA
polymerases, obviating any need for nuclear localization or chromosomal integration.
Therapeutic gene expression occurs entirely in the target cell cytoplasm. (Based on Moss B:
Science 1991; 2252:1662.)
24 Part I Principles of Urologic Oncology

vaccinia use for smallpox eradication and the relative ease somes containing plasmid DNA within a lipid envelope
of cloning genes into vaccinia vectors have uniquely but are rather particulate complexes in which plasmid
poised vaccinia and other poxvirus vectors for use as DNA is dispersed among the bound lipids.7,9,27
recombinant tumor vaccines. As with adenovirus vectors,
use of poxviruses for immunogene therapy is tempered
Gene Delivery
by potentially competitive, antivector, immune response
induction.26 These complexes promote cellular gene delivery by
hydrophobic interaction and fusion of the lipid–DNA
complex with the target cell membrane. Unlike viral vec-
NONVIRAL VECTORS: LIPOSOMAL
tors, however, no signal exists to facilitate transport of
GENE TRANSFER
the plasmid DNA containing the therapeutic gene to the
Nonviral approaches to gene therapy avoid disadvantages nucleus (Figure 2-5).7 Transfection efficiency is therefore
of viral vectors such as safety issues related to potential typically relatively inefficient (see Table 2-1). DNA com-
replication-competent virus formation and limited target plexes with copolymers, in contrast, offer the added
cell diversity related to receptor requirements for viral advantage of incorporation of targeting ligands, such as
envelope adsorption.7,9,10,17,18 The most extensively folate or transferrin. Early work in this regard has
developed nonviral gene delivery systems are liposome- demonstrated enhanced gene transfer with systemic
mediated gene transfer and high-velocity particle-mediated delivery of these targeted complexes.28,29
gene transfer, also known as the “gene gun.” In contrast, high velocity particle-mediated gene
transfer, or gene gun, technology allows the delivery of
thousands of copies of DNA into targeted cells. This is
Vector Production
achieved by coating 1–3 μm gold particles with plasmid
Plasmid–liposome complexes for gene delivery are DNA or mammalian chromosomal genomic DNA up to
comprised of DNA formulated with cationic lipids. 44 kb in size; a gene gun is then used to deliver the par-
Most of the cationic lipid–DNA complexes commonly ticles in vivo by generating a high-pressure gas burst that
used for gene delivery in clinical trials are not true lipo- accelerates the particles to a velocity sufficiently high for

Figure 2-5 Nonviral gene transfer by liposomal vectors. Liposomal vectors in current use
are complexes of cationic lipids and plasmid DNA. Although relatively safe and
immunologically inert, liposomal vectors require nuclear localization for access to target cell
transcription factors. (Based on Ledley FD: Hum Gene Ther 1995; 6:1129; Crystal RG:
Science 1995; 270:404.)
Chapter 2 Gene Therapy for Urologic Cancer: Basic Principles, Prospects, and Limitations 25

EMERGING VECTORS AND OTHER GENE


DELIVERY SYSTEMS
The preceding discussion has focused on gene transfer
vectors currently being used in clinical trials as investiga-
tive agents for urologic cancers. A variety of other novel
viral, as well as nonviral, vectors are currently under
development. Among emerging viral vectors are gene
delivery constructs derived from herpesvirus, parvovirus,
and adeno-associated viruses.32–34 Nonviral vectors
under development include eukaryote-derived vectors
Figure 2-6 Nonviral gene transfer via particle bombardment such as Listeria monocytogenes and recombinant BCG, syn-
(gene gun). In the helium pulse gene gun, motive force is thetic constructs such as dendrimers, and others.35 The
generated by release of a high-pressure burst of helium gas
ideal vector for most gene therapy applications will likely
from a reservoir (A) at a preset pressure (150-700 psi). A
release valve (B) discharges helium through a cartridge
evolve as a hybrid vector merging the desirable proper-
(C) containing DNA-coated gold particles. After being ties of viral vectors with advantageous attributes of non-
dispersed by an exit nozzle (D), the DNA-coated gold viral delivery systems.11
particles (E) penetrate target cells or tissue with sufficient
force to penetrate multiple cell layers and deliver plasmid
MOLECULAR TARGETS OF GENE THERAPY
DNA intracellularly. (Reprinted from Yang NS, Sun WH: Nat
Med 1995; 1:481, with permission.) Gene therapy for urologic malignancy can be catego-
rized into four distinct strategies based on the molecu-
penetration of multiple cell layers (Figure 2-6).8 As with lar target of gene transfer: immunogene therapy, direct
lipid–DNA complex gene transfer, translocation of tumor cell death induction, antioncogene therapy, and
plasmid DNA to the nucleus after gene gun delivery is tumor suppressor gene restoration (Table 2-2).
not a specifically targeted event. Immunogene therapy affects tumor growth indirectly
by inducing a tumor-specific immune response either
via immunostimulatory gene transfer ex vivo (followed
Gene Expression
by in vivo administration of genetically altered cells to
Plasmid DNA that does manage to translocate to the induce a tumor-specific immune response) or via direct
nucleus usually is not integrated into the target cell genome in vivo transfer of immunostimulatory or tumor anti-
and remains epichromosomal (see Figure 2-5).5 Similar to gen genes. Direct tumor cell death induction relies on
adenoviral vectors, expression relies on target cell tran- delivery of genes encoding cellular toxins or apoptosis-
scription factors and is typically relatively transient. inducing proteins. Antioncogene therapy specifically
inhibits or eliminates oncogene activity. Tumor sup-
pressor gene restoration therapy inhibits tumor growth
Attributes and Applications
by restoring genes that prevent transformation of the
The principal advantage of nonviral gene delivery sys- normal cell but have been functionally disabled during
tems, including cationic lipid–DNA complexes, the gene carcinogenesis. The preclinical rationale for these
gun, and other nonviral delivery systems, is that these gene therapy strategies, and consequent gene therapy
systems circumvent three potentially problematic charac- clinical trials treating urologic cancers are discussed
teristics of viral vectors: immune reactivity, reliance on (Table 2-3).
viral receptor expression by target cells, and safety issues
related to potential pathogenic recombinant contami-
Immunogene Therapy Via Ex Vivo Gene Transfer
nants in viral preparations. Enthusiasm and applicability
of nonviral vectors are tempered by relatively inefficient Gene therapy via transfer of immunostimulatory genes to
gene delivery and transient therapeutic gene expression induce a tumor-specific immune response is perhaps the
(see Table 2-1). As transient expression systems, nonviral most extensively evaluated strategy of gene therapy to
and gene gun delivery systems have been useful for date. This is partly because early gene transfer systems
induction of antitumor immunity. Induction of cytokine limited gene therapy to strategies using ex vivo (rather
secretion using the gene gun has been associated with than in vivo) gene transfer, and this approach is widely
reduction of renal cell carcinoma progression in a mouse applicable as immunogene therapy using, for example,
model.30 Clinical studies using lipid–DNA complexes patient-derived cultured tumor cells for a gene-modified
have shown induction of antitumor immune mediators in tumor cell vaccine.15,36–43 Generally, clinical applications
melanoma patients, and trials using these vectors for of these studies used retroviral vectors as the vehicles for
renal cell cancer are underway.27,31 gene transfer.15,44 Strategies of immunogene therapy
26 Part I Principles of Urologic Oncology

Table 2-2 Characteristics of Gene Therapy Strategies in Current Clinical Trials


Therapeutic Gene Extent of Potential Efficacy In Vivo* Relative Obstacles

Immunogene: ex vivo transfer Systemic Tissue procurement and cell culture


required

Immunogene: in vivo transfer Systemic Vector-specific immunity may


interfere with induction of
tumor-specific immunity

Cyto-toxicity/apoptosis Local-regional Requirement for highly efficient


gene delivery in vivo; possibility of
cytotoxic injury to normal cells

Antioncogene/antisense Local-regional Requirement for highly efficient


gene delivery in vivo and durable
expression of therapeutic gene

Tumor suppressor Local-regional Requirement for highly efficient gene


delivery in vivo and durable expression
of therapeutic gene
*Based on current vector limitations.

Table 2-3 NIH-Approved Clinical Trials of Gene Therapy for Urologic Cancer
Principal Vector: Therapeutic
Strategy Investigator Gene Cancer Histology Status

Immunotherapy: gene Gansbacher Retrovirus: IL-2 Renal cell Open


transfer ex vivo Simons Retrovirus: GM-CSF Renal cell Completed
Simons Retrovirus: GM-CSF Prostate Open
Paulson Liposome: IL-2 Prostate Pending

Immunotherapy: gene Vogelzang Liposome: class I Renal cell Completed


transfer in vivo MHC
Figlin Liposome: class I Renal cell Open
MHC
Lattime Poxvirus-vaccinia: Transitional cell Open
GM-CSF
Chen Poxvirus-vaccinia: Prostate Open
PSA

Cytotoxic Scardino Adenovirus: HSV-tk Prostate Open

Anti-oncogene Steiner Retrovirus: myc Prostate Pending


antisense

Tumor suppressor Small Adenovirus: Rb gene Transitional cell Pending


gene restoration

have also been formulated that rely on gene transfer in apy for urologic malignancies in preclinical studies and
vivo.45–49 It is therefore useful to evaluate immunogene clinical trials as well. For these therapies, tumor cells iso-
therapy strategies in the context of whether the particu- lated from fresh surgical specimens are genetically trans-
lar strategy requires ex vivo gene transfer or in vivo gene duced during tissue culture with an immunostimulatory
transfer (see Table 2-2). gene. The resected genitourinary cancer cells serve prin-
Therapies using genetically modified, patient-derived cipally as vehicles for autologous tumor antigens and are
cells for a genetically engineered tumor vaccine have transduced for immunostimulatory gene expression. The
comprised the principal use of ex vivo immunogene ther- gene-modified tumor vaccine is typically irradiated ex
Chapter 2 Gene Therapy for Urologic Cancer: Basic Principles, Prospects, and Limitations 27

vivo prior to being reinjected into the patient as a genet- cination using tumor cells derived from the same tumor
ically engineered tumor cell vaccine (Figure 2-7). but transduced to secrete IL-2; such vaccination pro-
Initial preclinical studies evaluating this strategy of tected animals from subsequent tumor challenge.36,37
gene therapy showed that a tumor-specific, T-cell Gamma-interferon gene transfer was shown to promote
mediated immune response could be augmented by vac- a similar protective effect.38 Subsequently, transfer of

100 Treatment
GROUP A Hanks BSS (control)
GROUP B XRT-MLL
80 GROUP C XRT-MLL + SOLUBLE huGM-CSF 8500 ng
Percentage cancer free

GROUP D XRT-MLL-MFG-huGM-CSF 140 ng/10/24


6
CELL DOSE 5 X 10
60

40
(Wilcoxon P=0.001)

20

0
0 15 30 45 60 75 90 105 120 135 150
Vaccine Rx
(Day 3,13,23)
Days after prostate cancer implantation
A
Primary
culture

Immunostimulatory gene
transfer

Irradiation of
human gene modified
Surgery
prostate cancer vaccine

Vaccination

B
Figure 2-7 Immunogene therapy via ex vivo gene transfer. A, Preclinical models have
shown that vaccination of tumor bearing animals with tumor cells that have been retrovirally
transfected ex vivo to produce immunogene products (in this case GM-CSF) can induce
complete or partial tumor regression at a distant metastatic site (as shown in the illustrated
experiment using hormone-refractory Dunning rat prostate cancer). (Reprinted from Sanda
MG, Ayyagari SR, Jaffee EM, et al: J Urol 1994; 151:622, with permission.) B, Schema of an
analogous human gene therapy protocol being evaluated in an ongoing clinical trial based
on experiments such as that in part-figure A. (Reprinted from Sanda MG, Simons JW:
Urology 1994; 44:617, with permission.)
28 Part I Principles of Urologic Oncology

granulocyte-macrophage colony-stimulating factor genes, and the other entails in vivo delivery of tumor
(GM-CSF) and other immunostimulatory genes into antigen genes by recombinant viral vectors vaccines.
tumor cells used for vaccination led to elimination of In vivo gene transfer of immunostimulatory genes has
preestablished microscopic tumor cell deposits in animal been evaluated using poxvirus and liposomal vectors
models. Antitumor immune mediators (such as helper T encoding GM-CSF, IL-2, IL-12, and other genes for
cells, cytolytic T cells, NK cells, and dendritic cells) are therapy of renal, bladder, and prostate cancer.30,31,48,53
activated by the expression of therapeutic immunostimu- Rather than removing tumor cells to achieve genetic
latory genes in close proximity to tumor-specific antigens modification in vitro and then using the gene-modified
present in the genetically engineered tumor vaccine cells. cells as a vaccine, the gene transfer vector is administered
The immune mediators then circulate and, ideally, erad- directly into tumor in vivo, such as by intravesical instil-
icate distant micrometastases. Preclinical in vivo efficacy lation or intratumoral injection. The transfected tumor
of such gene-modified tumor cell vaccines has also been cells essentially function as an in situ vaccine to induce
shown in several models of urologic malignancy, includ- activity both against the transfected primary tumor site
ing renal cancer, bladder cancer, and prostate cancer (see and distant metastases, without having undergone ex vivo
Figure 2-7).14,40–43 processing and culture. A potential advantage of this
Several clinical trials using immunogene therapy with approach is that genuine tumor antigen expression by the
ex vivo gene transfer specifically for urologic cancers are in vivo-transfected tumor cells is conserved, while inter-
underway or forthcoming (see Table 2-3).15,44,50,51 ference by in vitro artifact antigens is avoided (see Table
Therapeutic genes encoding IL-2 and GM-CSF targeted 2-2). This approach has also been extensively evaluated
prostate cancer or renal cell carcinoma in these studies. with other vector systems in nonurologic tumor models,
In one completed study, no dose limiting toxicities were and clinical trials in urologic and other tumors based on
encountered, and a dose-dependent lymphocyte infiltrate these studies have been undertaken.54–58
was noted at the vaccine site.15 The single patient who Some distinct advantages of bladder cancer and
exhibited a partial response in this phase I study also prostate cancer, specifically, support a focus on these
showed the greatest DTH response in the study group, malignancies with in vivo immunostimulatory gene
suggesting that GM-CSF secreting vaccine cells can transfer. First, regional targeting of localized bladder and
induce tumor-specific immune responses with minimal prostate cancer is potentially readily achieved in these
toxicity. Evaluation of potential clinical efficacy with this sites by either intravesical administration or transrectal
strategy awaits a larger phase II study. prostatic injection. Second, prostate cancer immunogene
A significant limitation of these ex vivo gene transfer therapy poses the possibility of using not only tumor
therapies, however, is the need for cell culture of cancer antigens, but also potentially of normal prostate antigens
cells that serve as targets for gene transfer (see Table 2-2). (such as prostate-specific antigen [PSA], expressed in
Problems associated with the need for cell culture normal and malignant prostate cells alike) as targets of
include: requisite surgery to procure adequate tumor vol- immune effector cells.
umes for vaccine cell production; unreliable tumor cell Use of recombinant vectors encoding specific tumor
yield with regard to both tumor cell number and tumori- antigens as agents for recombinant vaccination differs
genic genotype; and a requirement for cumbersome, from other immunogene therapy strategies in that the
expensive cell culture for each treated subject, limiting viral vector itself provides the antigen to stimulate a
the widespread applicability of this therapy.15,41,52 To cir- tumor-specific immune response. In this setting, the
cumvent these limitations of ex vivo tumor cell culture patient’s tumor cells are not relied on as an effective anti-
for gene transfer, the development of nonretroviral gene gen-presenting cell nor are they a required target of
transfer vectors has led to alternative immunogene ther- direct immunogene transfer. Instead of targeting tumor
apies using in vivo gene transfer techniques. cells as the recipients of the therapeutic gene (as shown
in Figure 2-4), systemically administered recombinant
vector vaccines target professional antigen-presenting
Immunogene Therapy Via In Vivo Gene Transfer
cells as recipients for the therapeutic gene (which in this
The advent of vectors capable of efficient and safe direct strategy encodes a tumor antigen). By using antigen-
gene transfer in vivo, such as poxvirus, adenovirus, and presenting cells, such as dendritic cells, to induce immune
liposome vectors, has provided an avenue for overcoming mediators that then recognize and eliminate tumor cells,
problems unique to ex vivo gene transfer therapies such this strategy avoids potential tumor cell mechanisms for
as the need for tumor cell procurement and culture (see actively suppressing immune induction, such as secretion
Tables 2-1 and 2-2). Two general approaches using in vivo of TGF-β.59–61 Initial studies using recombinant vectors
gene transfer for immunogene therapy have been devel- as tumor-specific vaccines focused on relatively simple
oped through preclinical studies to the arena of clinical vector constructs encoding a specific tumor antigen alone
trials; one entails in vivo transfer of immunostimulatory as the basis for induction of immunity. Along these lines,
Chapter 2 Gene Therapy for Urologic Cancer: Basic Principles, Prospects, and Limitations 29

clinical trials have been conducted using a vaccinia vac- Three general approaches to targeting cell death (inde-
cine encoding PSA for prostate cancer therapy.49,62 The pendent of tumor-specific immunity) are in development.
goal of vaccinia-PSA immune gene therapy is to induce First, gene transfer of a drug susceptibility gene (such as
an immune response against any cells expressing PSA herpesvirus thymidine kinase, HSV-tk) renders target
under the hypothesis that activated PSA-specific T cells cells sensitive to subsequent gancyclovir-mediated cyto-
will kill cancer cells that express PSA (as in the setting of toxicity.70 Second, transfection of cellular toxin genes can
recurrence after radical prostatectomy). Innate immune induce cell injury, disruption, and necrosis. Third, gene
tolerance to PSA as a normal self-antigen, however, will transfer of dominant apoptosis-inducing genes can trigger
need to be overcome to achieve the desired therapeutic organized cellular death, or apoptosis.71,72
effect. Gene transfer of HSV-tk, as a means of rendering
The efficacy of preclinical immunogene therapy stud- tumor cells susceptible to subsequent gancyclovir-
ies should be viewed in context. When evaluated in mediated cytotoxicity, was among the first approaches in
highly lethal, nonimmunogenic tumor models, which efforts to induce tumor cell death via gene transfer.70,73,74
most closely mimic human malignancy, the antitumor In this system, gancyclovir acts as a prodrug that becomes
effect has been modest—in the range of 4-log kill. This cytotoxic only after it is phosphorylated by HSV-tk.
would indicate that clinical efficacy of a gene-modified Mammalian cells normally lack HSV-tk; hence the
tumor cell vaccine approach may potentially be limited to requirement for gene transfer. Phosphorylation of gancy-
an adjuvant setting. In addition, characteristics common clovir by HSV-tk leads to the formation of gancyclovir
among urologic cancers, including deficient class I MHC triphosphate, a potent nucleotide competitor that inter-
expression, overproduction of immunosuppressive TGF-β, feres with DNA synthesis. A bystander effect, whereby
and heterogeneous target tumor antigen expression, all nontransduced adjacent malignant cells are killed in part
represent potential immune evasion mechanisms that to the transfer of the toxic analog via gap junctions or
may impede efficacy of immunogene therapies. The apoptotic vesicles, was initially described in HSV-tk gene
immunogene therapy patient conversely may harbor transfer studies.64 HSV-tk gene transfer can be accom-
generalized limitations to potential immune stimula- plished by retroviral or adenoviral vectors in vivo. Due to
tion.63 In addition, immune responses against the vector ease and efficiency of use, adenoviral vectors have been
backbone may interfere with tumor-specific immune used for HSV-tk gene transfer in animal models of
effectors. A new generation of recombinant vector vac- prostate cancer in which in vivo delivery was accom-
cines seek to address these and other obstacles by com- plished by intratumoral injection.75 Subsequent systemic
bining the advantages of immunostimulatory gene administration of gancyclovir led to significant reduction
transfer and vector-encoded tumor antigen gene transfer of tumor growth. This effect was synergistic with andro-
in vectors designed to deliver two therapeutic genes in gen withdrawal in a mouse model of androgen responsive
one vector: an immunostimulatory gene in tandem with prostate cancer.68 Clinical trials based on these findings
a tumor antigen gene.46 Despite potential obstacles, ther- and using intratumoral injection for delivery have been
apeutic efficacy in the setting of transient gene transfer completed with modest PSA responses.24,76
and durability of tumor-specific immunity comprise A limitation of HSV-tk gene transfer is the need to
advantages of immunogene therapy that fuel continued coordinate and optimize administration of two agents:
clinical development. the sensitizing gene transfer vector and the prodrug gan-
cyclovir. An alternative strategy for direct cytotoxicity is
gene transfer of cellular toxin genes, such as ricin and
Gene Transfer for Direct Induction of Target
diphtheria toxin, which disrupt protein synthesis result-
Cell Death
ing in lethal cellular injury.72 Although ricin gene trans-
Hypothetical barriers to tumor therapy by transfer of cell fer itself has not been applied to urologic tumors, direct
death genes traditionally included the potential need for administration of ricin gene products is cytotoxic to
nearly 100% efficient gene transfer in vivo to achieve human prostate cancer cells, providing rationale for fur-
remission and lack of effective strategies for targeting ther development of this strategy.77 Adenoviruses encod-
tumor cells specifically without concurrent death induc- ing diphtheria toxin have demonstrated remarkable
tion in normal tissues. The availability of vectors that potency at eliminating established tumors;71,78 however,
transfer genes efficiently in vivo, the discovery of the extreme potency of the toxin makes it capable of
bystander effects which allow transmission of cell death killing nontarget cells, even when used with a tissue-
signals to nontransduced cells, the characterization of specific promoter. Hence, unless better transcriptional
organized cell death (apoptosis) pathway abnormalities in control can be achieved, this approach will probably not
cancer cells, and the development of dominant cell death- be readily translated into clinical utility.
inducing genes, however, have all prompted a reevalua- Third and perhaps most promising of the gene ther-
tion of these hypothetical barriers (see Table 2-2).64–69 apy strategies, which aim to directly induce cell death, is
30 Part I Principles of Urologic Oncology

gene transfer of apoptosis-inducing genes. Organized also been encouraging in that they have demonstrated a
cell death in the form of apoptosis differs from toxic or clear dose response, with marked reductions of PSA
necrotic cellular disruption (such as ricin-mediated toxi- occurring in most of the men treated at the higher dose
city) in that apoptosis occurs as a normal entity of the levels. However, the response is limited, because the viral
eukaryotic life cycle in vivo, without concomitant inflam- infection lasts less than 2 weeks and not all of the cancer
mation or other local toxicity. Prostate biology revealed cells are transduced by the replicating virus. Given
some of the earliest evidence for apoptosis as a normal enough time, all the patients with clinical response
component of cellular homeostasis, and prostate cancer relapsed with a PSA progression. Of note, however, is the
was the first among several solid tumors whose growth fact that when these viruses are given directly into the
and progression has been shown to result from defective prostate, the presence of neutralizing antibodies had no
apoptosis rather than augmented proliferation.79 Gene significant clinical impact on the efficacy of the vector.
products involved in a cascade of intracellular events Hence it appears that while such neutralizing antibodies
mediating apoptosis have been successfully targeted for may present a serious obstacle to systemic oncolytic viral
induction of apoptosis in tumor cells. An ideal apoptosis- therapy, it is of a lesser concern with local therapy.
inducing gene would induce apoptosis in tumor cells Recent advances in the understanding of adenoviral
without altering homeostasis in normal cells. Candidate replication have discovered that the molecular pathways
genes that may exhibit such selective effects to some necessary for promoting viral replication are also pivotal
degree include caspases and p53; the ability of adenoviral in terms of sensitizing the cells to chemotherapy and
vectors encoding these genes to induce apoptosis in radiation therapy. The combination of radiation therapy
tumor cells in vitro and in vivo has been shown, and and CV706 results in a 6.7-fold enhancement of tumor
effects on normal cells and stem cells are under intensive reduction over the predicted response from the addition
study.20,22 Early findings indicate that gene transfer of of the two treatments alone.85 This synergy of effect has
Bclx-s with adenovirus vectors, for example, has little led to a robust enthusiasm for further clinical translation.
effect on normal cells while tumor growth was pro- If oncolytic virotherapy can potentiate radiation therapy
foundly affected in vivo.20 significantly, then the acceptance of this combination is
likely to meet with less resistance. As urologists, this
combination would be particularly attractive if the com-
Oncolytic Virotherapy
bined therapy could be administered contemporaneously
During the late 1950s, a variety of viruses were evaluated through a brachytherapy platform. Like other molecular
as cancer therapeutics; however, with the advent of therapies, oncolytic adenoviral gene therapy is still highly
chemotherapy, improved radiotherapy, and the lack of experimental and in its early developmental stages.
specificity of virotherapy, these strategies were largely However, it is becoming increasingly clear that the earli-
abandoned.80 In the early 1990s, however, a resurgence est clinical utility of these methods will be in combina-
in this concept occurred, when a herpesvirus was specifi- tion with conventional therapy and at least initially will
cally engineered to replicate selectively in central nerv- be limited to local-regional delivery.
ous system tumors.81 Subsequently, it was found that
naturally occurring mutants of certain viruses were capa-
Antioncogene Therapy: Approaches Using
ble of selective replication in cancer cells defective in cer-
Antisense and Ribozyme Constructs
tain pathways. In the case of the E1B deleted Onyx-015
virus, replication occurred selectively in those cells that Targeting oncogenes with gene transfer is theoretically
were defective in some way in the p53 axis of apoptosis advantageous because this strategy can potentially selec-
regulation.82 Similarly, others discovered that the tively affect tumor cell growth without affecting normal
reovirus was capable of enhanced replication in those cells, which may lack functionally expression of the target
cells with activated ras.83 However, not all malignancies oncogene. By exploiting the ability of complementary
share the same path to oncogenesis. Hence, efforts were RNA strands to bind to each other, delivery of genes con-
made to develop a conditionally replication-competent taining such complementary or “antisense” sequences to
oncolytic adenovirus (CRAd), which would only replicate specific oncogenes can revert the tumorigenic phenotype
and passively lyse cells when it was in a particular cell by inhibiting expression of specific oncogenes in target
type. The first of these CRAds was CV706, which pref- tumor cells. Interference with translational machinery due
erentially replicated and lysed prostatic epithelial cells by to pairing of antisense RNA constructs with their oncogene-
virtue of the PSA promoter and enhancer that were used encoding RNA targets is one mechanism of antioncogene
to activate the replicative genes, E1A and E1B.84 These activity postulated as active in this strategy. By interfering
CRAds demonstrated excellent activity and specificity in with translation of oncogene RNA, oncogenic proteins
vitro and in vivo and hence were rapidly translated into are produced at much lower levels, if at all. In addition to
clinical trials.23–25 The results of these initial trials have interfering with translation, moreover, antisense constructs
Chapter 2 Gene Therapy for Urologic Cancer: Basic Principles, Prospects, and Limitations 31

may activate endogenous ribonucleases, which in turn example, targeting H-ras in a bladder cancer cell line)
degrade the bound RNA. Regardless of the mechanism, with consequent repression of in vivo tumorigenesis.91,92
the net effect of antisense therapy is the reduction of The efficacy of this strategy in the setting of in vivo gene
oncogenic protein expression due to binding of oncogene delivery remains as yet untested. However, the direct tar-
RNA by the antisense gene product (see Table 2-2). get specificity of ribozyme-targeted antioncogene ther-
Although early antisense strategies focused on direct apy, in the setting of a well-characterized effector
administration of short antisense oligonucleotides (some- mechanism, suggests that ribozyme-based strategies may
times modified for improved solubility), more recently be the most promising antisense-based therapeutic strat-
the delivery of longer antisense constructs, as well as egy under current development.
dominant negative mutation constructs, via recombinant
vector systems has emerged.86–88 Retroviral transfer of a
Tumor Suppressor Gene Restoration
myc antisense gene (delivered by direct injection of retro-
viral vector into small prostate cancer nodules in The observation that renal cell cancer tumorigenicity can
rodents), for example, was found to impede in vivo be reversed by in vitro transfer of the von Hippel-Lindau
prostate cancer growth in a rodent model.89 Based on (VHL) gene prior to full biochemical and functional
these findings, a clinical trial of antioncogene therapy characterization of the VHL gene product attests to the
using intraprostatic injection of retroviral vector encod- potential utility of gene therapy targeting tumor suppres-
ing antisense myc has been proposed.89 sor gene restoration.93,94 This observation indicates that
The discovery of ribozymes, or RNA sequences, that restoring tumor suppressor genes may reverse tumori-
catalyze RNA cleavage and splicing, opened a promising genic potential of individual, in vitro transduced cells.
extension of gene therapy strategies based on oncogene Tumor suppressor gene transfer in vivo, however, has had
targeting via antisense recognition of oncogene RNA less impressive effects than in vitro transfection.95 At least
(Figure 2-8).90 Ribozymes can be designed to degrade two factors may contribute to the discrepancy between in
RNA containing a short segment of complementary vitro and in vivo effects of tumor suppressor gene
nucleotides. In theory, almost any RNA containing a restoration: first, intratumoral injection of vectors into
unique 15-base pair or longer sequence can be specifi- solid tumors is not a highly efficient approach for gene
cally degraded by designing a ribozyme containing a delivery—most of the vector is likely cleared before it
complementary binding motif. Adenoviral vectors have accesses tumor cells, and the initial vector distribution in
been used to deliver oncogene specific ribozymes (for injected tissue is unlikely to be uniform. Second, stable

Figure 2-8 Antioncogene ribozyme consensus sequence. The hammerhead ribozyme


contains three nonconserved helical regions (stems I, II, and III) along with the conserved
sequence of the central core. Stems I and III, which determine the specificity of the ribozyme
for its target, hybridize to target oncogene RNA. The target RNA is then cleaved at the site
indicated by the arrow, disabling oncogene expression. Nucleotides designated as N can be
any nucleotide. (Reprinted from Thompson JD, Macejak D, Couture L, Stinchcomb DT: Nat
Med 1995; 1:277, with permission.)
32 Part I Principles of Urologic Oncology

long-term integration (as with a retrovirus vector) of the adenovirus vector has been proposed (see Table 2-3).
therapeutic suppressor gene, in the setting of 100% effi- The association of Rb gene abnormalities in bladder can-
cient in vivo transduction, would be required to arrest cer and poor prognosis supports the rationale for intrav-
tumor growth (see Table 2-2). esical adenovirus-Rb gene therapy.96
This strategy could be optimized by using vectors For many tumor suppressor genes, restoration of sup-
capable of stably integrating the transgene into the target pressor gene function alone may not suffice for cytore-
cell genome, such as retroviral vectors, and also capable duction of established tumors even in the theoretical
of highly efficient in vivo transduction, such as adenovi- setting of totally effective and durable in vivo tumor sup-
ral vectors. In that no vector currently has both of these pressor gene transfer. Most tumor suppressor genes do
characteristics (see Table 2-1), using any vector system not encode signals for direct induction of cell death but
will have limited efficacy at present. For example, one rather affect tumor growth more indirectly, such as by
potential tumor suppressor target, c-cam, is a cellular regulating DNA repair, cellular attachment, or cell cycle
attachment molecule, which is absent in some prostate control.97 In this setting, restoration of normal suppres-
cancers, and thereby potentially contributes to the unin- sor gene via gene transfer may require accompanying
hibited and metastatic growth potential of these cells. cytoreductive systemic or regional therapies (chemotherapy,
Intratumoral injection of an adenoviral vector encoding radiation) to treat established tumors. The need for
c-cam, used to restore expression of this molecule in accompanying cytoreductive therapy is further evidenced
preestablished prostate cancer xenografts, slowed but did by the transient expression associated with the most effi-
not reverse tumor progression (Figure 2-9).95 Similar cient vector systems—once transgene expression fades,
effects have been seen with adenovirus vector-based ther- the tumorigenic phenotype associated with absence of
apy targeting restoration of other tumor suppressor genes. the suppressor gene will reappear (efficient in vivo vec-
Despite these limitations, survival of tumor-bearing ani- tors are required for this strategy as most, if not all, tar-
mals can be extended with in vivo suppressor gene get cells must directly express, or confer expression via
restoration therapy, and a clinical trial that evaluates the bystander effect, of the transferred gene for effective
efficacy of Rb gene delivery via intravesical instillation of tumor reduction)

Figure 2-9 Tumor suppressor gene therapy inhibits tumor growth in animal models.
Injection of adenovirus encoding c-cam1 (filled circles) into human prostate cancer nodules
grown in nude mice reduced tumor growth compared to saline (filled triangle) and vector
(open circle) controls. Delay of tumor growth without complete tumor remission is typical of
strategies relying on local-regional injection of recombinant vectors encoding therapeutic
tumor suppressor, antioncogene, or cytotoxicity genes. (Reprinted from Kleinerman DI,
Zhang WW, Lin SH, et al: Cancer Res 1995; 55:2831, with permission.)
Chapter 2 Gene Therapy for Urologic Cancer: Basic Principles, Prospects, and Limitations 33

Some tumor suppressor genes may also serve as gate- cells or tissues has therefore been achieved principally via
keepers for intracellular apoptosis-inducing signals. In the route of vector administration. The feasibility of con-
addition to suppressing the tumorigenic growth potential ferring specificity via the administration route, in the case
of individual cells, restoration of these tumor suppressor of urologic targets, has been demonstrated for renal cancer
genes should also be able to reduce established tumors after renal tubule vector infusion and for bladder epithe-
via apoptosis induction. The inability of an adenoviral lium after intravesical instillation of viral vectors.98,99 This
vector encoding p53 to eliminate preestablished malig- approach may be applicable to local-regional therapy of
nancy, however, indicates that near-complete transduc- early stage, organ-confined malignancy (see Table 2-2).
tion of all tumor cells may be required for optimal Systemic applications of cytotoxic, antioncogene, or
therapeutic effect. This level of efficiency is clearly not tumor suppressor therapeutic gene transfer, however, will
achieved by direct solid tumor injection with currently require specific targeting based not only on vector
available vectors. Improvements in vector and delivery administration route but also on molecular rather than
systems will be needed to optimize this, and other, gene mechanical targeting. Molecular vector targeting can be
therapy strategies that rely on direct effects of gene trans- achieved either by modifying tropism (altering the affin-
fer in the tumor cell target. ity of the vector coat for attachment and entry to a lim-
ited range of human target cells) or by restricting
transcription (constructing expression cassettes contain-
FUTURE DIRECTIONS: TARGETING VECTOR
ing promoters with selective activity in different tissues)
SPECIFICITY
(Figure 2-10).
Most vectors, which, at present, have shown functional
efficacy in vivo, lack significant specificity in target cell
Modified Vector Tropism
attachment or restriction in transgene expression. The
vector envelopes or coats of retroviruses, adenoviruses, Two approaches have been used for modifying vector
and liposomal vectors enter cells via families of receptors tropism. First, vectors can be derived from viruses with
that, as a group, are virtually ubiquitous. The promoters inherent tropism for a specific tissue target. Due to the
controlling transgene expression in these vectors are typ- relative paucity of molecular characterization of viruses
ically potent promoters susceptible to little or no regula- with natural and specific tropism for the genitourinary
tion by the host cell. Targeting of gene therapy to specific tract, this approach has limited utility. Nevertheless, at

Figure 2-10 Restricting target cell specificity of recombinant viral vectors. The ability
to specifically target gene delivery can facilitate systemic gene therapy with cytotoxic
vectors. Approaches to confer specificity include: (A) engineering vector coat specificity;
(B) restricting promoter-regulated transcription; (C) chemically modifying vector-target affinity.
(Based on Miller N, Vile R: FASEB J 1995; 9:190.)
34 Part I Principles of Urologic Oncology

least one virus (BK virus, which has specific tropism for In contrast to tissue-specific transcriptional targeting,
transitional epithelium) has shown potentially useful oncogene-associated regulatory sequences may promote
tropism specificity for transitional epithelium.100 selective expression of therapeutic genes in tumor cells
Recombinant BK episomal vectors were constructed that that harbor transcriptional overexpression of the onco-
led to reporter gene expression specifically in human gene. This has been demonstrated with a vector using
transitional cell (TCC) lines that are relative to absent ERBB2 promoter sequences to the cytotoxic gene cyto-
expression in other tumor cells. Limited characterization sine deaminase; this vector conferred selective sensitivity
of the elements regulating BK specificity and lack of repli- on ERBB2-overproducing cells.112
cation defective BK vectors, however, has limited further
development of this vector system thus far. SUMMARY
Second, molecular engineering and conjugate forma-
Based on a growing volume of preclinical data, clinical
tion to alter the native vector coats has been used to con-
trials of gene therapy for urologic cancer are underway.
fer specific tropism. In regards to retroviral vectors,
Therapeutic genes that are under current or forthcoming
engineering of envelope or vector coat sequences
clinical study include immunogenes, cell death-inducing
(pseudotyping) has been limited, principally due to the
genes, antioncogenes, and tumor suppressor genes.
potential of functionally disrupting the ability of engi-
Although systemic therapy with immunogenes is feasible,
neered envelopes to mediate target cell attachment and
other gene therapy strategies, which do not rely on an
entry. Pseudotyping has thus succeeded in producing
intervening antitumor immune response, are at present
vectors with extended or altered target cell tropism,
limited to local-regional targeting. This constraint is
without more restricted target specificity per se.11 In
largely due to limitations of gene transfer vectors, as well
contrast, a detailed analysis of the fiber gene has allowed
as in vivo gene delivery systems. Refinement of gene
the alteration of adenoviral tropism by either changing
transfer vectors, such as hybrid vectors construction, is
the knob domains for different adenoviral serotypes, or
actively being pursued to broaden the utility and applica-
by specifically introducing targeting ligands into portions
bility of direct gene therapy strategies.
of the knob that are unimportant for fiber folding and
The early phase of cancer gene therapy clinical trials
viral assembly.101,102 Molecular conjugate formation has
should be viewed in context. Preclinical models predict
successfully conferred altered and refined specificity to
modest, if any, therapeutic effects with current forms of
adenoviral, liposomal, and retroviral vectors, alike. This
human cancer gene therapy. Equally as important as clin-
has been achieved via covalent linkage of vectors with lig-
ical outcome in gene therapy clinical trials, however, are
ands such as growth factor receptors or antibody haptens,
biologic surrogate endpoints to guide continued
which confer the desired tropism for cells expressing the
improvement of gene therapy strategies. The earliest
specific receptor and via noncovalent association of
clinical trials have indeed shown the ability of clinical
hybrid vector components.103–106
gene therapy to alter biology of human urologic can-
cer.15,23–25 However, groundbreaking clinical trials of
Restricted Transgene Expression: gene therapy have also been associated with significant
Transcriptional Targeting toxicity, including a fatality and induction of vector-
induced leukemia.2,3 To attain its full potential, gene
An alternative to vector targeting at the target cell bind-
therapy must be approached with realistic expectations,
ing level is to limit expression of therapeutic genes by
respect for its potential toxicity, and a recognition of the
regulating transcription with a promoter region having
need for its continued refinement.
either tissue restricted activity, or preferential activity in
malignant cells. Such promoter-regulated specificity has
been used to target retroviral and adenoviral vectors
alike.11,107 Tissue-specific promoters are potentially REFERENCES
useful for regulating expression of cytotoxic genes in
vectors targeting nonvital tissues, such as prostate, to 1. Watson JD, Crick FH: Molecular structure of nucleic
widen potential therapeutic windows.108 To this end, acids. A structure for deoxyribose nucleic acid. Nature
vectors have been constructed with the promoter region 1953; 171:737.
2. Marshall E: Gene therapy death prompts review of
that normally regulates PSA expression used to control
adenovirus vector. Science 1999; 286: 2244–2245.
expression of a reporter gene.109,110 Cloning of thera-
3. Check E: Harmful potential of viral vectors fuels doubts
peutic cytotoxic genes, such as HSV-tk, into analogous over gene therapy. Nature 2003; 423:573–574.
vectors using tyrosinase promoter has been shown to 4. Danos O, Mulligan RC: Safe and efficient generation
inhibit tumor growth in melanoma animal models111; of recombinant retrovirus with amphotropic and
analogous vectors to target prostate cancer are under ecotropic host ranges. Proc Natl Acad Sci USA 1988;
development. 85:6460.
Chapter 2 Gene Therapy for Urologic Cancer: Basic Principles, Prospects, and Limitations 35

5. Ghosh-Choudhury G, Haj-Ahmad Y, Brinkley P, et al: cancer following radiation therapy. Cancer Res 2001;
Human adenovirus cloning vectors based on infectious 61:7464.
bacterial plasmids. Gene 1986; 50:161. 24. Miles BJ, Shalev M, Aguilar-Cordova E, et al: Prostate-
6. Moss B: Vaccinia virus: a tool for research and vaccine specific antigen response and systemic T cell activation
development. Science 1991; 2252:1662. after in situ gene therapy in prostate cancer patients
7. Ledley FD: Nonviral gene therapy: the promise of genes failing radiotherapy. Hum Gene Ther 2001; 12:
as pharmaceutical products. Hum Gene Ther 1995; 1955-67.
6:1129. 25. Freytag SO, Khil M, Stricker H, et al: Phase I study of
8. Mulligan RC: The basic science of gene therapy. Science replication-competent adenovirus-mediated double
1993; 260:926. suicide gene therapy for the treatment of locally recurrent
9. Crystal RG: Transfer of genes to humans: early lessons prostate cancer. Cancer Res 2002; 62:4968–76.
and obstacles to success. Science 1995; 270:404. 26. Wang M, Bronte V, Chen PW, et al: Active
10. Yang NS, Sun WH: Gene gun and other non-viral immunotherapy of cancer with a non-replicating
approaches for cancer gene therapy. Nat Med 1995; 1:481. recombinant fowlpox virus encoding a model tumor-
11. Miller N, Vile R: Targeted vectors for gene therapy. associated antigen. J Immunol 1995; 154: 4685.
FASEB J 1995; 9:190. 27. Nabel GJ, Nabel EG, Yang ZY, et al: Direct gene transfer
12. Rosenberg SA, Aebersold P, Cornetta K, et al: Gene with DNA-liposome complexes in melanoma: expression,
transfer into humans – immunotherapy of patients with biologic activity, and lack of toxicity in humans. Proc Natl
advanced melanoma, using tumor-infiltrating lymphocytes Acad Sci USA 1993; 90:11307.
modified by retroviral gene transduction. N Engl J Med 28. Ogris M, Wagner E: Tumor-targeted gene transfer
1990; 323:570. with DNA polyplexes. Somat Cell Mol Genet 2002;
13. Blaese M: The ADA human gene therapy clinical 27:85–95.
protocol. Hum Gene Ther 1990; 1:327. 29. Thomas M, Klibanov AM. Non-viral gene therapy:
14. Dranoff G, Jaffee E, Lazenby A, et al: Vaccination with polycation-mediated DNA delivery. Appl Microbiol
irradiated tumor cells engineered to secrete murine Biotechnol 2003; 62:27–34.
GM-CSF stimulates potent, specific, and long lasting 30. Sun WH, Burkholder JK, Sun J, et al: In vivo cytokine
anti-tumor immunity. Proc Natl Acad Sci USA 1993; gene transfer by gene gun reduces tumor growth in mice.
90:3539. Proc Natl Acad Sci USA 1995; 92:2889.
15. Simons JW, Jaffee EM, Weber C, et al: Bioactivity of 31. Vogelzang NJ, Sudakoff G, McKay S, et al: A phase I
human GM-CSF gene therapy using autologous study of intralesional (IL) gene therapy in metastatic renal
irradiated renal cell carcinoma vaccines. Submitted. cell cancer (RCC). Proc Annu Meet Am Soc Clin Oncol
16. Yang Y, Nunes F, Berencsi K, et al: Cellular immunity to 1995; 14: A641.
viral antigens limits E1-deleted adenoviruses for gene 32. Johnson PA, Miyanohara A, Levine F, et al: Cytotoxicity
therapy. Proc Natl Acad Sci USA 1994; 91:4407. of a replication-defective mutant of herpes simplex virus
17. Wickham TJ, Mathias P, Cheresh DA, Nemerow GR: type I. J Virol 1992; 66:2952.
Integrins αVβ5 and αVβ3 promote adenovirus 33. Dupont F, Tenenbaum L, Guo LP, et al: Use of an
internalisation but not virus attachment. Cell 1993; autonomous parvovirus vector for selective transfer of a
73:309. foreign gene into transformed human cells of different
18. Seth P: Adenovirus-dependent release of choline from origin and its expression therein. J Virol 1994; 68:1397.
plasma membrane vesicles at an acidic pH is mediated by 34. Flotte TR, Afione SA, Conrad C, et al: Stable in vivo
the penton base protein. J Virol 1994; 68:1204. expression of the cystic fybrosis transmembrane
19. Addison CL, Braciak T, Ralston R, et al: Intratumoral conductance regulator with an adeno-associated virus
injection of an adenovirus expressing IL-2 induces vector. Proc Natl Acad Sci USA 1993; 90:10613.
regression and immunity in a murine breast cancer model. 35. Pan ZK, Ikonomidis G, Lazenby A, et al: A recombinant
Proc Natl Acad Sci USA 1995; 92:8522. Listeria monocytogenes vaccine expressing a model tumor
20. Shariat SF, Desai S, Song W, Khan T, Zhao J, Nguyen C, antigen protects mice against lethal tumour cell challenge
Foster BA, Greenberg N, Spencer DM, Slawin KM: and causes regression of established tumours. Nat Med
Adenovirus-mediated transfer of inducible caspases: a 1995; 1:471.
novel “death switch” gene therapeutic approach to 36. Fearon ER, Pardoll DM, Itaya T, et al: Interleukin-2
prostate cancer. Cancer Res 2001; 61:2562–2571. production by tumor cells bypasses T helper function in
21. Eastham JA, Hall SJ, Sehgal I, et al: In vivo gene therapy the generation of an antitumor response. Cell 1990;
with p53 or p21 adenovirus for prostate cancer. Cancer 60:397.
Res 1995; 55:5151. 37. Gansbacher B, Gansbacher B, Bannerji R, Daniels B, et al:
22. Yang C, Cirielli C, Capogrossi MC, Passaniti A: Retroviral vector-mediated gamma-interferon gene
Adenovirus-mediated wild-type p53 expression induces transfer into tumor cells generates potent and long lasting
apoptosis and suppresses tumorigenesis of prostatic tumor antitumor immunity. Cancer Res 1990; 50:7820.
cells. Cancer Res 1995; 55(19):4210. 38. Gansbacher B, Zier K, Daniels B, et al: Interleukin
23. Weese TL, van der Poel H, Li S, et al: A phase I trial of 2 gene transfer into tumor cells abrogates tumorigenicity
CV706, a replication-competent, PSA selective oncolytic and induces protective immunity. J Exp Med 1990;
adenovirus, for the treatment of locally recurrent prostate 172:1217.
36 Part I Principles of Urologic Oncology

39. Golumbek PT, Lazenby AJ, Levitsky HI, et al: Treatment 54. Cordon-Cardo C, Fuks Z, Drobnjak M, et al: Expression
of established renal cell cancer by tumor cells engineered of HLA-A,B,C antigens on primary and metastatic tumor
to secrete interleukin-4. Science 1991; 254:713. cell populations of human carcinomas. Cancer Res 1991;
40. Connor J, Bannerji R, Saito S, et al: Regression of bladder 51:6372.
tumors in mice treated with interleukin-2 gene modified 55. Blades RA, Keating PJ, McWilliam LJ, et al: Loss of HLA
tumor cells. J Exp Med 1993; 177:1127. class I expression in prostate cancer: implications for
41. Sanda MG, Ayyagari SR, Jaffee EM, et al: Demonstration immunother-apy. Urology (in press).
of a rational strategy for human prostate cancer gene 56. Nouri AM, Hussain RF, Oliver RT: The frequency of
therapy. J Urol 1994; 151:622. major histo-compatibility complex antigen abnormalities
42. Moody DB, Robinson JC, Ewing CM, et al: Interleukin-2 in urological tumours and their correction by gene
transfected prostate cancer cells generate a local transfection or cytokine stimulation. Cancer Gene Ther
antitumor effect in vivo. Prostate 1994; 24:244. 1994; 1:119.
43. Vieweg J, Rosenthal FM, Bannerji R, Heston WDW, 57. Sanda MG, Restifo NP, Walsh JC, et al: Molecular
Fair WR, Gansbacher B, Gilboa E: Immunotherapy of characterization of defective antigen processing in human
prostate cancer in the Dunning rat model: use of prostate cancer. J Natl Cancer Inst 1995; 87:280.
cytokine gene modified tumor vaccines. Cancer Res 58. Lattime EC: Therapy of muscle-invasive bladder
1994; 54:1760. carcinoma with intravesical vaccinia. FDA Approval 1996:
44. Gansbacher B, Motzer R, Houghton A, Bander N: BB-IND-5002.
Immunization with Interleukin-2 secreting allogeneic 59. Torre-Amione G, Beauchamp RD, Koeppen H, et al: A
HLA-A2 matched renal cell carcinoma cells in patients highly immunogenic tumor transfected with a murine
with advanced renal cell carcinoma. RAC Report 1992; transforming growth factor type beta 1 cDNA escapes
9206-022. immune surveillance. Proc Natl Acad Sci USA 1990;
45. Kantor J, Irvine K, Abrams S, et al: Antitumor activity 87:1486.
and immune responses induced by a recombinant 60. Inge TH, Hoover SK, Susskind BM, et al: Inhibition of
carcinoembryonic antigen-vaccinia virus vaccine. J Natl tumor-specific cytotoxic T-lymphocyte responses by
Cancer Inst 1992; 84:1084. TGF-beta 1. Cancer Res 1992; 52:1386.
46. Bronte V, Tsung K, Rao JB, et al: Il-2 enhances the 61. Miyamoto H, Kubota Y, Shuin T, et al: Expression of
function of recombinant poxvirus-based vaccines in the transforming growth factor-beta 1 in human bladder
treatment of established pulmonary metastases. cancer. Cancer 1995; 75:2565.
J Immunol 1995; 154:5282. 62. Eder JP, Kantoff PW, Roper K, et al: A phase I trial of a
47. Lee SS, Eisenlohr LC, McCue PA, et al: Intravesical gene recombinant vaccinia virus expressing prostate-specific
therapy: in vivo gene transfer using recombinant vaccinia antigen in advanced prostate cancer. Clin Cancer Res
virus vectors. Cancer Res 1994; 54:3325. 2000; 6:1632-1638.
48. Lee SS, Eisenlohr LC, McCue PA, et al: In vivo gene 63. Catalona WJ, Chretien PB, Trahan EE: Abnormalities of
therapy of murine tumors using recombinant vaccinia cell-mediated immuno-competence in genitourinary
virus encoding GM-CSF. Proc Annu Meet Am Assoc cancer. J Urol 1974; 111:229–232.
Cancer Res 1995; 36:A1481. 64. Freeman SM, Abboud CN, Whartenby KA, et al: The
49. Sanda MG, Smith D, Charles LG, et al: Recombinant bystander effect: tumor regression when a fraction of the
vaccinia-PSA can induce a prostate-specific immune tumor mass is genetically modified. Cancer Res 1993;
response in androgen-modulated human prostate cancer. 53:5274.
Urology1999; 53:260–266. 65. Symonds H, Krall L, Remington L, et al: p53-dependent
50. Figlin RA: Phase I study of HLA-B7 plasmid apoptosis suppresses tumor growth and progression in
DNA/DMRIE/DOPE lipid complex as an vivo. Cell 1994; 78:703.
immunotherapeutic agent in renal cell carcinoma by 66. Oltvai ZN, Milliman CL, Korsmeyer SJ: Bcl-2
direct gene transfer with concurrent low dose bolus IL-2 heterodimerizes in vivo with a conserved homolog, Bax,
protein therapy. RAC Report 1995; 9508–121. that accelerates programmed cell death. Cell 1993;
51. Paulson D, Lyerly HK: A phase I study of autologous 74:609.
human IL-2 gene modified tumor cells in patients with 67. Boise LH, Gonzalez-Garcia M, Postema CE, et al: Bcl-x,
locally advanced or metastatic prostate cancer. RAC a bcl-2-related gene that functions as a dominant
Report 1995; 9510-132. regulator of apoptotic cell death. Cell 1993; 74:597.
52. Lahn M, Kohler G, Kulmburg P, et al: Parameters for 68. Raffo AJ, Perlman H, Chen MW, et al: Overexpression of
successful establishment of primary and long-term tumor Bcl-2 protects prostate cancer cells from apoptosis in vitro
cell cultures from renal cell carcinoma, melanoma and and confers resistance to androgen-depletion in vivo.
colon carcinoma for cellular immunotherapy. Gene Ther Cancer Res 1995; 55: 4438.
1994; 1:S15. 69. Martin S, Green DR: Apoptosis and cancer: failure of
53. Kawakita M, Rao G, Ritchey JK, et al: Canary-pox virus- controls on cell death and cell survival. Crit Rev Oncol
mediated cytokine gene therapy induces tumor specific Hematol 1995; 18:137.
and non-specific immunity against mouse prostate tumor. 70. Furman PA, McGuirt PV, Keller PM, et al: Inhibition by
J Urol 1996; 155:516A. acyclovir of cell growth and DNA synthesis of cells
Chapter 2 Gene Therapy for Urologic Cancer: Basic Principles, Prospects, and Limitations 37

biochemically transformed with herpes virus genetic 86. McManaway ME, Neckers LM, Loke SL, et al: Tumor-
information. Virology 1980; 102:420. specific inhibition of lymphoma growth by an antisense
71. Li Y, McCadden J, Ferrer F, et al: Prostate-specific oligodeoxynucleotide. Lancet 1990; 335:808.
expression of the diphtheria toxin A chain (DT-A): studies 87. Ogiso Y, Sakai N, Watari H, et al: Suppression of various
of inducibility and specificity of expression of prostate- human tumor cell lines by a dominant negative H-ras
specific antigen promoter-driven DT-A adenoviral- mutant. Gene Ther 1994; 1:403.
mediated gene transfer. Cancer Res 2002; 62:2576-82. 88. Georges RN, Mukhopadhyay T, Zhng Y, et al:
72. Hoganson DK, Batra RK, Olsen JC, Boucher RC: Prevention of orthotopic human lung cancer growth by
Comparison of the effects of three different toxin genes intratracheal instillation of a retroviral antisense k-ras
and their levels of expression on cell growth and bystander construct. Cancer Res 1993; 53:1743.
effect in lung adenocarcinoma. Cancer Res 1996; 56:1315. 89. Steiner MS and Holt JT: Gene therapy for the treatment
73. Moolten FL, Wells JM: Curability of tumors bearing of advanced prostate cancer by in vivo transduction with
herpes thymidine kinase genes transferred by retroviral prostate-targeted retroviral vectors expressing antisense
vectors. J Natl Cancer Inst 1990; 82:297. c-myc RNA. RAC Report 1995; 9509–123.
74. Culver KW, Ram Z, Wallbridge S, et al: In vivo gene 90. Thompson JD, Macejak D, Couture L, Stinchcomb DT:
transfer with retroviral vector-producer cells for treatment Ribozymes in gene therapy. Nat Med 1995; 1:277.
of experimental brain tumors. Science 1992; 256:1550. 91. Kashani-Sabet M, Funato T, Tone T, et al: Reversal of
75. Hall SJ, Mutchnik SE, Shaker M, et al: Adenovirus the malignant phenotype by an anti-ras ribozyme.
mediated HSV-tk gene transduction and gancyclovir Antisense Res Dev 1992; 2:3.
treatment for prostate cancer: suppression of metastasis in 92. Feng M, Cabrera G, Deshane J, et al: Neoplastic
an orthotopic model and synergism with androgen reversion accomplished by high efficiency adenoviral-
ablation. J Urol 1996; 155:528A. mediated delivery of an anti-ras ribozyme. Cancer Res
76. Scardino PT, Thompson TC, Woo SLC: Phase I study of 1995; 55:2024.
adenoviral vector delivery of the HSV-tk gene and the 93. Chen F, Kishida T, Duh FM, et al: Suppression of growth
intravenous administration of gancyclovir in men with of renal carcinoma cells by the von Hippel-Lindau tumor
local recurrence of prostate cancer after radiation therapy. suppressor gene. Cancer Res 1995; 55:4804.
RAC Report 1996; 9601-144. 94. Iliopoulos O, Kibel A, Gray S, Kaelin WG: Tumour
77. Rodriguez R, Carducci MA, Bartkowski LM, Simons JW. suppression by the von Hippel-Lindau gene product.
Cytoreductive agents for prostate cancer gene therapy. Nat Med 1995; 1:822.
Proc Am Assoc Cancer Res 1996; 37:346. 95. Kleinerman DI, Zhang WW, Lin SH, et al: Application
78. Lee EJ, Jameson JL: Cell-specific Cre-mediated activation of a tumor suppressor (C-CAM1)-expressing
of the diphtheria toxin gene in pituitary tumor cells: recombinant adenovirus in androgen-independent
potential for cytotoxic gene therapy. Hum Gene Ther human prostate cancer therapy: a preclinical study.
2002; 13:533–542. Cancer Res 1995; 55:2831.
79. Kyprianou N, Isaacs JT: Activation of programmed cell 96. Small EJ, Carroll PR: Gene therapy of bladder cancer
death in the rat ventral prostate after castration. using recombinant adenovirus containing the
Endocrinology 1988; 122:552. retinoblastoma gene (ACNRB): a phase I study. RAC
80. Southam C: Present status of oncolytic virus studies. Ann Report 1996; 9601–145.
New York Acad Sci 1960; 656–673. 97. Cordon-Cardo C, Dalbagni G, Sarkis AS, Reuter VE:
81. Martuza RL, Malick A, Markert JM, Ruffner KL, Coen Genetic alterations associated with bladder cancer.
DM: Experimental therapy of human glioma by means of Important Adv Oncol 1994; 71.
a genetically engineered virus mutant. Science 1991; 98. Moullier P, Friedlander G, Calise D, et al: Adenoviral-
252:854–856. mediated gene transfer to renal tubular cells in vivo.
82. Bischoff JR, Kirn DH, Williams A, et al: An adenovirus Kidney Int 1994; 45:1220.
mutant that replicates selectively in p53-deficient human 99. Bass C, Cabrera G, Elgavish A, et al: Recombinant
tumor cells. Science 1996; 274:373-376. adenovirus-mediated gene transfer to genitourinary
83. Strong JE, Coffey MC, Tang D, Sabinin P, Lee PW: The epithelium in vitro and in vivo. Cancer Gene Ther 1995;
molecular basis of viral oncolysis: usurpation of the Ras 2:97.
signaling pathway by reovirus. EMBO J 1998; 100. Cooper MJ, Miron S: Efficient episomal expression
17:3351–3362. vector for human transitional carcinoma cells. Hum
84. Rodriguez R, Schuur ER, Lim HY, Henderson GA, Gene Ther 1993; 4:557.
Simons JW, Henderson DR. Prostate attenuated 101. Gall J, Kass-Eisler A, Leinwand L, Falck-Pedersen E:
replication competent adenovirus (ARCA) CN706: a Adenovirus type 5 and 7 capsid chimera: fiber
selective cytotoxic for prostate-specific antigen-positive replacement alters receptor tropism without affecting
prostate cancer cells. Cancer Res 1997; 57:2559–2263. primary immune neutralization epitopes. J Virol 1996;
85. Chen Y, DeWeese T, Dilley J, et al: CV706, a prostate 70:2116–2123.
cancer-specific adenovirus variant, in combination with 102. Krasnykh VN, Mikheeva GV, Douglas JT, Curiel DT:
radiotherapy produces synergistic antitumor efficacy Generation of recombinant adenovirus vectors with
without increasing toxicity. Cancer Res 2001; 61: modified fibers for altering viral tropism. J Virol 1996;
5453–5460. 70:6839–6846.
38 Part I Principles of Urologic Oncology

103. Wu GY, Zhan P, Sze LL, et al: Incorporation of expression of tissue-specific promoters: an analysis of
adenovirus into a ligand-based DNA carrier system prostate-specific promoters. Cancer Gene Ther 2001;
results in retention of original receptor specificity and 8:927–935.
enhances targeted gene expression. J Biol Chem 1994; 109. Taneja SS, Belldegrun A, Dardashti K, et al: In vitro
269:11542. target specific gene therapy for prostate cancer utilizing
104. Chen J, Gamou S, Takayanagi A, Shimuzu N: A novel a prostate specific antigen promoter-driven adenoviral
gene delivery system using EGF receptor mediated- vector. Proc Annu Meet Am Assoc Cancer Res 1994;
endocytosis. FEBS Lett 1994; 338:167. 35:A2236.
105. Michael SI, Huang CH, Romer MU, et al: Binding- 110. Ko SC, Gotoh A, Kao C, et al: Tissue targeted toxic
incompetent adenovirus facilitates molecular conjugate- gene therapy for an androgen-independent and
mediated gene transfer by the receptor-mediated metastatic human prostate cancer model. Proc Am Assoc
endocytosis pathway. J Biol Chem 1993; 268:6866. Cancer Res 1996; 37:349.
106. Vieweg J, Boczkowski D, Roberson KM, et al: 111. Vile RG, Hart IR: Use of tissue-specific expression of
Efficient gene transfer with adeno-associated virus-based the herpes simplex virus thymidine kinase gene to inhibit
plasmids complexed to cationic liposomes for gene growth of established murine melanomas following
therapy of human prostate cancer. Cancer Res 1995; direct intra-tumoral injection of DNA. Cancer Res 1993;
55:2366. 53:3860.
107. Friedman JM, Babiss LE, Clayton DF, Darnell JE: 112. Harris JD, Gutierrez AA, Hurst HC, et al: Gene therapy
Cellular promoters incorporated into the adenovirus for cancer using tumor-specific prodrug activation. Gene
genome: cell specificity of albumin and Ther 1994; 1:170.
immunogloobulin expression. Mol Cell Biol 1986; 113. Sanda MG, Simons JW: Gene therapy for urological
6:3791. cancer. Urology 1994; 44:617.
108. van der Poel HG, McCadden J, Verhaegh GW, et al: A
novel method for the determination of basal gene
C H A P T E R

3 Principles and Applications


of Radiation Oncology
Steven J. Chmura, MD, PhD, Wendla Silverberg, MD,
and Ralph R. Weichselbaum, MD

The treatment of both benign and malignant diseases malignancies. While earlier technologies utilized gamma
with ionizing radiation (IR) began shortly after the dis- rays from radioactive sources, such as cobalt (Co60) to
covery of x-rays by Wilhelm Roentgen in 1895.1 The deliver photon therapy, modern linear accelerators
therapeutic applications of IR were quickly realized when (Figure 3-1) generate and deliver either high-energy
the first patient was treated by Emil Grubbe in 1896.2 photons (x-rays) or charged particles (electrons). The
Despite the initial enthusiasm in the clinical applications x-rays are produced through the deceleration of high-
of IR, subsequent experimental and clinical experience kinetic energy electrons (bremsstrahlung) within the
demonstrated the adverse effects on normal tissues when head of the linear accelerator as they strike a tungsten
attempting to treat tumors located deep below the skin. target.3 After striking the target, the electrons emit x-rays
The development of implantable radioactive sources with a spectrum ranging from zero energy to their maxi-
(brachytherapy) permitted high doses of radiation to be mum kinetic energy. The photons are emitted from a
delivered directly to the tumor tissue while decreasing point source, much like a flashlight, that diverges in a
the normal tissue toxicity with the first prostate patient cone shape. The energy of the photons decreases as the
treated in 1909. The introduction of high-energy (mega- inverse square of the distance (1/d2) from the source (the
voltage) external beam radiation therapy (EBRT) in the inverse square law).3 Through advancements in technol-
1950s allowed treatment to a higher dose of tumors deep ogy, the energy of radiation therapy has been greatly
within the body, while decreasing the surface dose to the increased since its clinical introduction, thus permitting
skin. Despite the advances in both the imaging of tumors treatment of tumors deep within the body.
and the hardware and software employed to deliver radi- The beam quality or energy employed in a particular
ation therapy, normal tissue toxicity remains the limiting patient refers to the highest energy photons generated.
factor in most human malignancies. The following sec- Modern linear accelerator energies span from the kilo-
tion reviews both the physical and biologic bases of radi- volt (kVp) to megavolt (MV) range. Outside of superfi-
ation therapy. Advances in both biologic modifiers and cial treatment of such lesions, EBRT is almost exclusively
new technologies to deliver radiation therapy that may delivered in the 4 to 18 MV range. For example, most
increase tumor cell killing while limiting normal tissue prostate cancer patients are treated with energies ranging
complications are also discussed. Specific examples of from 6 to 18 MV in order to spare superficial tissues and
particular interest for urologic oncology are highlighted. maximize the dose to the prostate.
The photons decelerate exponentially as they interact
with matter. The distance they travel through tissue is
THE PHYSICS OF RADIATION THERAPY
proportional to their initial energy (see above). Thus,
AND DELIVERY
higher energy beams are able to penetrate tissues deeper
External Beam Radiation Therapy
and result in fewer interactions at the skin surface. As the
EBRT is used to treat many tumor types, including head photons interact with matter, either superficially or deep
and neck, gynecologic, thoracic, and genitourinary within tissues, charged particles, such as electrons, are

39
40 Part I Principles of Urologic Oncology

Other types of particles have been employed as thera-


peutic modalities, including neutrons4 and protons.5
Neutron beams are similar to photon beams in that their
energy decreases exponentially in tissue. While neutron
beams have been employed to treat a variety of tumor
sites, including prostate and brain, they are seldom used
clinically, as multiple randomized studies have failed to
demonstrate a clear benefit when compared to photon
therapy in terms of tumor control and normal tissue tox-
icity. Proton beams are generated by a cyclotron. These
heavy charged particles are unique; they deposit their
dose near the end of their range. This phenomenon,
known as a Bragg peak, can be manipulated to deliver a
high dose to a small tumor deep within the body while
minimizing high doses to more superficial tissues.5
While there has been significant interest in expanding
the therapeutic applications of proton therapy, the sub-
stantial cost of a cyclotron (proton production machine)
along with limited clinical outcome data has resulted
in very few of them being constructed around the
world. As the technology to build cyclotrons becomes
cheaper to implement, more are expected to be con-
Figure 3-1 Example of a modern linear accelerator used to
structed. Theoretic and preliminary data suggest that
deliver photons or electrons in the clinical setting. proton therapy may be appropriate for prostate cancer
treatment.
set in motion that results in ionization and excitation of
other atoms. The energy absorbed in tissue by the sec-
Brachytherapy
ondary charged particles represents the dose delivered.
The accepted unit of dose is the gray (Gy), which is In contrast to EBRT, brachytherapy delivers dose
defined as the absorption of 1 J/kg. In radiation therapy through the placement of radioactive sources that remain
clinical outcome papers, the terms centigray (cGy) or in place either temporarily (minutes to days) or perma-
radians (rad) are commonly used with 1 cGy (or rad) nently. Initially, naturally occurring isotopes, such as
representing 0.01 Gy. Prior to 3D treatment planning radium, were used in brachytherapy. Newer artificially
techniques, dose was prescribed to a point, for example, produced isotopes have replaced radium due to their
in the middle of a patient or in the middle of a tumor. wider availability and improved safety profiles. The pre-
With modern 3D treatment planning techniques, dose scribed dose in brachytherapy is normally defined based
can also be prescribed to a volume of interest, for exam- on a limited number of 2D tissue points. Increasingly, 3D
ple, the tumor and areas of tumor spread. Current 3D dose prescriptions are becoming more common for
planning terminology of dose refers to the minimum brachytherapy.
dose absorbed by the volume of the target. The interac- Intracavitary brachytherapy involves placement of the
tion of the charged particles with tissue results in pro- sources within a body cavity. For example, most interme-
duction of free radicals along with direct damage to diate- to advanced-stage cervical cancers are treated, in
DNA. The biologic effects of absorbed dose are dis- part, with a Fletcher-Suit applicator. A hollow tube (tan-
cussed in detail later. dem) is inserted into the uterus, and two other hollow
Linear accelerators can also be configured to produce tubes (colpostats) are placed within the vagina against the
electrons by removing the tungsten target and guiding lateral fornices. Dose is delivered through insertion of
them through the accelerator toward the patient. Unlike radioactive sources into the hollow tubes that are secured
the photons that comprise x-rays, an electron is a charged in place. The sources (cesium 137) remain in place for 3
particle that travels a known range in tissue. By selecting to 4 days (low-dose rate [LDR] brachytherapy) depend-
the initial energy of the electrons, one can calculate the ing on the clinical scenario and choice of the physician.
depth of tissue that will be irradiated. Tissues beyond Intracavitary brachytherapy has also been used to treat
that range receive little irradiation. Since electrons are tumors of the head and neck, biliary tree, and bronchi. By
charged particles, they interact directly with matter in placing the hollow cylinders or catheters before loading
tissue by depositing dose and causing damage to the the radioactive sources (after loading), radiation exposure
tumor cell. to the staff is minimized.
Chapter 3 Principles and Applications of Radiation Oncology 41

conform the dose to the target depends on many factors,


including target location, external contour of the patient,
tissue density, beam energies availability, and the EBRT
hardware availability.10–12 Current 3D treatment plan-
ning and 3D treatment (3D-CRT) rely almost exclusively
on computed tomography (CT)-based imaging to gener-
ate a customized plan for each patient. The following
sections outline treatment planning and plan generation
using a prostate patient as a model assuming that 3D
treatment planning is available.

Simulation and Patient Immobilization


Most prostate cancer patients are treated with fractionated
(administered in multiple daily treatments) EBRT for 6 to
8 weeks. Patients normally lie supine on the treatment
table as shown in Figure 3-1. In order to deliver dose to
the prostate each day and minimize dose to the surround-
ing normal tissue (such as the rectum), the patient’s posi-
tion on the table must be reproduced during each
treatment. In addition, patient motion must be minimized
Figure 3-2 Initial pelvic film immediately following during the treatment that normally lasts 5 to 15 minutes.
implantation of permanent iodine125 prostate brachytherapy
Prior to initial treatment, a simulation of the actual
seeds. Due to subsequent swelling of the prostate, the seeds
will move and rotate during the next 60 days when they are
treatment technique is performed to determine the ideal
most radioactive. patient positioning. In order to ensure the reproducibil-
ity of the patient’s position, immobilization devices are
constructed out of foam cradles that will be used on all
Interstitial brachytherapy delivers dose directly subsequent patient treatment days. Through the use of
within a tumor or surgical bed. Hollow flexible these immobilization devices and newer imagining tech-
catheters are initially surgically inserted into the tumor niques, such as video-assisted setup, radiation therapists
or site of tumor resection. After 5 to 7 days of healing, are able to reproduce patients’ positions to within mil-
radioactive needles are inserted into the catheters to limeters daily.
deliver dose. Prostate brachytherapy involves the A CT simulator is a specialized scanner used to
placement of permanent radioactive seeds (iodine 125) directly acquire CT data while the patient is immobi-
within the prostate (Figure 3-2). These seeds deliver a lized in the desired treatment position. The CT data
dose over the course of many months to the prostate acquired from imaging guides radiation therapy plan-
and surrounding tissue. ning by providing geometric information on external
As discussed above, traditional LDR intracavitary patient contour and tumor size, shape, and location rel-
brachytherapy is delivered over 3 to 4 days as an inpatient ative to adjacent critical structures.13 Following the
procedure or sources with a short half-life left in perma- acquisition of the CT data, the physician defines three
nently as is the case with prostate brachytherapy. volumes to be used in the treatment planning process.
Recently, high-dose rate (HDR) techniques are increas- The gross tumor volume (GTV) represents the tumor
ing in popularity. High-activity iridium sources deliver visible on the CT simulation data. The clinical target
dose rapidly, permitting patients to be treated as an out- volume (CTV) is defined as the GTV and the draining
patient requiring only minimal anesthesia. Clinical stud- lymphatic and other tissues that may contain micro-
ies employing HDR have demonstrated efficacy in head scopic disease. The planning target volume (PTV) is
and neck, cervix, and prostate.6–8 created based on expansion of the GTV and CTV in
order to compensate for patient setup uncertainty, such
as patient and organ motion. For a typical prostate
TREATMENT PLANNING AND DELIVERY
patient plan, the PTV expansion typically ranges from
As previously described, the dose to be delivered during 0.6 to 1 cm.14 Normal tissues are also defined in order to
EBRT is prescribed to a volume in 3D conformal radia- design a plan that will minimize the dose to those
tion therapy (3D-CRT). The aim of the prescription is to organs, such as the rectum. The treatment planning is
uniformly irradiate the volume (target) while minimizing based on the volumes entered by the physician following
the dose to surrounding normal tissues.9 The ability to the simulation.
42 Part I Principles of Urologic Oncology

Treatment Planning Treatment Delivery and Verification


Treatment planning software permits physicists and Linear accelerators deliver the beam of photons or elec-
physicians to generate a dose distribution superimposed trons through a gantry that rotates 360 degrees around
on the CT images and volumes that have been designed. the patient (Figure 3-1). The rotation of the gantry
Although the specifics of the treatment planning software around the table permits the beam to enter the patient’s
may vary widely based on the technology available at body from any angle. The beam is shaped at each gantry
institutions, certain variables are universally required for angle by either a Cerrobend block cut by hand or a multi-
treatment planning. These variables include beam leaf collimator (MLC). Cerrobend attenuates (blocks)
energy, type of beam (photon or electron), number beam nearly all of the beam energy.3 These custom blocks are
angles, relative beam weights, and beam-modifying designed for each field during the treatment planning
devices. Superficial tumors can be treated with either stage, cut to the proper shape by a technician, and placed
low-energy photon beams (100 to 250 kVp) or electron manually in the head (Figure 3-1) of the gantry each day
beams. Tumors deep within the body, such as the of treatment. More recently, the MLC was developed to
prostate, are treated with energies ranging from 6 to 18 permit dynamic block shaping under computer control.
MV and from 4 to 9 beam angles. For example, most A typical MLC consists of 80 to 120 leaves that are fixed
prostate plans employ 4 to 5 beam angles with 6 MV to the gantry head and powered by small electric motors.
photons. The beam modifying devices include cus- The leaves are computer controlled and can be moved
tomized shielding blocks that alter the quality, intensity, either before (static) or during treatment (IMRT, dis-
and shape of the beam. These are further discussed in the cussed later). The leaves attenuate the beam similar to
section on intensity modulated radiation therapy the solid Cerrobend block permitting one to shape the
(IMRT). beam to virtually any shape.15
Dose volume histograms (DVH) provide a quantita- Quality assurance is usually performed at least on a
tive evaluation of treatment plans. The DVH represents weekly basis through the use of verification (x-ray) films.
the volume of a particular organ irradiated as a function These films are checked by the physician to ensure that
of dose (Figure 3-3). These data coupled with known tox- the patient is positioned properly on the table and the
icity research aid the physician in selecting the proper field shape (either through a Cerrobend block or MLC)
treatment plan. For example, based on both retrospective is correct. The treatment verification films are compared
and randomized data, most physicians planning a against those taken during the initial simulation process
prostate treatment attempt to limit the volume of rectum and the patients (or blocks) are moved to approximate the
receiving >70 Gy to <25% of the volume. original planning position.

Intensity Modulated Radiation Therapy


Recent advances in computer hardware and software
have dramatically changed the way in which 3D treat-
ment plans are constructed and EBRT is delivered to the
patient. With traditional 3D treatment planning, EBRT
is delivered with beams of uniform intensity and with a
static beam shape (though the use of a block or MLC).
(IMRT, as the name implies, permits nonuniform inten-
sity beams to be delivered to the patient. By allowing the
MLC to move during treatment, the shape of the beam
at each gantry angle can be altered. By modulating the
intensity and shape of the beam during treatment, high
doses of radiation can be delivered to the PTV while
minimizing dose to normal tissues.16 After the physician
defines the GTV, CTV, and PTV and determines the
normal tissue structures to avoid, the computer software
employs sophisticated algorithms to compute the inten-
Figure 3-3 DVH from a head and neck cancer case planned sity and shape of the beam for each gantry angle. This
with IMRT. The PTV contains the primary tumor and draining process is known as inverse planning.63,64 While no ran-
lymphatics plus a margin for microscopic disease and setup domized studies yet exist demonstrating the superiority
uncertainty. The IMRT permits the brain stem and spinal cord of this approach in terms of tumor control or normal tis-
to receive a very low dose of RT compared to the PTV. sue complications compared to conventional treatment
Chapter 3 Principles and Applications of Radiation Oncology 43

believed that accumulation of DNA double-stand breaks


(DSBs) are the main requirement for cell death to occur.
Following DNA damage, both tumor cells and normal
tissues respond by attempting to repair the damage, alter
the cell cycle, induce gene transcription, or promote cell
death.18,19 Cell death occurs through either a postmitotic
(necrotic) or apoptotic death that are discussed later. The
formation of ROS and the cellular response to DNA
damage are also influenced by cell membrane receptors
that initiate second messenger cascades that can mediate
cell death.20–23 The following sections describe cellular
response to IR and provide a brief overview of the bio-
logic events that determine whether a cell will live or die
following IR.

IONIZING RADIATION Interacts with Cellular


Targets and Generates Reactive Oxygen Species
IR interacts with matter either directly or indirectly
through production of ROS. IR causes ionization of cel-
lular H2O that produces free hydroxyl radicals. These
free radicals quickly interact with cellular targets. The
damage to the cellular targets, such as cell membranes,
Figure 3-4 Comparison of a conventional plan (top) with an
proteins, and DNA, is deposited randomly and is a func-
IMRT plan in the pelvis used to treat an anal cancer patient.
The arrows demonstrate that the IMRT plan conforms to the
tion of dose. Data demonstrate that damage by the short-
target delivering 100% of the prescribed dose, while the 2D lived free radicals represents a majority of the clinically
plan delivers 100% of the dose to a large volume of normal relevant damage produced by conventional photon ther-
tissue, including femoral heads and genitalia. apy and electron therapy.24 Experimental evidence
demonstrates that double-strand breaks (DSBs) in the
techniques, multiple prospective studies demonstrate at sugar phosphate backbone of DNA result in most of the
least equal tumor control compared to historical controls cytotoxicity to both tumor cells and normal tissues.
while greatly reducing normal tissue toxicity. For exam- While many factors contribute to the extent of DNA
ple, prostate cancer IMRT minimizes the volume of the damage following IR, cellular oxygen is critical as
bladder and rectum irradiated. This has permitted the hypoxic cells are up to 3-fold more resistant to killing by
dose delivered to the prostate to be increased from 66 to IR compared to well-oxygenated cells.25 The presence of
70 Gy to over 81 Gy at some institutions.17 An example cellular oxygen prolongs the lifetime of free radicals and
of a pelvic IMRT treatment plan from the University of appears to fix free radical damage in the DNA. The dif-
Chicago compared to a conventional 3D plan (Figure 3-4) ference between tumor cell killing in the presence and
demonstrates that the patient treated with the IMRT absence of oxygen is termed the oxygen enhancement
plan will receive less dose to the normal tissue that sur- ratio (OER). This is clinically relevant as tumor growth
rounds the target. may outpace its ability to generate new tumor vasculature
(angiogenesis). This growth results in areas of hypoxia
and necrosis within the tumor.26 The hypoxic regions,
IONIZING RADIATION AND ITS INTERACTION
while poorly vascularized, may remain resistant to IR
WITH TUMOR CELLS AND NORMAL TISSUES
during fractionated radiation therapy. As the tumor
While the therapeutic applications of x-rays were quickly changes composition during fractionated RT, reoxygena-
realized since their introduction, the biologic basis by tion of the once poorly hypoxic regions may occur dur-
which IR kills both normal tissues and tumor cells is a ing some fractions of therapy, again providing a rationale
subject of intense investigation. It is now clear that IR for fractionated treatment. In contrast to conventional
kills both tumor and normal tissues through several therapy, heavy charged particle radiation, such as proton
molecular mechanisms. The dose of IR absorbed in tis- therapy, kills almost exclusively through direct interac-
sues interacts with matter either directly or indirectly tion with cellular targets and is thus not affected by oxy-
through the hydrolysis of water and the formation of gen levels.27 Thus, through both direct and indirect
reactive oxygen species (ROS). While both single- and interactions with cellular structures, conventional EBRT
double-stranded DNA breaks occur following IR, it is damages cellular targets resulting in eventual cell death.
44 Part I Principles of Urologic Oncology

The accumulated damage to DNA and other struc- Attempts to enhance the therapeutic ratio have come in
tures eventually results in the inability of cells to divide the form of altered fractionation schemes, addition of
(clonogenic death). The cell may undergo multiple cell radiation sensitizing agents, and use of radioprotectors.
divisions before clonogenic death occurs. The activation These are discussed later.
of programmed cell death (apoptosis) also plays a signifi-
cant role in certain normal and tumor tissues and may
Enhancement of the Tumoricidal Effects of IR with
occur within hours of exposure of cells to IR. These find-
Radiation Sensitizers
ings demonstrate that tumor cells with a low mitotic index
(slow proliferation), such as some prostate cancers, may Clinical trials demonstrate that local control following
take many months for complete histologic regression.28 radiation therapy is significantly worse than in patents
with normal blood oxygen carrying capacity.35 Multiple
studies have demonstrated improvements in outcome of
Accumulation of DNA Damage Following IR
patients treated with hyperbaric oxygen.36 In addition,
Induces A Cellular Repair Response
compounds that act as hypoxic cell sensitizers, such as
The accumulation of damage, if not repaired, will result metronidazole and misonidazole, have been developed
in the clonogenic or apoptotic death of cells. Cells that and employed clinically. Despite promising studies in
acquire damage not beyond repair, termed sublethal vitro, few clinical studies have demonstrated improve-
damage repair (SLDR), may repair the damage to the ment in outcome at the expense of significantly increased
DNA between fractions of radiation. It is believed that toxicity.37 While laboratory studies have demonstrated
many tumors have a poor repair response compared to significant enhancement of the antitumor effects of
the surrounding normal tissues. This concept is crucial in hyperbaric oxygen or hypoxic cell sensitizers, the clinical
the attempt to permit recovery of normal tissues between application of these techniques has not led to significant
daily fractions while continuing to kill tumor cells. By therapeutic gain.
separating the doses of radiation, each day normal tissues Clinically, the most commonly employed agents to
can induce a repair response prior to the second dose of enhance radiation-mediated tumor cell killing are stan-
radiation. Tumor cells may be unable to repair their dam- dard chemotherapeutics, such as 5-fluorouracil (5-FU),
age thus it will accumulate until clonogenic or apoptotic cisplatin (CDDP), and paclitaxel. Combined treatment
death occurs. This concept has been demonstrated in with radiation therapy and chemotherapy has been
both in vitro and in vivo models of cell killing.29,30 demonstrated in randomized trials of head and neck,
The local environment of the cell also plays a role in gynecologic, and lung cancers to be superior to treatment
its capacity to repair its damage or its decision to die fol- with radiation alone. While the mechanisms of radiosen-
lowing exposure to IR. Damage to cellular structures that sitization vary, the combined use of these agents with IR
may be potentially lethal (PLDR) while a cell is exposed has led to improvements in local control of disease and
to certain growth factors, such as oxygen concentration, improved survival in many cancer sites.
and cell–cell contact may be repaired if postirradiation
conditions are altered to enhance cell survival.31 Based on
Protection of Normal Tissue Through the Use
both in vitro and in vivo studies, Weichselbaum and oth-
of Radioprotector Agents
ers suggest that PLDR significantly contributes to radia-
tion therapy failure.32–34 The understanding of these While the addition of chemotherapeutic agents to radia-
basic mechanisms has led to the development of agents tion therapy has enhanced the tumoricidal effects of IR,
and techniques to enhance the tumoricidal effects of IR. this benefit is usually accompanied by an increase in nor-
mal tissue toxicity. The balance between tumor cell
killing and normal tissue toxicity defines the therapeutic
Therapeutic Ratio of Radiation Therapy
ratio and ultimately limits the probability of cure. In
The probability of tumor cure is a function of many vari- order to increase the therapeutic ratio and limit toxicity
ables, including tumor type, size, radiation dose, and to normal tissues, agents that protect tissues from radia-
fraction size. For example, microscopic (subclinical) dis- tion (radioprotectors) have been employed clinically.
ease from an epithelial-derived tumor is usually curable Originally developed in the 1950s by the department of
with as little as 45 to 50 Gy. However, this dose would be defense, the radioprotector amifostine (Ethyol) acts as a
insufficient to cure microscopic disease derived from scavenger of free radicals. Following intravenous deliv-
high-grade astrocytomas, a large epithelial tumor, or T2 ery, the compound amifostine penetrates into normal tis-
prostate cancer. Most doses used in clinical practice have sues within minutes. However it is absorbed more slowly
been empirically determined. The therapeutic ratio is in the tumor tissue providing a rationale for preferential
defined as the probability of obtaining a tumor cure protection of normal tissues. Randomized studies in
divided by the chance of a normal tissue complication. head and neck and pelvic malignancies have shown some
Chapter 3 Principles and Applications of Radiation Oncology 45

protection from expected radiation sequelae while not DNA damage induces cell cycle arrest. The cell sig-
decreasing local control of disease.38 The use of amifostine naling pathways that monitor DNA damage and initiate
use, however, remains fairly limited due to the expense cell cycle arrest are referred to as checkpoint controls.
of the compound, limited randomized clinical data, For example, the commonly mutated tumor suppressor
and often severe acute side effects including nausea and gene and transcription factor p53 promotes cell cycle
hypotension. arrest at the G1 checkpoint following DNA damage
through transcription of p21 that inhibits progression to
S-phase. The exact phase of the arrest is dependent on
Enhancement of the therapeutic ratio through
the genetic mutations present in the tumor cell. Arrest of
gene therapy
the cell cycle in damaged cells represents an evolutionar-
While attempts to employ gene therapy with cancer as a ily conserved survival mechanism to ensure the fidelity
single modality have met with limited clinical success, and transmission of genetic information. By preventing
laboratory and clinical data demonstrate that combining progression through the cycle, the damage induced by IR
gene therapy with IR may improve tumor curability. can be repaired before the cell continues to grow and
Unlike combining IR with standard chemotherapy divide. Failure to repair the damage leads to genetic
agents that often have overlapping toxicity, gene therapy instability and cell death.44
can be tailored to target different and partially nonover- As previously noted, while IR induces both single-
lapping mechanisms of tumor cell killing. Current and double-strand DNA breaks, DSBs are responsible
research in gene therapy aims to: (1) replace mutated for most tumoricidal effects of IR. DNA damage is
genes (p53) that may sensitize cells to IR; (2) deliver repaired in mammalian cells through two distinct
genes encoding pro-drug converting enzymes into molecular pathways: homologous recombination and
tumor cells, allowing toxic agents to be generated in the nonhomologous recombination. Homologous recombi-
tumor bed, thereby decreasing the risks of systemic tox- nation involves various repair proteins, such as rad51,
icity; (3) construct genetically engineered viruses with that identify homologous DNA to be used as a template
therapeutic genes under the control of radiation to direct error-free repair of the genetic information.45
inducible promoters; and (4) enhance replication com- However, the predominant mechanism of repair in
petent viruses that proliferate preferentially in tumor human cells is nonhomologous recombination. During
cells following IR. While current technology limits the this process, DSBs are identified by protein complexes
efficiency of gene transfer to tumor cells, radiation may and the ends joined by ligases (xrcc4).46 Due to the sim-
enhance the “bystander effect” of specific gene therapy ple end-joining and lack of a template for repair, non-
designs that employ a diffusible product.39–42 Clinical homologous recombination results in mutant
studies are currently underway employing a wide variety chromosomes. The protein sensors of DSBs that initi-
of these techniques. ate the repair cascade are a focus of intense investiga-
tion since inhibition of those sensors would sensitize
tumor cells to IR.
DNA Damage, the Cell Cycle, and Repair
Tumors represent a heterogeneous population of grow-
Ionization Radiation-Induced Cell Death:
ing cancer cells that are distributed unevenly throughout
Apoptosis and Necrosis
the cell cycle with most in the resting or G1 phase. As
previously discussed, damage to the DNA represents the One major biologic determinant of radiation therapy fail-
critical target for IR-induced cell death. In vitro and in ures is tumor radioresistance.47–49 As previously dis-
vivo data demonstrate that the extent of DNA damage cussed, exposure of mammalian cells to IR results in a
and tumor cell death is, in part, dependent on the phase loss of cellular reproductive capability (mitotic death) by
of the cell cycle. While exceptions exist, the most inducing DNA DSBs and lethal chromosomal aberra-
sensitive phases of the cell cycle are G2/M. Cells are least tions.50 Morphologic characteristics of IR-induced
sensitive to IR in late S-phases.43 Identification of differ- mitotic death include multinucleated giant cells, cell–cell
ential sensitivity based on cell cycle provides further fusions, and loss of membrane integrity that is also a
rationale for fractionated treatment regimens. Since characteristic of necrotic cell death. Daughter cells with
tumor cells are an asynchronous population, after each limited divisional potential can arise from lethally irradi-
fraction those in the most sensitive phases will be killed ated mother cells to form abortive colonies. Death dur-
while the resistant phases will progress to other phases of ing cell division due to lethal mutations or damaged
the cycle. This process is termed reassortment. Thus, chromosomes following IR is a well-studied mechanism
with standard EBRT with photons, fractionation permits of tumor cell killing.51
cells to be exposed to radiation while the cells are in their In contrast to necrotic death, apoptosis induced by IR
most sensitive phases. results in activation of a genetic program initiated by
46 Part I Principles of Urologic Oncology

cytoplasmic or nuclear events culminating in cytoplasmic randomized studies demonstrate improvements in local
blebbing, chromatin condensation, and DNA fragmenta- control of disease, organ preservation, and overall sur-
tion.52–54 The induction of apoptosis may occur immedi- vival when comparing RT alone with combined modality
ately after irradiation (interphase death), after arrest in treatment in diseases that include the head and neck,
the G2 phase,55 or following one or more cell divi- bladder, cervix, esophagus, and lung. Postoperative
sions.56,57 Apoptosis may represent an important mecha- chemoradiotherapy is also employed for gastric, rectal,
nism of death in some cell types following IR, such as and head and neck tumors. Concomitant RT appears to
lymphomas.56 It has been proposed that the direct effects produce better local control in some disease sites while
of x-rays on the nucleus and DSBs initiate a cascade of producing significant more acute toxicity. Thus, current
signaling events, such as p53 induction, that culminates clinical trials seek to identify more active chemother-
in the activation of proteases (caspases) that execute the apy agents with fewer overlapping toxicities and to deter-
cell death program.55,56 While the signal to undergo mine the optimal timing and delivery of chemotherapy
apoptosis following IR is generally considered to origi- with RT.
nate in the nucleus,55,56,58,59 studies demonstrated that
the production of the lipid second messenger ceramide
Palliative Radiation Therapy
from sphingomyelin hydrolysis immediately following IR
for Metastatic Disease
contributes to the apoptotic response.60–62 Current
research efforts aim to enhance the cytotoxicity of IR Painful metastatic disease is effectively treated with a
through development of small molecules that exploit the combination of pain medication and RT. Osseous metas-
apoptotic program. tases are commonly observed in advanced breast, lung,
and prostate patients. Pain relief from an abbreviated
course of RT can reduce or eliminate pain in 70% to
THE CLINICAL PRACTICE OF RADIATION
80% of cases.67 A recently completed randomized study
ONCOLOGY
by the Radiation Therapy Oncology Group (RTOG)
Definitive and Adjuvant Radiation Therapy
compared a large single fraction of 8 Gy to the more con-
Therapeutic radiation (RT), delivered via external beam ventionally used 10 fractions (3 Gy) to 30 Gy in breast
or brachytherapy, by itself can cure many human malig- and prostate patients.68 Equivalent outcome in terms of
nancies, including head and neck, cervix, lymphoma, pain relief and complications were found suggesting that
prostate, and pure seminomas. RT alone demonstrates shorter courses of palliative RT may be as effective in the
equivalent local control compared with surgery in head patient with metastatic disease. Longer follow-up is
and neck malignancies while better preserving normal needed, however, to determine if long-term complica-
organ function.63,64 Similar results have also been seen in tions and duration of pain relief will remain equivalent.
early cervical cancer trials.65 To date, no completed ran- Patients who develop multiple cerebral metastases
domized study employing modern surgical and radiation from lung, bladder, breast, and other tumors suffer neu-
techniques for prostate cancer exists. rologic sequelae from both the expanding tumor mass
RT is commonly employed as part of a combined itself and subsequent edema formation. Combining ini-
modality approach in cancer therapy. It is routinely tial steroid treatment with fractionated RT to the entire
used as an adjuvant to surgical excision to sterilize brain (typically 30 and 3 Gy fractions) improves neuro-
potential microscopic disease in lymph nodes or from logic function in over 50% of patients. Patients with a
surgical margins. Preoperative RT is used for large single brain metastasis benefit from combined surgical
unresectable tumors, such as sarcomas or rectal cancer, resection and postoperative RT.
in an attempt to permit eventual surgical excision or Spinal cord compression is also effectively treated
organ conservation.66 More commonly, postoperative with palliative RT. Again, the neurologic compromise is
RT is employed following definitive surgical findings often treated with a combined modality approach. High-
of margin status, tumor pathology, and stage of disease. dose steroids reduce the surrounding edema and provide
The decision to employ postoperative RT can then be often rapid neurologic improvement. A recent random-
made and treatment type and dose tailored to the spe- ized study demonstrates that surgical excision and stabi-
cific case. Common postoperative RT applications lization of the vertebral bodies involved with tumor
include tumors of the head and neck, lung, breast, gen- followed by RT provide superior pain relief and improve-
itourinary, gastrointestinal tract, and central nervous ment of neurologic function compared with RT alone.69
system. In patients deemed surgically inoperable, RT is normally
As previously discussed, chemotherapy may be com- delivered in 3 Gy fractions to a total dose of 30 Gy.
bined with RT in order to increase tumor cell killing. The dose of 30 Gy provides good tumor control while
Commonly, chemotherapy is given prior to RT (neoad- offering almost no additional risk that the radiation itself
juvant) or simultaneously (concomitant) with RT. Recent will damage the spinal cord.
Chapter 3 Principles and Applications of Radiation Oncology 47

tum or lung, as the volume of tissue irradiated increases


Acute and Chronic Radiation Sequelae
so does the probability of complications. An example of
As previously stated, the therapeutic index of radiation this volume and complication relationship is seen in the
therapy is defined as the probability of tumor cure recently completed randomized 3D conformal dose-
divided by the probability of severe toxicity. The nor- escalation study for prostate cancer. In order to achieve
mal toxicity of radiation therapy is a function of the vol- better local control of low and intermediate risk
ume and type of tissue exposed to IR, dose fraction, and prostate cancer with EBRT, Pollack et al. randomized
total dose. The currently employed radiation sensitiz- patients to 78 Gy versus 70 Gy in 2 Gy fractions.69
ers, standard chemotherapy, also increase acute and With 100 months of follow-up, the 78-Gy group
chronic toxicity of RT. Acute radiation sequelae occur showed significantly better PSA-free survival in the
through damage to actively dividing normal tissues.70 intermediate risk group demonstrating that increased
Commonly seen acute toxicities during or shortly fol- dose leads to more prostate cancer cures in this popu-
lowing fractionated radiation therapy include skin lation. However, grade 2 (TABLE 3-1) or greater rec-
desquamation, diarrhea, and mucositis. Chronic seque- tal complications were also increased. Upon further
lae normally develop months to years after radiation analysis a dose/volume relationship can be seen in
therapy and result from fibrosis and subsequent scarring terms of complications. If the percentage of rectum
of normal tissues. treated to >70 Gy was <25%, grade 2 or higher com-
While acute toxicity can normally be managed dur- plications were <15%. However, if the volume
ing the course of treatment, chronic toxicity remains exceeded 25%, the probability of complication rose to
the limiting factor when determining the maximum 45%. These data from randomized dose escalation
dose to deliver in the effort to cure the tumor. The series for prostate, as well as other disease sites, guide
probability of causing a chronic toxicity in 5% of the the radiation oncologist when balancing the desire to
treated population at 5 years is termed the TD5/5. deliver higher dose for increased probability of cure
Injury to a portion of certain organs (i.e., spinal cord) with the chance of chronic complications.
markedly impairs the function of the entire organ. For
these organs radiation oncologists have developed
SUMMARY
empiric dose constraints to limit observed toxicity to
almost 0%. For example, a dose of 45 to 50 Gy maxi- Radiation therapy has been used to treat cancer patients
mum to any point on the spinal cord is accepted as the since 1896. The development of modern linear accelera-
standard of care. For other structures, such as the rec- tors, sophisticated computer planning software, and an

Table 3-1 RTOG Toxicity Scoring System


Organ 0 Grade 1 Grade 2 Grade 3 Grade 4

Large None Mild diarrhea Moderate diarrhea Surgery is required Fistula or perforation
intestine/ and cramping (bowel movement due to bleeding or develops
rectum with bowel > 5 times/day) or obstruction of
movement < 5 excessive rectal bowel
times/day mucus or mild
but intermediate
bleeding

Spinal cord None Mild Lhermitte’s Severe Lhermitte’s Neurologic findings Paraplegia
syndrome syndrome on examination
that can be
accounted for by
damage at the level
of spinal cord in
the radiation field

Bladder None Microscopic Increased frequency Severe frequency Bladder capacity


hematuria of urination with or dysuria < 100 cc, hemorrhagic
occasional Macroscopic cystitis, or necrosis
macroscopic hematuria
hematuria Bladder capacity
reduced to < 150 cc
48 Part I Principles of Urologic Oncology

increased understanding of the basic biology that under- conformal treatment planning for stage C prostate cancer.
lies the cytotoxicity of IR has allowed the field to cure Int J Radiat Oncol Biol Phys 1995; 33(5):1009–1017.
more human tumors, decrease the need for large and 13. Kuszyk BS, Ney DR, Fishman EK: The current state of
debilitating surgical procedures, and decrease the proba- the art in three-dimensional oncologic imaging: an
overview. Int J Radiat Oncol Biol Phys 1995;
bility of complications. Current clinical trials in most dis-
33(5):1029–1039.
ease sites demonstrate that combined modality therapy
14. Roeske JC, et al: Evaluation of changes in the size and
with chemotherapy and radiation remains superior to location of the prostate, seminal vesicles, bladder, and
radiation alone despite the increase in toxicity. Future tri- rectum during a course of external beam radiation
als with more novel chemotherapeutic agents aim to pro- therapy. Int J Radiat Oncol Biol Phys 1995;
vide similar radiation sensitization while decreasing the 33(5):1321–1329.
overlapping toxicities. More advanced molecular biology 15. Boyer AL, et al: Clinical dosimetry for implementation
techniques have enabled gene therapy to be combined of a multileaf collimator. Med Phys 1992;
with radiation in order to achieve a similar goal. We 19(5):1255–1261.
expect the advancements in both hardware, software, and 16. Ling CC, et al: Intensity-modulated radiation therapy.
biology will lead to further increases in the therapeutic In Devita V, Hellman S, Rosenberg SA (eds): Cancer:
Principles and Practice of Oncology. Philadelphia, PA,
ratio in the future.
Lippincott-Raven, 2001.
17. Zelefsky MJ, et al: High dose radiation delivered by
intensity modulated conformal radiotherapy improves the
REFERENCES outcome of localized prostate cancer. J Urol 2001;
166(3):876–881.
1. Roentgen W: On a new kind of rays [preliminary 18. Dizdaroglu M, Measurement of radiation-induced damage
communication]. Translation of a paper read before to DNA at the molecular level. Int J Radiat Biol 1992;
the Physikalische-medicinischen Gesellschaft of 61(2):175–183.
Wurzburg on December 28, 1895. Br J Radiol 1931; 19. Dizdaroglu M: Oxidative damage to DNA in mammalian
1(4):32–36. chromatin. Mutat Res 1992; 275(3–6):331–342.
2. Coutard H: Roentgentherapy of epitheliomas of the 20. Lin X, Fuks Z, Kolesnick R: Ceramide mediates
tonsillar region, hypopharynx and larynx from 1920 to radiation-induced death of endothelium. Crit Care Med
1926. Am J Roentgenol 1932; 1932(28):313–332. 2000; 28(Suppl 4):N87–N93.
3. Khan F: The Physics of Radiation Therapy, 2nd edition. 21. Billis W, Fuks Z, Kolesnick R: Signaling in and
Baltimore, MD, Williams and Wikins, 1994. regulation of ionizing radiation-induced apoptosis in
4. Laramore GE, et al: Fast neutron radiotherapy for locally endothelial cells. Recent Prog Horm Res 1998; 53:85–92
advanced prostate cancer: results of an RTOG (Discussion 93).
randomized study. Int J Radiat Oncol Biol Phys 1985; 22. Bodmer JL, Schneider P, Tschopp J: The molecular
11(9):1621–1627. architecture of the TNF superfamily. Trends Biochem Sci
5. Bonnett DE: Current developments in proton therapy: a 2002; 27(1):19–26.
review. Phys Med Biol 1993; 38(10):1371–1392. 23. Sartorius U, Schmitz I, Krammer PH: Molecular
6. Leung TW, et al: High dose rate brachytherapy for mechanisms of death-receptor-mediated apoptosis.
carcinoma of the oral tongue. Int J Radiat Oncol Biol Chembiochem 2001; 2(1):20–29.
Phys 1997; 39(5):1113–1120. 24. Ward JF: DNA damage produced by ionizing radiation in
7. Stitt JA, et al: High dose rate intracavitary brachytherapy mammalian cells: identities, mechanisms of formation, and
for carcinoma of the cervix: the Madison system. I. reparability. Prog Nucleic Acid Res Mol Biol 1988;
Clinical and radiobiological considerations. Int J Radiat 35:95–125.
Oncol Biol Phys 1992; 24(2):335–348. 25. Littbrand B, Revesz L: The effect of oxygen on cellular
8. Rodriguez RR, Demanes DJ, Altieri GA: High dose rate survival and recovery after radiation. Br J Radiol 1969;
brachytherapy in the treatment of prostate cancer. 42(504):914–924.
Hematol Oncol Clin North Am 1999; 13(3):503–523. 26. Thomlinson H, Gray L: Histological structure of some
9. Purdy JA, Wong JW, Harms WB, Emami B, Matthews human lung cancers and the possible implications for
JW: State of the art of high energy photon treatment radiotherapy. Br J Cancer 1955; 9:539–563.
planning. Front Radiat Ther Oncol 1987; 21:4–24. 27. Raju MR, et al: A heavy particle comparative study.
10. Graham MV, et al: Preliminary results of a prospective Part III: OER and RBE. Br J Radiol 1978;
trial using three-dimensional radiotherapy for lung cancer. 51(609):712–719.
Int J Radiat Oncol Biol Phys 1995; 33(5):993–1000. 28. Cox JD, Kline RW: Do prostatic biopsies 12 months or
11. Sibley GS, et al: The treatment of stage III nonsmall more after external irradiation for adenocarcinoma, Stage
cell lung cancer using high dose conformal radiotherapy. III, predict long-term survival? Int J Radiat Oncol Biol
Int J Radiat Oncol Biol Phys 1995; 33(5):1001–1007. Phys 1983; 9(3):299–303.
12. Yang FE, et al: The potential for normal tissue dose 29. Fowler JF: Review: total doses in fractionated
reduction with neoadjuvant hormonal therapy in radiotherapy—implications of new radiobiological data.
Chapter 3 Principles and Applications of Radiation Oncology 49

Int J Radiat Biol Relat Stud Phys Chem Med 1984; 48. Lowe SW, et al: p53 status and the efficacy of cancer
46(2):103–120. therapy in vivo. Science 1994; 266(5186):807–810.
30. Hall E: Radiobiology for the Radiologist, pp 17–160. 49. Lowe SW, et al: p53 is required for radiation-induced
Philadelphia, PA, JB Lippincott, 1988. apoptosis in mouse thymocytes [see comments]. Nature
31. Hahn GM, Little JB: Plateau-phase cultures of 1993; 362(6423):847–849.
mammalian cells: an in vitro model for human cancer. 50. Hall EJ: Radiobiology for the Radiologist. New York,
Curr Top Radiat Res Q 1972; 8(1):39–43. Harper & Row, 1988.
32. Weichselbaum RR, et al: Radiation-resistant and repair- 51. Radford IR: The level of induced DNA double-strand
proficient human tumor cells may be associated with breakage correlates with cell killing after X-irradiation. Int J
radiotherapy failure in head- and neck-cancer patients. Radiat Biol Relat Stud Phys Chem Med 1985; 48(1):45–54.
Proc Natl Acad Sci USA 1986; 83(8):2684–2688. 52. Jacobson MD, Burne JF, Raff MC: Programmed cell
33. Weichselbaum RR, Schmit A, Little JB: Cellular repair death and Bcl-2 protection in the absence of a nucleus.
factors influencing radiocurability of human malignant EMBO J 1994; 13(8):1899–1910.
tumours. Br J Cancer 1982; 45(1):10–16. 53. Raff MC, et al: Programmed cell death and the control of
34. Guichard M, et al: Potentially lethal damage repair as a cell survival. Philos Trans R Soc Lond B Biol Sci 1994;
possible determinant of human tumour radiosensitivity. 345(1313):265–268.
Radiother Oncol 1984; 1(3):263–269. 54. Weil M, et al: Constitutive expression of the machinery
35. Evans J, Bergsio P: The influence of anemia on the results for programmed cell death. J Cell Biol 1996;
of radiotherapy in carcinoma of the cervix. Radiology 133(5):1053–1059.
1965; 84:709–712. 55. Radford IR, Murphy TK: Radiation response of mouse
36. Dische S, et al: Carcinoma of the cervix–anaemia, lymphoid and myeloid cell lines. Part III. Different signals
radiotherapy and hyperbaric oxygen. Br J Radiol 1983; can lead to apoptosis and may influence sensitivity to
56(664):251–255. killing by DNA double-strand breakage. Int J Radiat Biol
37. Lee DJ, et al: Results of an RTOG phase III trial (RTOG 1994; 65(2):229–239.
85-27) comparing radiotherapy plus etanidazole with 56. Dewey WC, Ling CC, Meyn RE: Radiation-induced
radiotherapy alone for locally advanced head and neck apoptosis: relevance to radiotherapy. Int J Radiat Oncol
carcinomas. Int J Radiat Oncol Biol Phys 1995; Biol Phys 1995; 33(4):781–796.
32(3):567–576. 57. Martin SJ, Green DR: Apoptosis as a goal of cancer
38. Buntzel J, et al: Selective cytoprotection with amifostine therapy. Curr Opin Oncol 1994; 6(6):616–621.
in concurrent radiochemotherapy for head and neck 58. Szumiel I: Ionizing radiation-induced cell death.
cancer. Ann Oncol 1998; 9(5):505–509. Int J Radiat Biol 1994; 66(4):329–341.
39. Ram Z, et al: The effect of thymidine kinase 59. Sanchez Y, Elledge SJ: Stopped for repairs. Bioessays
transduction and ganciclovir therapy on tumor 1995; 17(6):545–548.
vasculature and growth of 9L gliomas in rats. 60. Kolesnick RN, Haimovitz-Friedman A, Fuks Z:
J Neurosurg 1994; 81(2):256–260. The sphingomyelin signal transduction pathway
40. Tanaka T, Yamasaki H, Mesnil M: Stimulation of mediates apoptosis for tumor necrosis factor, Fas, and
intercellular communication of poor-communicating cells ionizing radiation. Biochem Cell Biol 1994;
by gap-junction-competent cells enhances the HSV- 72(11–12):471–474.
TK/GCV bystander effect in vitro. Int J Cancer 2001; 61. Haimovitz-Friedman A, et al: Ionizing radiation acts on
91(4):538–542. cellular membranes to generate ceramide and initiate
41. Belyakov OV, et al: Direct evidence for a bystander effect apoptosis. J Exp Med 1994; 180(2):525–535.
of ionizing radiation in primary human fibroblasts. Br J 62. Verheij M, et al: Requirement for ceramide initiated
Cancer 2001; 84(5):674–679. SAPK/JNK signaling in stress-induced apoptosis. Nature
42. Andrade-Rozental AF, et al:, Gap junctions: the “kiss of 1996; 380:75–78.
death” and the “kiss of life.” Brain Res Brain Res Rev 63. Million RR: Carcinomas of the oral cavity and
2000; 32(1):308–315. oropharynx. Curative treatment with preservation of
43. Terasima T, Tolmach LJ: Variations in several responses of function. Front Radiat Ther Oncol 1993; 27:20–30.
HeLa cells to x-irradiation during the division cycle. 64. Mendenhall WM, et al: Radiotherapy for carcinoma of
Biophysics J 1963; 3:11–15. the supraglottic. Otolaryngol Clin North Am 1997;
44. Iliakis G: Cell cycle regulation in irradiated and 30(1):145–161.
nonirradiated cells. Semin Oncol 1997; 24(6):602–615. 65. Landoni F, et al: Randomised study of radical surgery
45. Kanaar R, Hoeijmakers JH, van Gent DC: Molecular versus radiotherapy for stage Ib-IIa cervical cancer. Lancet
mechanisms of DNA double strand break repair. Trends 1997; 350(9077):535–540.
Cell Biol 1998; 8(12):483–489. 66. Pahlman L, et al: Preoperative irradiation of primarily
46. Haber JE: The many interfaces of Mre11. Cell 1998; non-resectable adenocarcinoma of the rectum and
95(5):583–586. rectosigmoid. Acta Radiol Oncol 1985; 24(1):35–39.
47. Weichselbaum RR, et al: In vitro radiobiological 67. Price P, et al: Low dose single fraction radiotherapy in the
parameters of human sarcoma cell lines. Int J Radiat treatment of metastatic bone pain: a pilot study. Radiother
Oncol Biol Phys 1988; 15(4):937–942. Oncol 1988; 12(4):297–300.
50 Part I Principles of Urologic Oncology

68. Hartsell WF, et al: Phase III randomized trial of 8 Gy in 1 70. Stevens K: The stomach and intestines. In Moss W, Cox
fraction vs. 30 Gy in 10 fractions for palliation of painful JD (eds): Radiation Oncology, Rationale, Techniques,
bone metastases: preliminary results of RTOG 97-14. Int pp 362–408. St Louis, MO, CV Mosby, 1989.
J Radiat Oncol Biol Phys 2003; 57(2 Suppl):S124.
69. Pollack A, et al: Prostate cancer radiation dose response:
results of the M.D. Anderson phase III randomized trial.
Int J Radiat Oncol Biol Phys 2002; 53(5):1097–1105.
C H A P T E R

4 Principles of Chemotherapy
for Genitourinary Cancer
David I. Quinn, MBBS, PhD, FRACP,
Patrick J. Creaven, MD, PhD,
and Derek Raghavan, MD, PhD

Cytotoxic chemotherapy has been in use for the manage- [EGF]) and the impact of cytotoxic agents, with potential
ment of advanced cancer for more than a century,1 aris- for resulting synergistic or antagonistic effects.
ing from concepts developed by Lissauer, Ehrlich, and Normal tissues are composed predominantly of a
many others. The initial attempts at such treatment were static population of cells (which rarely undergo cell divi-
characterized by a lack of specificity, with a fine balance sion), an expanding population (which retain the ability
between the toxicity to the tumor and that experienced to grow, under stringent physiologic control mecha-
by the host. As reviewed in detail elsewhere,1 during the nisms) and a self-renewing population (for tissues that
past century, there has been some refinement in our turn over rapidly, such as bone marrow and gastrointesti-
application of chemotherapy to the treatment of cancer, nal mucosa). In this situation, balance is maintained
predicated on an improved understanding of the bio- between natural attrition and replacement.
chemical basis of its action and a clearer insight into the The static, or terminally differentiated, population
cellular and molecular mechanisms underlying normal usually includes cells that do not undergo cell division
and malignant growth. after fetal life, such as skeletal muscle and neuronal tis-
sue. The cells of an expanding population do not nor-
mally undergo continuous growth and division but may
TUMOR CELL BIOLOGY IN RELATION
respond to stress (such as injury) with a period of replace-
TO CHEMOTHERAPY
ment growth. For example, hepatocytes can respond to
The anticancer agents are a varied collection of drugs surgical resection of liver tissue by re-entering the cell
that act through a range of mechanisms predominantly cycle and replacing the lost tissue. Another example of
focused on interference with cell reproduction. The dif- the expanding population is the stem cells found in the
ferences between the growth characteristics of normal bone marrow; these normally rest in the G0 phase but can
and malignant tissues form the major basis of the effec- re-enter the cell cycle. The fact that they are predomi-
tive use of cytotoxic chemotherapy.2 Differences between nantly in G0 may protect them, in part, from the effects
the cellular transport characteristics of these agents may of cytotoxic agents.
also contribute to the difference in response to some By contrast, the self-renewing cell populations (e.g.,
cytotoxics. It is also apparent that there are important gastrointestinal tract, hair follicles, bone marrow) are in
differences between intracellular metabolic functions, a continuous proliferative state, with constant cell
such as the expression of glutathione, an intracellular turnover, and are thus most commonly injured by cyto-
scavenger that interacts with some alkylating agents and toxic chemotherapy; the static cell populations are the
the platinum complexes to inactivate them. More recent least vulnerable to the effects of chemotherapy.
data suggest that there may be subtle interactions Malignant growth is essentially uncontrolled, occur-
between the expression of growth controlling factors ring as a result of a breakdown in the mechanism(s) that
(such as the receptor for the epidermal growth factor turn off growth. The patterns that contribute to tumor

51
52 Part I Principles of Urologic Oncology

growth may include reduction in the duration of the cell unregulated cell proliferation. The classic negative regu-
cycle, a decrease in the rate of cell death, or an increase lator of cellular proliferation is transforming growth fac-
in recruitment of cells into the active cell cycle. In gen- tor beta (TGFβ). TGFβ acts through a number of
eral, it appears that malignant growth follows a mechanisms to inhibit cell cycle progression.14 It directly
Gompertzian pattern,2 in which a period of exponential induces increased production of cyclin-dependent kinase
growth is followed by a slowing of the growth rate.3,4 (CDK) inhibitors such as p15INK4B, p27Kip1, and
This may occur through the tumor outgrowing its vascu- p21Cip1.15–17 These proteins, in turn, act to block the
lar supply,5 due to the development of toxic breakdown activity of cyclin: CDK complexes responsible for pRb
products associated with cellular turnover, or through phosphorylation.18,19 TGFβ also suppresses the expres-
other subtle cell–cell interactions. In addition to unregu- sion of the c-myc gene, which has a physiologic role in
lated cell growth, malignancy is characterized by tissue increasing the rate of G1 to S phase transition through
invasion and metastasis, sustained angiogenesis, and eva- mechanisms that are still being elucidated.20,21 EGF,
sion of apoptosis or programmed cell death.6 which binds to a cell membrane associated receptor to
initiate a chain of signal transduction, acts to increase the
rate of cell cycle proliferation in part by decreasing the
Cellular Kinetics and Cell Cycle Control
quantity of p27Kip1 present.22 In cell line experiments,
The kinetics of tumor cell growth, both in vitro and in blockade of the epidermal growth factor receptor
vivo, has been the subject of considerable study,2 (EGFR) results in increased p27Kip1 and arrest of the can-
although our concepts regarding this topic remain quite cer cell in G1 phase as it is unable to transit into S
fluid. Surprisingly little information is available regard- phase.23 Furthermore, p53 regulates cell cycle progres-
ing the kinetics of human tumor growth,7 although a sion through synthesis of the CDK inhibitor, p21Cip1.16
greater body of information is available regarding the Hence, cellular transit through the G1/S transition may
growth of animal tumors.2 It is generally agreed that be influenced by a number of factors, including growth
tumor cells grow through an orderly sequence of steps: factor concentration and effect, receptor expression and
activity, cell cycle regulatory molecule expression, and pRb
1. The initial growth phase (G1) is characterized by action. For example, a more rapid cell cycle could arise
synthesis of RNA and protein, as well as DNA repair; from any or a combination of increased EGF concentra-
this is a period of variable length, and its duration will tion, EGFR receptor overexpression, loss of CDK
determine the length of the total cell cycle of the inhibitor expression, cyclin overexpression, or retinoblas-
individual cell. toma gene mutation. The net rate of cell proliferation is
2. This is followed by the synthetic (S) phase, in which a synthesis of a multitude of potential effectors on the
new DNA is synthesized. pRb pathway.
3. The cells progress through the G2 phase, in which Several oncogenes, such as ras, c-myc, and src have
the total DNA content is double that of the normal activity in the M phase of the cell cycle; in fact, they may
cell. actually interact in the process leading to activation of
4. The mitotic (M) phase sees the division of the the M-phase promoting factor, which in turn controls
chromosomes and separation into two offspring cells. entry into mitosis (the second major cell cycle control
5. After mitosis, the cells may spend a variable period of point at the G2/M phase transition).24 Variation in the
time in a resting state known as G0—the cells are out activity of these oncogenes, whether induced by amplifi-
of active cycle and appear not to be affected by cation, mutation, or exogenous regulators, can result in
chemotherapy to any major extent. altered transition through mitosis.24–26 Tumor progres-
sion may be a function of a series of events that involve
A detailed description of the molecular biology of cell progressive loss of control over entry into the S phase
cycle control is beyond the scope of this chapter, but the and loss of regulation of M phase; such progression is due
principles have been reviewed elsewhere.6,8,9 In brief, to genetic instability and is central to the evolution of
several candidate genes and growth factors appear to reg- malignancy.24 These changes may, in part, be contingent
ulate the various steps of the cell cycle. Entry into G1 on the loss of M phase checkpoint function, a mechanism
from S phase is regulated by a range of growth factors by which the cell cycle pauses transiently and allows
and interrelated molecular mechanisms culminating in checking of the accuracy of replication.27,28
retinoblastoma protein (pRb) phosphorylation. When The concept of the cell cycle is of great importance to
hypophosphorylated, pRb blocks proliferation by seques- our understanding of cytotoxic action. Most agents affect
tering and altering the function of E2F transcription fac- some aspect of the synthesis of DNA, RNA, or protein,
tors that control the expression of banks of genes acting at different points within the cell cycle. This may
essential for the progression from G1 to S phase.10–13 be very important when adding agents in a combination
Disruption of the pRb pathway liberates E2Fs and allows regimen, for example, the use of a spindle poison (such as
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 53

a vinca alkaloid) may hold up cells from entry into the G1 chapter but has recently been reviewed.41–43 The interac-
phase and thus reduce the impact of an agent that acts tion between cell proliferation and apoptosis determines
predominantly at that point of the cell cycle. In turn, inhi- the net growth of a tumor mass. These factors have been
bition of G1/S phase transition may invalidate the effect of evaluated in relation to cancer development and progres-
agents that act at or subsequent to this point, such as sion in a number of human cancers, including prostate
topoisomerase inhibitors.29 These potential effects are cancer (PC). Normal prostate epithelial cells have a very
limited by the fact that many agents act at multiple points low replicative rate that results from replacement of the
in the cell cycle. Inhibition of checkpoint function may cell every 500 days and no net increase in cell number.44
explain how tumor cells can be more vulnerable to the In “low grade” prostatic intraepithelial neoplasia (PIN),
effects of cytotoxic agents than are normal cells—thus the epithelial cells undergo proliferation in excess of dying so
vulnerability of cancer cells to agents that target the S that they have a doubling time of around 150 days. Upon
phase or M phase may occur because the malignant cells development of “high grade” PIN, there is an increase in
proceed unchecked through the cell cycle despite a series both replication and cell death so that there is no net
of errors, whereas the normal cells stop at finite check- increase in cells but the turnover time is around 56 days.
points while needed repairs occur. In addition, there is an Subsequent transition to invasive PC involves little
increasing trend toward designing drugs that target spe- change in proliferation but rather a reduction in cell
cific parts of the cell cycle machinery. Examples of mitotic death and a mean doubling time of around 500 days.
phase regulators include vinca alkaloids, taxanes, and Metastatic PC cells have both increased proliferation and
epothilones30,31 whereas flavopiridol and UCN-01, which cell death rates with a mean doubling time of around 33
are currently under development, inhibit cyclin-dependent days for lymph node and 54 days for bone metastases in
kinases active in G1/S phase transition.32–35 hormone sensitive cases.44 In metastatic androgen-
Cell cycle characteristics can be measured in several independent PC, there is no further increase in cellular
ways, including the use of labeling of mitoses,2 flow proliferation and an increase in cell death. The reasons
cytometry,36 and surrogate markers of cell cycle transit or for this increase in cell death are unclear but may relate
proliferation such as Ki67 immunohistochemical index.37 to local or general nutrient delivery. Hence, PC has a
When considering the biology of tumor growth as very long doubling time even in metastatic disease with
assessed by flow cytometry, the proportion of cells in relatively subtle changes in proliferation and cell death
G1 + S phases is thought to be most important, although that mark transition between stages of the progression
the level of aneuploidy (proportion of cells that do not of the disease.44 The relative balance between prolifer-
have a normal or diploid DNA content) appears to be ation as determined by cell cycle transit and pro-
important as a prognostic determinant in some tumors. grammed cell death varies between cancer types and
Another more direct parameter of the cell cycle is meas- may influence sensitivity to a variety of anticancer ther-
urement of tumor doubling time.2 apies. When PC is treated with androgen ablation, the
Also of importance is the growth fraction (proportion result is increased apoptosis in a minority of cells with
of cells within a tumor that are in active proliferative decreased proliferation in the majority of cells, demon-
phase), which can range from 25% to more than 90% in strating the importance of this balance in response to
human tumors, often with areas of variation within indi- therapeutic interventions.45
vidual tumor deposits. The rate of cell loss is also impor- In other tumors, varying amounts of reduction in cell
tant—in most tumors, it is high, ranging from 70% to proliferation and increase in apoptosis are seen among
greater than 90%.2 In general, the length of the G1 phase tumors exposed to hormonal and cytotoxic therapy.46 In
is one of the primary determinants of proliferative urologic oncology, the balance between reduced cellular
behavior—thus, if G1 is short, the duration of the cell cycle proliferation and increased cell death is best illustrated in
is usually rapid, whereas cells with a long G1 or those that the response of metastatic nonseminomatous germ cell
spend considerable time in G0 have a much longer cell tumors to chemotherapy. More than 90% of patients
cycle and are less sensitive to the impact of chemotherapy. with this condition are cured. In responding to
chemotherapy a major proportion of cancer cells
undergo apoptosis. However, certain cells within the
The Balance between Cell Proliferation
tumor, particularly within teratomatous elements, are
and Apoptosis
resistant to apoptosis and undergo reduction in prolifer-
Apoptosis or programmed cell death occurs in normal as ative rate and may differentiate into “mature” ter-
well as neoplastic cells. The process is complex and reg- atoma.47–50 These areas may remain as quiescent masses
ulated by a variety of molecular pathways. The best for protracted periods of time but have the potential to
understood of these pathways center on the tumor sup- de-differentiate and manifest cancer recurrence, which
pressor gene, p53,38,39 and the oncogene, Bcl2.40 A typically is resistant to further chemotherapy.51,52 Based
detailed review of apoptosis is beyond the scope of this on this, the therapeutic approach to these masses involves
54 Part I Principles of Urologic Oncology

surgical excision, particularly where teratoma has been by the extravascular approach, which includes oral,
identified in the primary tumor.53–55 Recent work sug- intramuscular, intrathecal, intravesical, and intraperi-
gests that mature teratoma may have a molecular signa- toneal routes. The route of extravascular delivery will
ture that distinguishes it from cancer cells undergoing influence absorption. Factors that determine the
apoptosis and those that appear morphologically unal- uptake characteristics of a drug include the structure
tered after chemotherapy.56 Whether this molecular and size of the molecule and its pKa and, thus, its solu-
approach will be of therapeutic utility remains to be seen. bility characteristics.
The clinical activity of specific agents may vary sub-
stantially with the nature of the route and schedule of
Clonality of Tumor Cell Populations
administration and consequent absorption. For example,
In animal tumors, which tend to be clonal in nature, first cyclophosphamide can be administered orally in a dose of
order kinetics appear to apply in response to chemother- 100 mg/m2/day for 14 days to patients with advanced PC,
apy—that is, a dose of single-agent chemotherapy will and is well tolerated, causing only modest myelosuppres-
kill a fixed proportion of tumor cells. For example, if a sion and gastrointestinal toxicity.64 When administered
tumor mass containing 107 cells is treated with an agent to similar populations of patients by intravenous bolus
that kills 90% of the cells, 9 × 106 cells will be killed by a injection (e.g., 750 to 1000 mg/m2 every 3 weeks), the
single dose, leaving behind 106 viable cells (or 10% of the side effects may be more substantial65 with no apparent
original tumor mass). A second dose will kill 9 × 105 cells, improvement in therapeutic outcome.
leaving 105 cells still alive. If treatment is not repeated, Successful intravesical chemotherapy is predicated
these cells will regrow, and the mass will rapidly return to on the desire for cytotoxics to be active locally without
its former size. This is also influenced by the proportion systemic absorption, thus protecting the patient from
of cells that undergo spontaneous cell death, as well as systemic side effects while maximizing the concentra-
the proliferative rate of any remaining viable cells. tion at the tumor surface. Thus, thiotepa, a small,
In the human setting, the situation is much more com- readily absorbed molecule is potentially less useful in
plex, as many human tumors appear not to be purely uni- this context than larger molecules, such as doxoru-
clonal in nature but rather are composed of multiple bicin or mitomycin C.66–68 Furthermore, the level of
subpopulations of cells with different characteris- systemic absorption of thiotepa can be increased if it
tics.52,57–61 It is not clear whether this is due to the evolu- is administered soon after transurethral tumor resec-
tion from single clonal populations (stem cells) or is due to tion in the presence of a residual denuded bladder
initial evolution of multiple clones in response to an initial epithelium.69
carcinogenic stimulus. Heterotypic signaling mechanisms Ultimately the key to therapeutic effectiveness of any
between the diverse cell types that comprise a human cytotoxic agent is a function of the product of its concen-
tumor are likely important regulators of primary tumor tration and the time available at the tumor site (C × T).
cell proliferation and other malignant characteristics.62,63 Most cytotoxics are administered by intravenous or
intraarterial routes, and the calculation of the actual
plasma C × T equation is made accordingly.
PHARMACOLOGY OF ANTICANCER AGENTS
The time course of drugs in the body is determined by
Distribution and Transport
the rates of drug absorption, distribution, metabolism,
and excretion. The mathematical description of these The amount of cytotoxic agent available at the tumor
rate processes is referred to as pharmacokinetics. Often target and the length of time during which it is present
the data can be fitted to mathematical models, which are determines its level of efficacy. Several factors will influ-
simplified descriptions of the complex physiologic reali- ence this, including the lipid solubility of the drug, its
ties. Many of the processes involved are so-called “first binding to protein and other carriers (with consequent
order”; that is, the rate at which the process occurs is pro- variation on free drug concentrations), and the mecha-
portional to the drug concentration, although some nisms available to allow entry into the tumor (such as
processes that are enzyme or carrier-dependent follow passive diffusion or active transport). A major factor will
Michaelis Menten kinetics in which the process is first be the plasma levels of the drug, a major determinant of
order at low concentrations and zero order (i.e., occur- which is its distribution characteristics. Following rapid
ring at a fixed rate) at high concentrations of the drug. intravenous injection, the plasma concentration (Cp ) of
the drug will initially fall rapidly. With the elapse of
time after the dose, the rate of decline will decrease. A
Absorption
plot of the natural logarithm (ln) Cp against time (semi-
Cytotoxic agents may be administered directly into the log plot) will generally show two components of the
circulation (intravenous or arterial administration) or plasma decay: an initial rapid component and a subse-
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 55

Metabolism
quent slower component, both of which have the char-
acteristics of log-linear, that is, first order, processes. Drug metabolism may occur both outside and within
Mathematically, such plasma decay can be fitted to a tumor cells. There are two important types of metabolism
two-compartment model in which the body is conceptu- of antitumor agents. Antitumor agents, which resemble
alized as consisting of two compartments, a central com- normal metabolites, are often metabolized by the same
partment into which the drug is introduced and a mechanisms as the normal metabolites that they resemble.
peripheral or tissue compartment into which it diffuses, Most purine and pyrimidine antimetabolites require activa-
ultimately to equilibrium. The second component of the tion to a nucleotide (usually the triphosphate) in order to be
plasma decay consists of elimination processes consist- active, and these reactions are carried out by the mecha-
ing of metabolism or excretion. The rate of this second nisms in the cell used to metabolize the corresponding nor-
process will give the half-life of the drug in the body and mal preformed purines and pyrimidines (the so-called
is an important pharmacokinetic characteristic of all salvage pathways). Some of the antifolates undergo polyg-
drugs (t1/2β). For some drugs, such as the anthracyclines, lutamation by the mechanisms used for folates.
a third component of the plasma decay is seen indicat- Degradative pathways such as those responsible for reduc-
ing a so-called deep tissue compartment, usually corre- ing 5-fluorouracil (5-FU) to dihydro-5-fluorouracil and
sponding to the binding of the drug to some tissue converting cytosine arabinoside to the corresponding uracil
component, such as nucleic acid, from which the drug is arabinoside by deamination,73–75 are also active in the cell.
slowly released. An even simpler model in which the These reactions occur in the cells of the tumor and in the
body is regarded as a single compartment can sometimes cells of normal tissues. In the case of capecitabine, a novel
be used; however, for many drugs it can lead to major oral fluoropyrimidine carbamate, intracellular conversion
errors in computing the important pharmacokinetic of parent drug to 5-FU occurs preferentially so that there
parameters. It is important to recognize that these is more active drug in cancer tissue than adjacent normal
“compartments” are mathematical constructs, which cells.76,77 This occurs under the enzymatic effects of cyti-
usually have little or no correspondence with actual dine deaminase and thymidine phosphorylase, which
physiologic compartments. The total area under the metabolize capecitabine to 5′-deoxy-5-fluorocytidine, 5′-
plasma concentration-times-time curve (AUC or C × T) deoxy-5-fluorouridine, and fluorouracil.76,78 In addition to
is an important measure of the total exposure of the tis- these specific metabolic reactions, compounds that do not
sues to the drug. Other important pharmacokinetic show resemblance to physiologic substrates are metabo-
parameters, which can be calculated from the indices of lized primarily in the liver by the pathways used for detox-
plasma decay, are the total body clearance (ClB) and the ification of xenobiotics. The most important of these is
apparent volume of distribution (VD), the theoretical oxidation, often followed by conjugation. Oxidation is car-
volume required to dissolve the total body content of ried out by cytochrome P450, a family of enzymes located
the drug if it were uniformly distributed in the concen- primarily in the microsomal or smooth endoplasmic retic-
tration found in plasma. ulum fraction of the liver.79,80 This pathway is very non-
In general, the drug is distributed between the specific in terms of structural requirements and oxidizes
intravascular, extracellular and intracellular water but has most lipid soluble compounds. It is this pathway that is
to cross a membrane to pass from one of these to another. responsible for the initial oxidation of the oxazaphospho-
In addition, certain sites are protected from easy drug rine ring of the oxazaphosphorines cyclophosphamide and
access. Such sanctuaries are usually characterized by ifosfamide, a reaction leading to the conversion of these
lower drug concentration than other tissues. Conversely, compounds to their active metabolites.81–84
some drugs are disproportionately concentrated in par- Knowledge of the metabolism of cytotoxic agents is
ticular tissues as exemplified by the accumulation of important in designing treatment strategies, for example,
estramustine in prostatic tissue.70 intravesical delivery of cyclophosphamide would make
The presence of sanctuary sites may be of real impor- no sense, as the drug requires hepatic metabolism to its
tance; for example, the blood-brain barrier appears to active form to be effective (see later). In the patient with
protect the brain against the local uptake of cytotoxic hepatic dysfunction or failure, impaired hepatic conjuga-
agents, and thus the brain may be the site of first relapse tion and/or oxidation will alter the metabolism of dox-
in tumors that are otherwise quite responsive to orubicin, the taxanes, irinotecan, and the vinca alkaloids,
chemotherapy.71 Similarly, it appears that the testis may whereas the microsomal activation of cyclophosphamide
functionally constitute a sanctuary site against the effect may be impaired in this clinical setting.85–91
of chemotherapy—up to a third of patients treated for
metastatic testis cancer before surgical removal of the
Excretion
affected testis will have residual cancer within the testis at
subsequent orchiectomy, despite the attainment of extra- Excretion of cytotoxic agents occurs predominantly in
testicular complete response.72 the kidneys and liver, and abnormalities in the function of
56 Part I Principles of Urologic Oncology

either or both organs may substantially influence the pat- demonstrated that response to the cellular effects of
tern of toxicity.85 Renal dysfunction will particularly agents as diverse as the vinca alkaloids, actinomycin D,
affect the disposition of the platinum complexes, estramustine, mitoxantrone, and doxorubicin is reduced
methotrexate, and bleomycin,92–95 and this may be an in normal and malignant cells that express a protein
important issue in a patient with renal tract outflow complex on the cell surface, coded by a series of mul-
obstruction due to a large primary tumor of bladder, tidrug resistance (mdr) genes.112–114 This occurs as a
prostate, or urethra or with obstruction from enlarged result of reduced intracellular concentrations of the
retroperitoneal nodes. agents due to increased cellular efflux.115 Expression of
the mdr phenotype may be present before therapy or
appear as a result of induced resistance during treat-
Factors Modifying Pharmacokinetics
ment.116 More recent research has elucidated a diverse
Absorption of drugs may be affected by diseases of the GI family of P-glycoproteins that acts physiologically to
tract, previous surgery, compounds that change the pH facilitate influx and efflux of xenobiotics from a variety of
of the gut, coadministration of other drugs, and a variety cells.117,118 Alteration in the expression and activity of
of other factors.96–98 Distribution can be influenced by these molecules can have a significant effect of drug
disease states, such as cardiac failure, ascites, pleural effu- pharmacokinetics both at a whole body and cellular
sion, and edema.99 Age and amount of adipose tissue may level.119
have an impact on the clearance and toxic effects of cyto- Expression of mdr phenotype has been identified in
toxic agents. For example, obesity appears to reduce the renal carcinoma,120 although its significance has been dif-
clearance of both doxorubicin and ifosfamide,100,101 ficult to define as most renal carcinomas are resistant to
although it is not clear that this has an impact on toxicity the available cytotoxic agents, and the presence/absence
of the individual drugs. Age may alter disposition of dox- of this phenotype does not correlate with outcome. The
orubicin; for example, Robert and Hoerni102 demon- study of the multidrug phenotype in bladder cancer cells
strated reduced clearance in older patients, compared has also proved to be a difficult problem, as the expres-
with younger cohorts. Numerous factors have been sion of P-glycoprotein in bladder cancer has been highly
shown to affect the rate at which drugs are metabolized variable and inconstant.121–124 It has now been shown
by cytochrome P450 microsomal enzymes in the gut and that expression of P-glycoprotein may be upregulated in
liver.79,98,103A large number of compounds can induce resistant populations of bladder cancer cells after treat-
P450, including phenobarbital, carbamazepine, rifampin, ment with the MVAC regimen.125 In other tumor types,
and phenytoin, chlorinated hydrocarbon insecticides, multidrug resistance can occur in the absence of expres-
food additives, and tobacco consumption. Some antineo- sion of the 170 kD P-glycoprotein, whereas other pro-
plastic agents may inhibit drug metabolism, as may the teins may be associated with very similar patterns of
occurrence of hepatic disease.104,105 Excretion of com- resistance,126 perhaps explaining this phenomenon in the
pounds by the kidneys depends heavily on renal function. absence of expression of this P-glycoprotein.
Coincidental administration of compounds that can com- Ultimately, apart from its predictive function, this
pete for tubular reabsorption may have an effect on renal work is unlikely to be of great importance unless the mdr
clearance of certain compounds,106 as may urinary pH if phenotype can be overcome at a functional level.127 For
the compound is able to become ionized.94,107,108 example, the calcium channel blockers, such as vera-
pamil, have been shown to reverse multidrug resist-
ance,128 although the toxic side effects of this approach
MECHANISMS OF DRUG RESISTANCE
have precluded routine use while some data suggest that
There are several mechanisms of resistance to cytotoxic compensatory effects such as altered blood flow may
chemotherapy (Table 4-1). In general, these can be clas- diminish tumor sensitization at clinically attainable con-
sified on the basis of cellular distribution. Intracellular centrations.129 Although clinical trials have not yet been
factors include those that act at the cell surface, others published in bladder cancer, work initiated in our labora-
within the cytoplasm, and those that function at the level tories suggests that verapamil can overcome the impact
of the nucleus. In addition, there are extracellular factors, of the mdr phenotype, at least in bladder cancer cell lines
such as those that affect the distribution and metabolism in vitro.130
of the drugs, including competitors for cellular transport Furthermore, Brandes et al.131,132 have shown that
mechanisms. N,N-diethyl-2-[4-(phenylmethyl)phenoxy]-ethanamine
Some cytotoxic agents can be exported from tumor (DPPE, Tesmilifene), an intracellular histamine antago-
cells through a mechanism based on the cellular surface, nist, enhances the anticancer effect of cytotoxic agents
the so-called multidrug efflux pump, which was origi- and that DPPE may modulate the function of the P450
nally characterized by the expression of a specific 170 kD hepatic enzyme system and may alter the expression of
protein complex, P-glycoprotein.109–111 It was initially multidrug resistance phenotype. They have shown that
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 57

Table 4-1 Mechanisms of Tumor Drug Resistance


Example or Potential Cause

Macro-pharmacokinetic mechanisms: ineffective dose of drug in the body

Inadequate dose Miscalculation or decreased dose due to prior excessive toxicity

Inadequate absorption Short bowel syndrome after surgical resection or coingestion


of drug binder such as cholestyramine

Induced metabolism P450 microsomal enzyme-inducing drug for liver metabolized agent such as
paclitaxel or docetaxel

Failure to metabolize to active drug Ifosfamide or cyclophosphamide

Induced excretion

Micro-pharmacokinetic mechanisms: ineffective intracellular concentration of drug

Impaired tumor blood flow Local radiotherapy or scarring after surgery, sanctuary site such as central nerves
system or testis

Impaired tumor uptake Methotrexate

Induced intracellular metabolism 5-FU metabolism, methotrexate


of drug to inactive or exportable
metabolite

Drug efflux P-glycoprotein and doxorubicin

Pharmacodynamic intracellular mechanisms

Mutated target that does Tubulin mutations and taxane or vinca alkaloid therapy. Androgen receptor
not respond to drug mutation for androgen deprivation

Overexpressed target that Thymidylate synthetase and fluoropyrimidines


overcomes drug effect

Compensation by cellular mechanisms DNA repair enzyme over expression and platinum drugs. Apoptosis resistance due
to mutated p53 or overexpressed Bcl2. Lack of cell cycle response due to pRB
mutation, cyclin overexpression, or loss of CDK inhibition

DPPE appears to enhance the anticancer efficacy of cellular availability. Inhibitors of GSH synthesis, such as
cyclophosphamide against hormone-refractory PC buthionine sulfoximine, have been shown to cause a
(HRPC), an observation that we have confirmed for decrease in intracellular levels of GSH with a concomi-
mitoxantrone.133 tant increase in the cytotoxicity of some anticancer agents,
The mechanisms of resistance to the platinum coordi- such as the alkylating agents, cisplatin136 and paclitaxel.137
nation complexes have been studied in detail, particularly Although much of the experimental data regarding the
in relation to ovarian cancer and malignant melanoma. significance of glutathione in cisplatin resistance has been
Although several mechanisms have been identified, derived from models of ovarian cancer, very high levels of
including factors that influence cellular accumulation, sig- glutathione are present in cell lines derived from bladder
nal transduction, ionic fluxes, and intracellular enzyme cancer, and this may correlate with cisplatin resistance.138
function,134,135 the function of the intracellular scavenger, It should be emphasized that our understanding of
glutathione (GSH), has increasingly been the focus of these mechanisms is relatively crude, and there appear to
particular attention in the context of the resistance of be other factors that influence the responsiveness of
bladder cancer to the effects of cisplatin. GSH is found in bladder cancer to cytotoxic chemotherapy. It appears that
most mammalian cells and has many functions, including the expression of several oncogene products may influ-
regulation of protein and DNA synthesis and detoxifica- ence resistance to cytotoxic agents. The exact nature of
tion. It appears to react with cisplatin, reducing its intra- this interaction is not yet clear and is particularly difficult
58 Part I Principles of Urologic Oncology

to define as several of these products code for specific cells, the greater the chance of spontaneous mutation.
aspects of cellular growth control irrespective of expo- As a consequence, they proposed that the most effective
sure to cytotoxic agents. For example, it has been shown mechanism for cancer killing would be to initiate
that the interaction of EGF and its specific receptor chemotherapy early (with a small cellular burden) and
(EGFR) are involved in the regulation of growth of blad- to introduce multiple agents in an attempt to overcome
der cancer. However, in vitro treatment with EGF can the various mechanisms of resistance. To date, this
increase cellular sensitivity of epithelial tumors to cis- hypothesis has not been validated in clinical trials,
platin.139,140 Conversely, it has also been shown that spe- although most of the studies reported have been flawed
cific monoclonal antibodies and small molecules block and have not truly evaluated the principles of this
EGFR function,141 and treatment with these agents plus hypothesis.
cisplatin can cause a synergistic antitumor effect.142 Another model of resistance, proposed after a
These data are particularly difficult to interpret in view reassessment of the model presented by Goldie and
of the previously documented impact of expression of Coldman,155 leads to the conclusion that cytotoxic
EGFR on the natural history of bladder cancer, and the agents can be used most effectively in sequence rather
demonstration that erbB-2 gene amplification and over- than as combination schedules. Thus an initial series of
expression is an adverse prognostic determinant in blad- treatments with the less effective of two drugs would
der cancer.143–145 Recent clinical trials have not eliminate a proportion of the tumor cells present, leav-
delineated a clear additive or synergistic effect of anti- ing behind a resistant population that may respond to
EGFR therapies with cytotoxic chemotherapy but this is several courses of treatment with the more effective
not surprising given the conflicting preclinical model drug; in this way, it is postulated that the impact of the
data available. less effective therapy can be maximized.155
Another complex relationship has been demonstrated
between the expression of p53 (a suppressor gene prod-
ASSESSMENT OF THE EFFICACY
uct), growth regulation, and cytotoxic response in blad-
OF ANTICANCER TREATMENT
der cancer. Alterations of the p53 gene are among the
most frequent genetic abnormalities found in human
The efficacy of anticancer treatment can be assessed in a
cancer146 and appear to have a broad range of postulated
variety of ways, depending on the endpoints under con-
roles in cell growth control, including involvement in
sideration. In most current studies, a reduction in
cellular repair and apoptosis.38 Apoptosis is regarded as
tumor-related symptoms (subjective response), the
one of the forms of physiologic cell death as it represents
development of tumor shrinkage (objective response),
a genetically determined cellular sequence that is part of
length of disease-free or overall survival or improvement
the normal tissue homeostatic mechanism. It has been
in quality of life are regarded as significant indices of
shown that p53, which is normally present only tran-
patient benefit in response to treatment. In order to
siently, can be induced to accumulate within the cell by
assess these outcomes objectively and accurately, we use
exposure to cytotoxic agents, such as cisplatin and mito-
the process of the clinical trial wherein a defined popu-
mycin C, and conversely that p53-dependent apoptosis
lation of patients, who fit specified criteria, are managed
modulates the cytotoxicity of radiotherapy, 5-FU, and
according to a defined protocol with specified measure-
doxorubicin.147–151 These issues may be of particular
ment of outcome. A clinical trial may be one of two
importance as it has already been postulated that p53
types66,156: (i) An “explanatory” trial has the primary aim
expression may constitute an independent prognosticator
of acquisition of information. Thus it usually only
of response to the MVAC regimen.152,153 As p53 may be
requires small numbers of patients and often has a bio-
induced by cytotoxic exposure, it is possible that the tim-
logic endpoint (e.g., tumor response). A typical example
ing of tissue sampling may be critical in determining the
is the assessment of the new anticancer agent, gem-
expression of this potential prognostic factor, especially if
citabine, in the treatment of advanced bladder cancer,
intravesical or systemic chemotherapy has been used
which provides the documentation of response and tox-
previously.
icity but does not give comparative information versus a
standard type of treatment. It is often appropriate to
Models for Overcoming Drug Resistance of Tumor report only the patients who are fully treated (complete
Populations: Clinical Implications the minimum requirements of the protocol of assess-
ment) in order to minimize the chance of rejecting an
Several models have been proposed to explain the vary-
active new approach. However, this may overestimate
ing levels of resistance to the impact of chemotherapy
the potential benefit of the drug in the general patient
seen in human tumors. For example, Goldie and
population. (ii) A “pragmatic” trial is concerned with the
Coldman154 proposed that tumors have a spontaneous
assessment of comparative patient benefit, which can
mutation rate and that the larger the number of tumor
then be used as a guide to decision-making in treatment.
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 59

For example, the recent intergroup randomized trials comes, have been published elsewhere.163–165 In general,
that compare neoadjuvant intravenous chemotherapy phase II studies require relatively small numbers of
plus locoregional treatment versus locoregional treat- patients (usually less than 50, depending on the number
ment alone for invasive bladder cancer are pragmatic tri- of responses seen).
als and require large numbers of patients.157–159 As the Phase III trials are designed to define the role of a
goal of a pragmatic trial is to determine overall benefit new treatment compared to a standard approach. These
to all eligible patients, endpoints should include both studies are pragmatic in concept and usually require ran-
quality and quantity of survival, and it is important to domization and relatively large numbers of patients. The
report the outcome for all entered patients, irrespective number of required patients is predicated on the level of
of treatment received. statistical confidence (power) that is required for the
Another classification of clinical trials is according to outcome.
the descriptors, “phase I,” “phase II,” “phase III,” and A more recent term, the “phase IV” trial, is used to
“phase IV”. A phase I trial is designed to determine the describe a post-marketing study—that is, one in which a
appropriate clinical dosage of a new treatment (often pharmaceutical company attempts to develop additional
being translated from preclinical studies) and to assess information regarding the indications for a newly
the side effects of this treatment. Such trials will often approved agent (which is now being marketed); sometimes
incorporate pharmacologic studies that will allow a such studies are actually marketing (or “seeding”) studies,
clearer understanding of the distribution and metabolism in which the primary purpose of the trial is to increase the
of the novel agent in humans. Although anticancer level of interest or familiarity among physicians.
response is recorded, this is not a primary endpoint of a The requirements of these clinical trials and a detailed
phase I trial, and the process of consent must make discussion of the potential sources of error (Table 4-2)
patients understand that the chance of a significant are beyond the scope of this chapter but have been
tumor shrinkage is only small. Patients will often partic- reviewed elsewhere.66,166
ipate in such trials in the hope that they may secure the
small percentage chance of success, or occasionally for
SPECIFIC AGENTS COMMONLY USED AGAINST
reasons of altruism.160 Regrettably, it has become
GENITOURINARY CANCERS
increasingly clear that patients often have a very poor
Platinum Complexes
understanding of the true purpose of phase I trials and of
the small likelihood of individual patient benefit.161 Cisplatin (cis-diamminedichloroplatinum II) was intro-
Phase I trials are explanatory in nature and should be duced into the clinic in 1972 when observations by
designed to minimize the expenditure of patient Rosenberg167 of its activity in interfering with the growth
resources, usually with only 3 to 4 patients being entered of Escherichia coli led to the demonstration of broad-
at each dose level.162 These numbers allow relatively spectrum antitumor activity in experimental tumors. The
accurate identification and description of toxicity at each lead compound of the platinum antitumor drugs, cis-
dose level, without the necessity to treat unreasonably platin (Figure 4-1), has contributed in a major way to the
large numbers of patients at potentially subtherapeutic development of cure in testicular germ cell tumors and is
doses. highly active in combination in bladder cancer.
Unless the toxicity from a phase I trial is prohibitive or The mode of action of the platinum complexes
truly unpredictable and dangerous, the new agent will involves the formation of positively charged (electron-
usually proceed to phase II testing. In most cases, an deficient) moieties that form adducts with DNA. These
absence of antitumor activity in phase I testing will not
necessarily preclude initial phase II assessment, although Table 4-2 Potential Sources of Error in Clinical Trials
demonstration of activity in phase I testing tends to raise Failure to establish uniform reporting criteria of efficacy
the priority of a novel agent for further investigation. and toxicity
Phase II trials are also explanatory and attempt to define
the extent to which a new approach has antitumor activ- Measurement error
ity. These studies usually involve treatment of defined
Bias in patient sampling: selection, exclusion
groups of patients with a particular disease with a prede-
termined dosage of a novel agent, with assessment of Insufficient patient numbers
tumor shrinkage and toxicity as primary endpoints. The
demonstration of tumor shrinkage does not equate with Use of historical controls
long-term patient benefit, which is assessed subsequently
Stage migration
in a phase III trial. Detailed mathematical assessments of
patient numbers required to document true anticancer Inadequate follow-up
efficacy, minimizing false positive and false negative out-
60 Part I Principles of Urologic Oncology

O O
H3N Cl H3N O NH2 O C

Pt Pt Pt

H3N Cl H3N O NH2 O C


O O

1 2 3

Figure 4-1 Structures of cisplatin (1), carboplatin (2), oxaliplatin (3).

adducts are primarily intrastrand cross-links. Interstrand combination regimens that include the vinca alkaloids,
cross-links also form but in much smaller amounts. They neurotoxicity is an important side effect. This may pres-
are, however, much more lethal than the intrastrand ent clinically as peripheral or autonomic neuropathy and
cross-links. The contribution of the two types of adducts may be seen more commonly on nerve conduction test-
to the overall cytotoxicity of platinum complexes is a ing.179 A range of cardiovascular side effects has been
matter of dispute. Based on the sites of action of cisplatin, reported, including hypertension, myocardial infarctions,
its major focus of activity is in the G1 and S phases of the EKG abnormalities, and an increased risk of peripheral
cell cycle. Resistance to the platinum complexes may be thromboembolic phenomena and Raynaud’s phenome-
due to impaired cell uptake of the drug,168 inactivation of non.180 However, in some cases, the relationship between
the active species by intracellular thiols (primarily glu- cisplatin and these side effects is obscured by the impact
tathione),138,69,170 and enhanced capacity of the cell to of antecedent cigarette smoking by the population of
repair the damage to DNA.171–174 patients under treatment. Myelosuppression may occur
Cisplatin is usually administered intravenously or with standard doses of cisplatin, with anemia occurring in
intraarterially, and in both instances it has been shown particular. Occasionally a Coombs’ positive hemolytic
that its most dangerous toxic side effect, nephrotoxicity, anemia has been documented. Leukopenia and thrombo-
is markedly reduced or eliminated if a high urine output cytopenia occur sometimes but are usually not dose lim-
is maintained (via enforced parenteral hydration before, iting. In an attempt to overcome the substantial toxic
during and after cisplatin administration, augmented by effects of this useful agent, a range of analogs has been
the use of mannitol, and in some instances small doses of developed in an effort to reduce toxicity without loss of
furosemide). In early studies, cisplatin was administered activity (see later).
for intravesical use by instillation through a urinary Cisplatin is still among the most widely used cytotoxic
catheter, but unpredictable allergic reactions with occa- agents in the management of genitourinary cancer. In
sional anaphylaxis were documented, and this indication addition to being one of the drugs of choice in the man-
is no longer under investigation. agement of germ cell tumors and bladder cancer, cisplatin
Measurement of total plasma platinum after adminis- has activity against adenocarcinoma and small cell carci-
tration of cisplatin shows a triphasic disappearance curve noma of the prostate, adrenal cancer, and penile cancer.
with half-life values of 20 minutes, 1 hour, and 24
hours92,175; the terminal phase probably reflects the slow
Carboplatin
release of platinum from plasma proteins. More than
90% of excretion is via renal mechanisms (a combination As noted earlier, cisplatin is one of the most active and
of glomerular filtration and tubular secretion), with less useful agents in the management of genitourinary malig-
than 10% due to biliary excretion. The plasma decay of nancy. However, its substantial toxicity has led to the
unchanged cisplatin generally has a monophasic pattern search for analogs with equivalent anticancer activity but
with a t1/2 of less than 1 hour.176 a reduced pattern of side effects, culminating in the intro-
Renal failure, with acute tubular necrosis, degenera- duction of several second-generation compounds into
tion, and interstitial edema, is the dose-limiting toxic clinical trials. One of these, carboplatin, was developed
effect, as noted above.177,178 Additional side effects of largely at the Institute for Cancer Research in the United
parenteral administration include severe nausea and Kingdom and is now commercially available in the
vomiting, diarrhea, and occasional anaphylaxis. United States. While considerably less toxic than cisplatin
Ototoxicity, manifested by high frequency hearing loss, (it has very little nephrotoxicity, considerably less neuro-
has been well documented, with a greater level of dam- toxicity and ototoxicity, and produces much less nausea
age demonstrated by audiometry than on clinical and vomiting), it is highly cross-resistant with the older
grounds. With higher doses of cisplatin, and especially in compound. This may be related to the fact that once the
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 61

Anticancer Antibiotics
cyclobutanedicarboxylato-leaving group has separated
Bleomycin
from the molecule, the structure of the active hydrated
species is identical to that of cisplatin (see Figure 4-1). Bleomycin is isolated from the fungus Streptomyces vertic-
Thus the major difference is in the kinetics of the forma- ullus and consists of a complex molecule of high molecu-
tion of the active species.181 Carboplatin is much more lar weight. It is composed predominantly of so-called A2
stable in serum than cisplatin and diffuses into tissues rap- peptides, which contain a DNA-binding portion and an
idly so that only 24% of a dose is bound by plasma pro- iron-binding component at the opposite end (Figure
tein 4 hours after a dose.182 The terminal serum half-life 4-2). Its primary action is to produce single-strand and
of platinum is similar in patients given either carboplatin double-strand breaks in DNA through the action of oxy-
or cisplatin.93 The elimination of carboplatin occurs pre- gen radicals formed in association with activation of the
dominantly through the kidneys, where renal clearance Fe++–bleomycin complex that binds to nitrogen-containing
closely parallels glomerular filtration rate.183,184On this groups in the bleomycin molecule. The entire complex,
basis, Calvert has developed a formula, based on creati- consisting of Fe++–bleomycin–oxygen binds to DNA,
nine clearance, that predicts safe carboplatin doses for through intercalation by the bithiazole groups of
individual patients relative to the chance of myelotoxicity, bleomycin, and DNA damage is created through the for-
specifically thrombocytopenia.184 mation of superoxide or hydroxyl radicals. Tumor cells
Carboplatin has activity in testicular, urothelial, and are most sensitive to the effects of bleomycin in late G2
prostate185–187cancers but not renal cell cancer. Despite or the M phase,206 although cells may also be killed in G1
this it has failed to establish itself as first-line therapy in phase.207 The mechanism of resistance to bleomycin has
any urologic malignancy. Its equivalence with cisplatin not been delineated with certainty, although it does
has been demonstrated against lung cancer188,189 and in appear that a cytosolic hydrolase may function to inacti-
ovarian cancer,190,191 but this has not been the case in vate the drug.208
genitourinary cancer.192,193 Of particular importance, it Bleomycin is administered either by subcutaneous
appears that carboplatin is inferior to cisplatin for man- bolus dosing, intravenously (bolus or continuous infu-
agement of cancers of the testis and bladder. However, in sion) or by intramuscular injection. The optimal route
combination carboplatin may be incorporated into regi- has not been defined, although it is most commonly used
mens that will eventually prove to have favorable efficacy (in combination with cisplatin and etoposide) as a weekly
toxicity profiles compared to those incorporating cis- bolus dose. Bleomycin has a biphasic clearance, with t1/2α
platin. The results of trials directed at proving this in of about 20 minutes and a t1/2β of about 3 hours.
urologic cancer have been inconclusive or disappointing Bleomycin is excreted predominantly via the kidneys,
to date.194–197 Carboplatin remains a well-tolerated, with more than 50% being excreted in the first 24
largely second-line therapy in a range of genitourinary hours.95 Of importance, the dosage should be modified in
malignancies. a patient with impaired renal function, and caution
should be used when the drug is administered after cis-
platin, which can cause transient renal dysfunction.
Oxaliplatin
The major toxic effect of bleomycin is interstitial
Oxaliplatin was synthesized by Kidani et al.,198 and there pneumonitis, the pathogenesis of which is poorly under-
is evidence that in solutions containing high chloride stood. The syndrome may be characterized by the evolu-
content the oxalato group is replaced by chloride.199 The tion of subtle pulmonary symptoms, including cough,
drug produces a characteristic peripheral neuropathy that dyspnea on exertion, chest pain, and fever, or occasion-
is characterized by a cold-induced dysesthesia, which ally may be revealed only by changes in lung function
tends to be cumulative.200,201 Oxaliplatin has undergone tests. Pulmonary toxicity occurs especially in older
extensive phase II and phase III clinical trials predomi- patients, those who have previously received pulmonary
nantly in gastrointestinal malignancies, while demon- or mediastinal irradiation, those with underlying lung
strating some activity in a range of cancers including disease, and in particular in cigarette smokers. In a study
urothelial tumors in phase I testing.202 A group of of the chronic toxic effects of bleomycin, we were unable
German investigators has shown significant anticancer to demonstrate major pulmonary toxicity in patients who
effect in patients with previously treated germ cell did not smoke.179 In our experience, the acute toxicity of
tumors. As a result of these trials, oxaliplatin has recently bleomycin can usually be ameliorated by corticosteroids,
been licensed internationally for the treatment of col- although care should be taken to avoid tapering the dose
orectal cancer.203 It will be further evaluated in other pri- too quickly. Other side effects include mild myelosup-
mary tumor sites, including bladder cancer and germ cell pression, allergic reactions, fever, Raynaud’s phenome-
tumors, for increased activity compared to cisplatin and non, and cutaneous pigmentation.
carboplatin204,205 or for activity in platinum-resistant The major indication for the use of bleomycin is in the
tumors. management of germ cell tumors, and most attempts to
62 Part I Principles of Urologic Oncology

NH2

H2N O NH2

N
O O
H
R1

N
O
H
O HO N
H2N 6
O NH N
N N O
H H HO 6

N
O CH3
HO N BLEOMYCIN A2 –NH(CH2)3 S+
OH OH
O H CH3

O NH2
1 OH
OH BLEOMYCIN B2 –NH(CH2)4 N +
O H
OH NH2

O NH2

O OH
O OH
O
O CH2O C NH2 OH

H2N OCH3
O
CH3 CH3O O OH
N N
3
CH3 O
O NH2
2
HO

H
N
OH O HN OH

OH O HN OH
N
H
4

Figure 4-2 Structures of bleomycin (1), mitomycin C (2), doxorubicin (3), mitoxantrone (4).
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 63

delete it from standard protocols have resulted in nea, accompanied by changes in pulmonary function
reduced pulmonary toxicity but at the expense of a tests, and responds dramatically to high dose corticos-
reduced cure rate.209,210 In addition, it has also been teroids.218 Mitomycin C is a vesicant and causes extensive
applied in some series to the management of advanced soft tissue reaction if extravasated; sometimes the ulcera-
cancer of the penis. There appear to be no current indi- tion will occur as a delayed phenomenon.
cations for its use in the treatment of bladder cancer, The major indications for the use of mitomycin C
renal carcinoma, or PC. include intravesical administration for superficial bladder
cancer and intravenous administration for advanced blad-
der cancer; activity of mitomycin has also been demon-
Mitomycin C
strated in the management of adenocarcinoma of the
Mitomycin C is an anticancer and antibacterial antibiotic prostate and in advanced penile carcinoma. There is no
isolated from Streptomyces caespitosus.211 It is unusual in routine role for this agent in the management of renal
that it undergoes activation in a hypoxic environment, a carcinoma, germ cell tumors, or adrenal carcinoma.
characteristic of solid tumors. It is activated by visible
(but not ultraviolet light) and is unstable in acidic and
Doxorubicin and Other Anthracyline Antibiotics
extreme basic conditions. Mitomycin is activated by
reduction of a quinone moiety, mediated by microsomal Doxorubicin (Adriamycin) is a high molecular weight
enzymes, such as cytochrome reductase, DT-diaphorase, anthracycline antibiotic isolated from Streptomyces
xanthine oxidase or cytochrome P450 reductase, or even peucetius var. caesius. Its structure includes a water-soluble
by exposure to acidic pH. This releases a methanol side basic reducing amino sugar linked by a glycosidic bond to
chain from the molecule and allowing an aziridine ring to carbon atom 7 on the D ring of the chromophore agly-
open (see Figure 4-2). This exposes the alkylating site. cone, adriamycinone. This four-ring planar structure
The alkylating agent cross-links complementary DNA constitutes the anthraquinone nucleus (see Figure 4-2).
strands. Oxygen-free radicals may also participate in the Several analogs are in clinical use, including epirubicin
cytotoxic function of mitomycin C through the produc- (which differs in the steric configuration of the −OH
tion of DNA strand breaks.212,213 group on the 4′ position of the daunosamine sugar) and
Mitomycin is usually administered by intravenous idarubicin (which lacks an A ring −OCH3 substitution).
injection or through a urinary catheter for intravesical A detailed discussion of each of these agents is beyond
use. Its large molecular weight allows it to be retained the scope of this review but has been covered elsewhere
within the luminal surface of the bladder, without signif- in detail.219
icant systemic absorption. Because the drug is degraded There appear to be several possible mechanisms of
at acid pH, the urinary pH must be maintained at alka- cytotoxic action of the anthracycline antibiotics, includ-
line levels for intravesical use. ing the formation of free radicals, intercalation between
Because of its ubiquitous metabolism, mitomycin is nucleotide pairs in the DNA molecule, and the initiation
cleared rapidly after intravenous or intraarterial injec- of topoisomerase type II-dependent DNA fragmenta-
tion. Renal excretion is minor (10% of administered tion.220 Doxorubicin appears to be active in all phases of
dosage), and hepatic excretion seems to play only a minor the cell cycle, with its maximal effect in S phase; however,
role despite the importance of hepatic inactivation. it is not phase specific. For example, its topoisomerase
Hepatic dysfunction does not appear to alter the phar- type II function is concentrated in G2.
macokinetics of this drug to any great extent.214,215 Doxorubicin and epirubicin can be administered by
The major toxic effect of mitomycin is myelosuppres- bolus or continuous intravenous or intra-arterial injec-
sion, which involves platelet, leukocyte, and erythrocyte tion or via urinary catheter for intravesical use. These
precursors.62 Of importance, this myelosuppression may drugs have a characteristic red color. Doxorubicin is
be delayed and can occur 6 to 7 weeks after treatment. extremely vesicant, and great caution should be used in
The level of myelosuppression correlates with dosage, its administration. After intravascular administration,
and there appears to be a cumulative effect as well. Other the pharmacokinetics of doxorubicin and analogs are
side effects include gastrointestinal disturbances, occa- best represented by two- or three-compartment model-
sional stomatitis, and renal toxicity.216,217 Mitomycin C ing.221 The plasma half-lives are 11 minutes, 3 hours,
can occasionally induce microangiopathic hemolytic ane- and 25 to 30 hours; it is likely that the long terminal
mia with associated renal failure (the hemolytic-uremic phase is due to slow dissociation from DNA. These
syndrome); this syndrome can be delayed in onset.216,217 agents are rapidly distributed (into liver, lymph nodes,
Not infrequently, mitomycin C will cause interstitial muscle, bone marrow, fat and skin but not central nerv-
pneumonitis, especially in high cumulative doses and in ous tissue), and more than 70% is bound to plasma pro-
patients who have had prior chest irradiation; the syn- teins. These drugs are extensively metabolized and are
drome is characterized by progressive cough and dysp- excreted predominantly in the bile and feces. Biliary
64 Part I Principles of Urologic Oncology

excretion accounts for more than 40% of each dose,87 anticancer effect of standard cisplatin-based combination
whereas less than 10% is excreted in the urine. regimens. Limited activity against adrenal carcinoma has
Obstructive hepatic dysfunction interferes with the dis- been recorded,226,227 but the drug appears to be inactive
position of these agents and mandates dose modifica- against renal carcinoma.228
tion, whereas hepatocellular dysfunction without
impaired conjugation does not seem to require these Mitoxantrone
changes in dosing.222 One potentially attractive feature
of idarubicin is its retention of antitumor activity when Mitoxantrone is an anthracenedione, structurally related
administered orally. Although apparently useful in the to the anthracyclines.229,230 In comparison with anthracy-
treatment of leukemia and lymphoma, its utility against clines the structure of mitoxantrone maintains the planar
the solid tumors appears more limited. polycyclic aromatic ring associated with intercalation but
The most common dose-limiting acute toxic effect is is without their characteristic sugar moiety associated
myelosuppression, with leukopenia at 10 to 14 days after with production of intracellular free radicals implicated
treatment. Thrombocytopenia and anemia occur but are in anthracycline cardiotoxicity (see Figure 4-2). It has
less clinically important. As noted above, doxorubicin is a characteristic blue color and is administered as an intra-
significant vesicant and can produce extensive ulceration venous infusion.230
and soft tissue necrosis after extravasation. The other Mitoxantrone is highly protein bound in serum
dose-limiting toxic effects are found in the heart. Dose- (>95%) and has a very large volume of distribution (450
related cardiomyopathy has been extensively described223 to 5200 l/m2).231–235 It undergoes hepatic metabolism and
and usually occurs with cumulative doses greater than biliary excretion with only around 7% of drug found
500 to 550 mg/m2 in previously untreated patients. unchanged in the urine.231 This suggests that dose reduc-
These effects are more pronounced with conventional 3 tion in liver disease may be warranted while adjust-
weekly bolus dosing than if weekly low dose bolus dosing ment for renal impairment may not be of value.236 The
or 96-hour continuous low dose infusion is implemented. terminal half-life is long ranging from 8.9 to 189
Other factors that predispose to cardiac toxicity include hours.235,237,238 Mitoxantrone acts through DNA interca-
known cardiac disease, prior mediastinal or cardiac irra- lation, DNA–protein cross-linkage and DNA–DNA
diation, age greater than 70 years, and chronic hyperten- linkage, and topoisomerase II inhibition.236
sion. In addition to cardiomyopathy, other cardiac The major acute toxicity of mitoxantrone is myelo-
effects have been recorded, including nonspecific EKG suppression, especially neutropenia, while nausea and
changes, occasional rhythm disturbances, an uncommon vomiting tends to be mild. Cardiac toxicity occurs with
myocarditis-pericarditis syndrome, and rarely sudden mitoxantrone but is less common than with the anthra-
death. It has been suggested that epirubicin may be less cyclines. Alopecia secondary to mitoxantrone administra-
cardiotoxic than doxorubicin, although the contrary view tion is normally mild.239 Mitoxantrone has significant
has been expressed that equiactive doses of the two activity in PC but not other genitourinary cancers.240,241
agents are equitoxic.224 It has been licensed by the FDA for use in symptomatic
Of importance, doxorubicin is a radiosensitizer and HRPC based on the results of a randomized trial con-
has a radiomimetic function—thus it will cause recall ducted by National Cancer Institute of Canada showing
reactions in areas previously treated with radiotherapy.225 major improvement in quality of life.242–244 The potential
Other toxic effects include nausea, vomiting, stomatitis, role of mitoxantrone in earlier stage disease is under
and alopecia. intense investigation245,246 and is the subject of a ran-
The clinical indications for the use of this group of domized trial being carried out by the Southwest
agents against the genitourinary cancers have been Oncology Group.
defined on the basis of the activity of doxorubicin. At
present, it is still not clear whether the analogs, epiru- Drugs That Act Through Tubulin Modulation
bicin and idarubicin, offer any clear advantage in this Vinca Alkaloids
context. Doxorubicin has been identified as one of the
agents of choice for use against superficial and advanced The vinca alkaloids, which occur naturally in the peri-
bladder cancer and has significant activity against adeno- winkle (Catharanthus roseus), are formed from two multiple-
carcinoma and small cell carcinoma of prostate. In previ- ring planar units, an indole nucleus and a dihydroindole
ous experience, in particular at the University of Indiana nucleus. The two most commonly used drugs, vincristine
and at the Memorial Sloan Kettering Cancer Center in and vinblastine, are almost identical, differing only in a
New York, doxorubicin was built into combination single substitution on the dihydroindole nucleus.247
chemotherapy regimens for germ cell tumors but has They both act by binding to tubulin and inhibit micro-
been deleted in more recent trials, based on randomized tubule assembly, which in turn inhibits mitotic spindle
trials in Indiana in which it appeared not to add to the formation. This causes an accumulation of cells in
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 65

metaphase. Although the vinca alkaloids are thought to relatively mild.256 An initial study of the pharmacokinet-
be cell cycle phase specific for mitosis, the cytotoxic ics by Bore et al.,257using tritiated vinorelbine and also a
effect probably occurs in S phase and its effect seen in the radioimmunoassay, showed a low urinary excretion based
M phase. After intravenous administration, the vinca on elimination. The primary excretion route was the
alkaloids are extensively bound by serum proteins and feces, with 34% to 58.4% of the total dose being excreted
blood components and are rapidly cleared from the by this route over a period of 21 days.257More recently,
plasma and concentrated in various tissues. The disposi- Rowinsky et al.258 published the results of a bioavailabil-
tion is triphasic,90,248 with a rapid initial half-life (t1/2α ity study in which the drug was given intravenously and
less than 5 minutes), t1/2β of 1 to 2 hours, and t1/2γ of 1 to orally to the same patients. Bioavailability was 27% ± 14%.
3 days. They approximate total body water in their dis- The volume of distribution was large, 20.02 ± 8.55 l/kg,
tribution. The major route of excretion is hepatic,249 with and the terminal phase half-life was 18 hours. Plasma
appreciable amounts appearing in the stool, and liver dis- decay was triphasic.258
ease may necessitate a change in dosage. A small compo- Vinorelbine has activity in HRPC259 but has not pro-
nent of excretion is urinary. duced impressive results in limited trials in patients with
Although similar in structure, the vinca alkaloids have testicular germ cell tumors, renal cell carcinoma, or
substantially different profiles of toxicity. Vincristine is urothelial cancer.204,260 The activity of vinorelbine in PC
predominantly associated with neurologic side effects, is apparently increased by the concurrent administration
including a range of peripheral sensorimotor neuropathic of estramustine but with the consequent side effects
changes, autonomic neuropathy, jaw pain, and central attributable to the estrogenic effects of that drug.261–263
nervous system effects. Less commonly vincristine causes The potential of this combination relative to other
myelosuppression, gastrointestinal effects, alopecia, and chemotherapeutic approaches in this setting remains to
the syndrome of inappropriate ADH production. By con- be determined.
trast, vinblastine is associated with predominant myelo-
suppression (in particular granulocytopenia), more
marked gastrointestinal toxicity (especially paralytic Taxanes
ileus), and alopecia; the neurologic toxicities, while quite A very important new class of drugs that is being exten-
prominent, occur less frequently and less severely than sively evaluated in a broad spectrum of tumor types is the
for vincristine. Both agents are significant vesicants. taxanes. The two FDA-licensed drugs in this class are
The vinca alkaloids have had particular application in paclitaxel and docetaxel; the structures are shown in
the treatment of advanced germ cell tumors,250,251 small Figure 4-3. Paclitaxel is extracted from the bark of the
cell cancer of the prostate, and in bladder cancer (vin- western yew tree264; and, initially, this severely limited
blastine, in particular)252 but appear to have less single the supply of the drug for investigation. Compounded by
agent activity in the treatment of prostate adenocarci- the low water solubility of the drug, this slowed the
noma, adrenal carcinoma, and renal carcinoma. development of the drug. However, these difficulties
As noted earlier, the vinca alkaloids have substantial were overcome because of the interest generated by the
clinical activity against the urologic malignancies, in par- high degree of activity of the drug in experimental sys-
ticular germ cell tumors and urothelial malignancy. For tems and because the drug was shown to have a unique
this reason, the development of new vinca alkaloids with mode of action.265–270 The drug acts on the microtubules,
a higher degree of experimental antitumor activity is of but unlike the vinca alkaloids, which also interact with
interest. A potentially promising new agent in this class is microtubules, the taxanes stabilize the microtubule. It
vinorelbine (Navelbine), which has been under investiga- has been shown that a microtubule is in dynamic equilib-
tion in Europe and has recently been introduced into the rium with the tubulin heterodimers, which make up its
United States. The compound is 5′-noranhydrovinblas- structure. These associate at one end and dissociate at the
tine. Although it shares the mode of action of the other other. The action of the vinca alkaloids is to prevent the
vinca alkaloids, discussed earlier, in experimental systems tubulin from binding and thus the microtubules undergo
it has less affinity for axonal microtubules compared with spontaneous disassembly. The action of the taxanes is the
its affinity for the microtubules of the mitotic spindle. opposite. The microtubule is stabilized, so that growth
This indicates the possibility that it might produce inter- continues by association of tubulin but disassembly does
ruption of mitosis at concentrations that would not give not occur. Thus the cell becomes filled with a tangle of
rise to neurotoxicity.253 It has also shown a higher degree elongated but functionally useless microtubules, leading
of activity than vincristine or vinblastine against some to cell death.
experimental tumors.254,255 In phase I testing, neutrope- Paclitaxel has undergone very extensive testing and is
nia was the dose-limiting toxicity, and the maximam tol- approved for use in ovarian carcinoma271 and lung can-
erated dose (MTD) was 35 mg/m2/week. It did give rise cer. In phase I studies, it was initially given by short infu-
to peripheral neuropathy at higher doses, but this was sion.272,273 However, acute allergic reactions and
66 Part I Principles of Urologic Oncology

R C O
O
O O R
R OH
R
C NH C CH C O
R
H OH
HO
O
O
R=CH3 O
C
C R
1 O
O
HO H O OH
H
CH3
H OH H3C
O CH3
H
CH3
CH3 NH H O H
H3C HO H O

H3C O O O O H O
O

CH3

Figure 4-3 Structures of paclitaxel (1), docetaxel (2).

disturbances of cardiac rhythm led to progressive length- drug is currently under evaluation. These developments
ening of the infusion time to 24 hours, which became the are important in enabling the drug to be given on an out-
standard for administration of the drug. When given by patient basis. The pharmacokinetics of paclitaxel were
24-hour infusion, the MTD was 200 to 275 mg/m2 given evaluated during the early clinical studies using high per-
every 3 weeks. The dose-limiting toxicity was neutrope- formance liquid chromatography (HPLC) assay for the
nia, which was often profound but short lived and which drug. Both a biphasic and a triphasic plasma decay have
relatively uncommonly led to toxic death.274 Other toxi- been described, with the terminal phase half-life being in
cities, as noted above, included acute cardiac effects the range of approximately 4 to 7 hours for the biphasic
(mainly arrhythmias), hypersensitivity reactions, peripheral plasma decay and in the range of approximately 10 to 50
neuropathy, and gastrointestinal toxicity. In a phase I hours where a gamma phase was described.280 The earlier
study in leukemia,275 in which grade IV myelosuppres- studies reported linear pharmacokinetics, but recent
sion was accepted as routine, mucositis was the dose-lim- reports have indicated nonlinear pharmacokinetics with
iting toxicity at a dose of 390 mg/m2. Total alopecia was shorter infusions. Volumes of distribution have been
another feature, and it involved the hair on the head, variable in the reported studies and have been generally
body hair, and eyelashes. It was subsequently recognized in the range of 50 to 100 l/m2; however, very large vol-
that paclitaxel-related cardiac arrhythmias276 (and in par- umes of distribution have been described in the study
ticular bradycardia, the most common of the paclitaxel- reported by Huizing et al.281 Very little of the drug is
related arrhythmias) are clinically less dangerous than eliminated unchanged through the kidneys and systemic
initially feared. With suitable premedication with clearance appears to result from metabolism, biliary
steroids, H1 and H2 histamine antagonists, the drug can excretion, and binding to tissue components. The main
be safely given by a 3-hour infusion.277 This markedly route of metabolism is by cytochrome P450-dependent
reduces the myelosuppression without apparently reduc- hydroxylation, the principal hydroxylated metabolite in
ing the antitumor effect.278,279 One-hour infusion of the bile being 6-hydroxy paclitaxel.282 Of interest, cisplatin
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 67

given first in combination with paclitaxel is more toxic therefore best considered a combination hormonal-
than the reverse sequence. This appears to be related to cytotoxic therapy. However, the mechanism by which
the fact that cisplatin inhibits the metabolism of paclitaxel, estramustine exerts its antineoplastic effect is unclear.
causing an increase in the area under concentration × Following oral ingestion estramustine phosphate is rap-
time curve (AUC) of the latter drug.283 For the same idly dephosphorylated and absorbed with a bioavailabil-
schedule of administration, the degree of myelosuppres- ity of 37% to 75%.301 Concurrent ingestion of calcium
sion is related to the total drug exposure as measured by rich foods, such as dairy products, can interfere with
the AUC. A shorter infusion gives less toxicity for the estramustine absorption.302 After absorption estramus-
same AUC than a longer infusion, suggesting that dura- tine is metabolized to estromustine, which is preferen-
tion of exposure to a minimum cytotoxic concentration tially taken up by and retained in prostate tissue and PC
may be an important factor in the toxicity of the drug.284 cells by estramustine-binding protein.303 Estramustine is
Paclitaxel has shown antitumor activity in early studies of metabolized in the liver and excreted in the bile with very
germ cell tumors and transitional cell carcinoma of the minimal renal excretion.301 The terminal half-life of
urothelium. Patients with germ cell tumors, with prior estromustine is 10 to 20 hours.301
therapy, showed a 24% response rate to paclitaxel As a single agent, estramustine has not demonstrated
250 mg/m2 24-hour infusion with 2 CR and 4 PR in 25 benefit over continued or alternate hormonal therapy in
patients.285 Previously untreated patients with transi- PC.304 However, in combination with selected cytotoxic
tional cell carcinoma of the urothelium received the same agents, the current clinical wisdom is that estramustine
dose and schedule of paclitaxel and showed a 42% appears to contribute to increased response,259,305–307
response rate.286 Studies in renal cell carcinoma have although this has not been proven in well-structured tri-
been negative.287 Early studies with paclitaxel in HRPC als. While the mechanism for this is unclear, the effect of
show modest single agent activity,288 and subsequent tri- estramustine may occur through a phase activation of PC
als have examined the potential of adding other agents to cells so that they are more sensitive to subsequent or con-
the taxanes including estramustine and carboplatin (see current cytotoxic effect. Estramustine alters cellular
later).185,289 microtubular configuration and may have synergy with
Docetaxel (taxotere, Figure 4-2) is a synthetic analog other drugs that act on microtubules such as taxanes
of paclitaxel that is prepared from 10 diacetyl baccatin (paclitaxel, docetaxel) and vinca alkaloids (vincristine,
III, a compound extracted from the needles of the vinblastine, and possibly vinorelbine).305,308–310 Recent
European yew tree, Taxus baccata. It is thus derived from reports from phase I and phase II trials suggest that the
a biologically renewable source. It is more potent in vitro combination of estramustine and docetaxel is well toler-
than paclitaxel.290 Although its mode of action is similar, ated and produces a decrease of >50% in serum PSA in
it is reportedly active against cells, which are resistant to around 50% of HRPC cases treated.296,297,311–315 Given
paclitaxel.290 It has undergone phase I testing in 1-hour, significant estrogenic side effects related to estramustine
2-hour, 6-hour, and 24-hour infusions291–293 and as a 1- therapy, particularly thromboembolic phenomena, its
hour infusion daily for 5 days.294 The MTD ranged from place in combination therapy with cytotoxic agents for
70 to 115 mg/m2, and neutropenia was dose limiting in HRPC still requires proof of efficacy over the cytotoxic
all of the studies, although mucositis was also a major agents alone, as well as the optimization of dose and
toxicity in the study of the 24-hour infusion.293 It has scheduling to minimize toxicity.
been suggested that anaphylactic reactions are less com-
mon with docetaxel than with paclitaxel but that skin tox-
Alkylating Agents
icity may be more marked. Plasma decay is triphasic with
a gamma phase t1/2γ of 11.8 ± 6.7 hours.292 Docetaxel has The alkylating agents are a group of chemical com-
activity in HRPC,295 where it is currently most com- pounds of diverse structure, which share the common
monly combined with estramustine,296–298 urothelial can- property of labile, electrophilic alkyl groups that can
cer,299 penile cancer, and testis cancer. It has limited react with most biologic molecules to form adducts. The
activity in renal cell carcinoma.300 At present, it is diffi- alkyl groups can be added to oxygen, nitrogen, phospho-
cult to be dogmatic regarding the respective merits of rus, or sulfur atoms, and thus can function at extremely
paclitaxel and docetaxel with respect to antitumor effi- diverse sites. However their most important reactions are
cacy, but it is quite clear that they have different spectra with the nitrogen atoms of DNA, particularly the N7
of toxicity. position of guanine residues, altering the structure or
function of the DNA. These drugs are all cell cycle
dependent, but not cell cycle specific; that is, they exert
Estramustine
their effects on cells throughout the cell cycle (analogous
Estramustine is constituted by the carbamate linkage of to radiation). They appear most active against rapidly
estradiol and nor-nitrogen mustard molecules and is proliferating cells. Agents included in this group include
68 Part I Principles of Urologic Oncology

cyclophosphamide, ifosfamide, thiotepa, melphalan, which is particularly irritating to the bladder mucosa, can
nitrogen mustard, busulfan, chlorambucil, and the cause hemorrhagic cystitis, and this can lead to bladder
nitrosoureas. fibrosis or even transitional cell carcinoma of the bladder.
The alkylating agents, through their common mecha- In the context of intravenous administration, this toxic
nism of action, are potentially cytotoxic and carcino- effect can be avoided by the use of sodium-2-mercap-
genic. Although sharing common functional traits, toethane sulfonate (MESNA), which is excreted in the
differences in their chemical structures account for the urine, providing reactive thiols that bind to the acrolein,
variation in their pharmacologic characteristics. This is protecting the bladder mucosa. Occasionally, cyclophos-
of particular importance as an explanation for why cross- phamide may cause interstitial pneumonitis, gonadal
resistance may not occur in all situations. atrophy, anaphylaxis, and in higher doses, the syndrome
of inappropriate ADH production and cardiotoxicity
(including acute cardiac necrosis when given in trans-
Cyclophosphamide
plant-intense dosage).
Cyclophosphamide is 2-bis-(2-chloroethyl)amino- The most common application of cyclophosphamide
tetrahydro-2H-1,3,2-oxazaphosphorine-2-oxide mono- for genitourinary cancer is in the treatment of adenocar-
hydrate, a cyclic phosphamide ester of nor-nitrogen cinoma of the prostate,64,65 and it has also been used
mustard. The monohydrate is not ionized and is lipid sol- in the past in the treatment of advanced germ
uble. This agent is a bifunctional substituted nitrogen cell tumors.250,319 It is less frequently used for germ cell
mustard, which must be activated in the liver before it is tumors in current practice because of the risks of car-
active. Its activation is a multistep process that occurs in cinogenicity and infertility. The drug is inactive in the
the hepatic microsomal P450 enzyme system.83,316 There treatment of renal and adrenal carcinomas and has only
is thus no rationale to using the drug for intraarterial limited activity in the treatment of bladder cancer.
chemotherapy nor as an intravesically administered
agent. It can be usefully administered as an oral agent
Ifosfamide
(90% bioavailability) or intravenously. Hepatic inactiva-
tion is the major mechanism of active drug elimination, Ifosfamide is a structural analog of cyclophosphamide,
whereas after intravenous administration about 15% of differing only in the position of one of the two
the drug is excreted unchanged in the urine and the rest chloroethyl groups. It is also a metabolically activated
as metabolites. The plasma half-life is approximately 5 to alkylating agent and must first undergo hydroxylation by
6 hours.316 hepatic microsomes.316 However, the change in its struc-
As cyclophosphamide is bifunctional, it can cross-link ture has resulted in changed pharmacology, and its acti-
the two strands of DNA, yielding an interstrand cross- vation within the liver occurs more slowly than for
link, or can produced intrastrand cross-links, or even can cyclophosphamide.
bind DNA to protein. The binding of the active metabo- Ifosfamide is well absorbed orally and can also be
lite of cyclophosphamide to DNA does not cause cell administered intravenously. Its pharmacology is similar
death per se; rather the cells progress slowly through the to that of cyclophosphamide. Its plasma half-life has been
S phase and arrest and subsequently die in the G2 phase. reported to be as short as 5 to 6 hours, either after oral or
Information regarding potential drug interactions intravenous administration,82,320 although Creaven et
with cyclophosphamide is relatively scant. As it must be al.81 documented a plasma half-life of radioactively
metabolized by hepatic microsomes to be active, drugs, labeled ifosfamide of nearly 14 hours. In current clinical
which induce this system (such as the barbiturates, practice, it is most commonly administered intra-
phenytoin, and carbamazepine) may increase the conver- venously, with schedules varying from a single infusion,
sion of cyclophosphamide to its metabolites; similarly to multiple-day schedules, with MESNA coverage to
cyclophosphamide may have an impact on the activity of prevent hemorrhagic cystitis. Creaven et al.81 have sug-
the barbiturates. It has also been reported that cimetidine gested that the alkylating activity ratio of ifosfamide is
increases the toxicity of cyclophosphamide via a change 1:5, when compared to cyclophosphamide. The pattern
in the concentration × time relationship of its active of toxicity is similar to that of cyclophosphamide,81 but
metabolites.317,318 with less myelosuppression and a greater tendency to
Cyclophosphamide produces significant leukopenia cause cystitis. In addition, ifosfamide has a greater preva-
and immunosuppression but is not usually associated lence of central nervous system toxicity, including altered
with thrombocytopenia.83,89 Nausea and vomiting may mental status, cerebellar dysfunction, seizures, and
occur, although usually in association with high dose extrapyramidal effects.81,321,322 It is not really clear
intravenous usage. Similarly alopecia is more commonly whether there is a definite dose-response relationship for
associated with high dose administration. Of particular ifosfamide, and thus an optimal dose has not been
importance, excretion of acrolein, one of the metabolites, defined.
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 69

Ifosfamide is currently used in salvage and high-risk O O


regimens for advanced germ cell tumors323–235 and for O
metastatic urothelial cancer.326–328 In broad-based phase O
I and phase II trials, little activity has been reported in
O
PC, although the drug has not been assessed since PSA
has been introduced as a surrogate marker of response. O
H
Ifosfamide is inactive in renal carcinoma, but single agent O
activity has been identified in squamous cell carcinoma, O
including cancer of the penis.
O
O
Thiotepa H
N,N′,N′′-triethylenethiophosphoramide (thiotepa) is an
aziridine drug, a polyfunctional alkylating agent, which
can produce interstrand cross-links, DNA adducts and O O
strand breaks, and a range of other DNA lesions.329 After
O
IV injection the t1/2α was 7.7 minutes and the t1/2β 25
minutes in the study by Cohen et al.330 Figure 4-4 Structure of etoposide.
Thiotepa is administered by intravenous or intracavi-
tary routes, the latter including intraperitoneal, intrathe-
cal, and intravesical delivery. It is not a vesicant and does Each compound has a complex structure composed of
not cause local soft tissue reactions. a multiringed structure linked to a glucopyranoside sugar
In the standard dose range, thiotepa is associated with (see etoposide structure in Figure 4-4). Both drugs are
myelosuppression, both granulocytopenia and thrombo- routinely administered intravenously, although etoposide
cytopenia, as well as nausea, vomiting, headache, and is now formulated for oral administration. The optimal
occasionally alopecia. In transplant-intense doses, schedule of administration has not been defined,
thiotepa may cause mucositis, cutaneous changes, and although prolonged schedules (multiple daily short infu-
occasional organic brain syndrome.331 When adminis- sions or continuous infusion) are most commonly
tered intravesically, thiotepa causes little systemic toxicity employed because of the phase-specific mode of action.
unless used within a few days of extensive transurethral The pharmacokinetics is biphasic, with half-life values of
resection; thiotepa, a relatively small molecule, is 90 minutes and 3 to 11 hours.334 After administration
absorbed significantly through an extensively denuded of radiolabeled etoposide to humans, 40% to 90% of
bladder surface. radioactivity is recovered in the urine within 48 hours,
The major clinical application for thiotepa has been in fecal recovery is less than 20%, and biliary excretion is
the intravesical chemotherapy of superficial bladder can- only minimal.332.
cer,67 although this agent is now used less frequently The major toxicity of the epipodophyllotoxins is dose-
because of its risk of carcinogenicity and the develop- related myelosuppression, with predominant leukope-
ment of more effective treatment options. nia.333 Gastrointestinal complications, such as nausea,
vomiting, and anorexia are usually mild. Other side
effects include alopecia, headache, fever, and hypoten-
Epipodophyllotoxins
sion. Severe hypotension may occur if the drug is infused
Podophyllotoxin, a derivative of the mandrake root, has too rapidly, and this can occasionally be accompanied by
been known to have antimitotic properties for more than bronchospasm or rarely by anaphylaxis. It is believed that
half a century, and extracts of the mandrake root have been these allergic phenomena may be due to the use of the
used for medicinal purposes for centuries.332 Although the diluent, cremophor. Etoposide phosphate, a water-
early podophyllotoxin derivatives were excessively toxic, soluble derivative that is rapidly hydrolyzed to etoposide
two derivatives (etoposide and teniposide) have shown in the plasma, is currently undergoing clinical trial.
substantial clinical activity with a tolerable profile of side Recently there has been emerging information that
effects.332–334 Although initially believed to act by binding etoposide is occasionally associated with iatrogenic acute
to tubulin and inhibiting microtubule assembly, additional myeloid leukemia, and such cases have now been
studies have suggested that the epipodophyllotoxins arrest recorded among patients cured of germ cell tumors.180
cells in late S phase or early G2 phase,335 rather than at G2- The indications for the use of the epipodophyllotoxins
M. More recently, these agents have been shown to exert for genitourinary cancer are relatively limited, with the
their anticancer effects by impeding the function of the most common application being in the management of
topoisomerase II enzyme.336 advanced germ cell tumors.250,251 Most studies have sug-
70 Part I Principles of Urologic Oncology

gested that there is only very limited activity against Methotrexate may be given by oral, intramuscular,
bladder cancer and prostatic adenocarcinoma, although intravenous, intraarterial or intrathecal routes. An optimal
there is a clear role for this drug in the treatment of small dosing route and schedule has not been defined. A broad
cell anaplastic PC. Etoposide is inactive in the treatment range of parenteral dosing has been reported, with doses as
of renal carcinoma, and only scant data are available with varied as in the range between 50 and 15,000 mg/m2,
respect to penile and adrenal cancers. predicated on the ability to rescue normal tissues with
calcium leucovorin. Caution must be exercised as the
higher dose range is potentially lethal if sufficient leu-
Antimetabolites
covorin is not administered. Alkalinization (as measured
Antifolates
by assessment of the urine pH), by increasing the solu-
The role of the antimetabolites in cancer treatment was bility of the drug and of the 7-hydroxy metabolite will
first explored 50 years ago with the investigation of also reduce toxicity. Methotrexate levels must be moni-
aminopterin, an analog of folic acid. This early work gave tored after high dose treatment to determine the length
rise to the development of methotrexate, the 4-amino, of leucovorin rescue.
10-methyl analog of the parent compound, which has More than 50% of the drug is bound to plasma pro-
come to be one of the most widely used agents for the teins. Methotrexate is widely distributed in the body. In
genitourinary cancers. standard doses, methotrexate is excreted unchanged in
Methotrexate inhibits dihydrofolate reductase urine, whereas in high doses some metabolism of the
(DHFR), an important enzyme in folic acid metabolism, drug occurs. Plasma decay of methotrexate is either
which catalyzes the reduction of dihyrofolate to biphasic or triphasic,341–343 with the terminal phase of
tetrahydrofolate. DHFR maintains the intracellular excretion having been reported to be in the range of 10
reduced folate pool, which in turn is required for the to 26 hours. Methotrexate is highly schedule dependent;
synthesis of thymidine and purines and thus for the pro- its toxicity is more a function of the time during which
duction of DNA, RNA, and protein. Methotrexate plasma levels are maintained above a minimum cytotoxic
undergoes polyglutamation intracellularly to varying concentration than of the total AUC. Thus any factor
extents; the polyglutamated forms do not traverse cellu- that can lead to a prolonged low level of drug will greatly
lar membranes. Polyglutamation occurs to a greater increase its toxicity. Principal among such factors are
extent in tumor cells than in normal cells, and this may impaired renal function and localization in “third space”
explain the selective action of the drug. The polygluta- compartments such as pleural effusion or ascites with a
mated form is retained preferentially within cells, some- slow release into the circulation. In these situations,
times for very lengthy periods337 and can directly methotrexate must be used with caution; rescue with leu-
inhibit other folate-dependent enzymes, including covorin employed as necessary.
thymidylate synthase.338 The spectrum of toxicity of methotrexate is a function
Methotrexate may also cause single- and double- of age, dose, the use of calcium leucovorin, drug metabo-
stranded breaks in DNA.339 It is most active against rap- lism, drug interactions, and renal function.340,344 Patients
idly proliferating cells and appears to exert its major may experience no toxic effects at all. Hematologic
effect during S phase; and is thus classified as a cell cycle side effects include leukopenia, thrombocytopenia, and
phase-specific antimetabolite. anemia. Gastrointestinal toxicity includes nausea, vomit-
Methotrexate uses the same active transport mecha- ing, diarrhea, and stomatitis. Hepatotoxicity has been
nisms to enter cells as does folic acid. At least two such reported in patients receiving chronic low dose oral ther-
mechanisms have been identified, including a low-affinity apy and in high dose parenteral administration.
carrier that transports methotrexate and reduced folates Occasionally, methotrexate can cause self-limited pneu-
and a high-affinity system, which is more avid for monitis. One of the most dangerous toxic effects is the
reduced folates than for methotrexate. The metabolic induction of renal failure, as adequate renal function is
block induced by methotrexate can be circumvented by necessary to ensure satisfactory excretion of the drug.
the use of calcium leucovorin, which feeds into the folic Methotrexate can cause skin rash, pruritus, urticaria,
acid cycle beyond the block of DHFR. Calcium leucov- alopecia, and a range of other cutaneous side effects. Less
orin and its metabolite, 5-methyltetrahydrofolate, share commonly, central nervous toxicity can occur, especially
the latter common transport mechanism with methotrex- after intrathecal administration.
ate. It appears that normal cells can be selectively rescued
by calcium leucovorin, either because of differences in
Fluoropyrimidines
transport or because of differences in the rate of DNA
synthesis between normal and cancer cells. The complex 5-Fluorouracil (5-FU) was synthesized to act as a false
biochemistry of methotrexate, its reversal and mecha- pyrimidine and thus to inhibit the formation of thymi-
nisms of resistance, is beyond the scope of this chapter dine.345 There are several proposed mechanisms of
but has been detailed elsewhere.340 action, including inhibition of thymidylate synthase by an
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 71

active metabolite (FdUMP), incorporation of the triphos- inhibits DHD, decreases the first-pass effect and enables
phate 5-FUTP into RNA, and incorporation of the 2′ 5-FU to be given orally.
deoxy triphosphate 5-FdUTP into DNA.346 The pres- Capecitabine is an orally bioavailable prodrug of
ence of reduced folate is critical to the function of 5-FU 5-FU.359 It is generally administered on a twice-daily basis
in inhibiting thymidylate synthase. Gastrointestinal either continuously or with a week-off therapy every 3 or
tumors with increased expression of thymidylate synthase, 4 weeks. After absorption capecitabine is metabolized by
with or without increased levels of the metabolizing hepatic carboxylesterase to 5′-deoxy-5-fluorocytidine
enzymes, thymidine phosphorylase, and dihydropyrimi- and then converted to 5′-deoxy-5-fluorouridine by cyti-
dine dehydrogenase, tend to be resistant to fluoropyrim- dine deaminase, which is also present in the liver.76
idines.347 Testing for expression levels in individual 5′-Deoxy-5-fluorouridine then enters cells and is metab-
tumors may identify genitourinary cancers with suscepti- olized by thymidine phosphorylase to 5-FU. The result is
bility to this group of drugs, but further testing of this a cellular 5-FU concentration that exceeds that achieved
hypothesis is required.348 in the serum and surrounding normal tissue.76
5-FU can be administered by oral, intravenous, Capecitabine has been evaluated as a potential alternative
intraarterial, or intraperitoneal routes. Bioavailability is to 5-FU infusion in a number of cancer types.360.361 It has
poor and erratic when the drug is given by mouth,349 and modest activity in renal cell carcinoma,362,363 but is yet to
this route of administration is no longer used (however, be fully evaluated in other genitourinary cancers.
see following discussion on modulation and capecitabine). The primary toxicities of the fluoropyrimidines are
When given intravenously the drug has an extremely gastrointestinal toxicity, stomatitis and diarrhea, and
short terminal phase half-life measured in minutes.73,350 myelosuppression. The pattern of toxicity varies with the
This is due to its rapid degradation by dihydrouracil schedule of administration and is also influenced by the
dehydrogenase (DHD) to 5,6-dihydro 5-FU that then presence of modulators. Weekly bolus 5-FU produces
undergoes ring rupture and is degraded to small mole- mainly myelosuppression (primarily leukopenia). Loading
cules.346 A rare deficiency of this enzyme can lead to dose 5-FU, prolonged infusions, and 5-FU used with leu-
severe toxicity following administration of 5-FU.346 covorin produce primarily gastrointestinal toxicity; cere-
Because of its mode of action, 5-FU is an S-phase spe- bellar toxicity and cardiotoxicity have been described364
cific drug and this combined with its extremely short half- but are uncommon at standard doses. Capecitabine may
life would lead one to anticipate that it would be highly produce tenderness and desquamation of the hands and
schedule dependent. However, paradoxically, rapid infu- feet (the so-called hand-foot syndrome), in addition to the
sions of 5-FU are more toxic than the same dose given over side effects seen with other fluoropyrimidines.
a period of several hours. This is probably due to the fact,
although that the drug itself has an extremely short plasma
Gemcitabine
half-life, the persistence of the active metabolite FdUMP
intracellularly is measured in days.351 The bolus injection of A new pyrimidine antimetabolite, 2′deoxy-2′difluoro-
the drug probably temporarily exceeds the capacity of cytidine (gemcitabine), has recently been introduced into
DHD, thus making drug available for conversion to active clinical practice for urothelial and a range of other
metabolites. The nucleoside derivative of 5-FU, 5-fluoro- tumors. In preclinical systems gemcitabine showed very
2′-deoxyuridine (FUdR, floxuridine) is extremely schedule significant activity against experimental solid tumors365
dependent. The dose that can be given by short-term infu- and human tumor xenografts.75 Its structure is shown in
sion is approximately three orders of magnitude greater Figure 4-5. Like cytosine arabinoside, an antimetabolite
than the dose that can be given when the drug is given by a which it resembles structurally, it is activated intracellu-
long-term continuous infusion.352 larly to the triphosphate, 2′deoxy-2′difluoro-cytidine
A number of compounds have been used in conjunction 5′-triphosphate (dFdCTP), and in this form is incorpo-
with fluoropyrimidines to enhance their activity through rated into DNA.366–368 Gemcitabine entry to cells
biochemical modulation. These include interferon, PALA requires the presence of the nucleoside transporter sys-
(N-phosphonoacetyl-L-aspartate), and leucovorin.353 Of tem, with cells deficient in this transporter being gemc-
these, leucovorin has been the most extensively investi- itabine resistant.366 The presence of dFdCTP within the
gated and has proven to be the most clinically useful by cells inhibits its own degradation via deamination by
raising the response rate to 5-FU in colorectal cancer by a deoxycytidine deaminase and promotes 2′deoxy-2′diflu-
factor of about three.354 It acts by leading to stabilization oro-cytidine phosphorylation by deoxycytidine kinase
of the ternary complex formed between the active metabo- (dCK), at least in part by ribonucleotide reductase (RR)
lite FdUMP, the active site of the enzyme thymidylate syn- inhibition. Forced overexpression of cytidine deami-
thase, and a reduced folate cofactor (5, 10 methylene nase,369 increased expression of RR370 and decreased
tetrahydrofolate), which is derived from leucovorin.355 An expression of dCK371,372 are associated with decreased
investigational modulator eniluracil has been evaluated in sensitivity to gemcitabine in cell line systems. The anti-
a clinical trial setting.356–358 This compound, which neoplastic effect of gemcitabine derives, in part, from
72 Part I Principles of Urologic Oncology

NH2 can act.378–381 The net result of faster infusions is that the
maximal 2′,2′-difluorodeoxycytidine triphosphate con-
centration is achieved for a shorter time with the addi-
N tional amount of drug wasted therapeutically but
potentially contributory to side effects. On this basis and
N given that fixed dose rate infusion at 10 mg/m2/minute
does not produce more toxicity than more rapid infusion,
HOCH2 administration of gemcitabine in this manner is standard
O practice.
Antitumor activity for gemcitabine against bladder
F cancer was noted in the phase I study by Pollera et al.382
in 14 patients receiving gemcitabine at doses greater than
875 mg/m2, they observed 1 CR and 2 PR. A response
OH F
rate of 28% was observed in a phase II trial in previously
Figure 4-5 Structure of gemcitabine. untreated patients with bladder cancer.383 Subsequently,
the combination of gemcitabine and cisplatin was
assessed in a phase II trial with a response rate of 41%.384
dFdCTP incorporation into DNA instead of dCTP This combination was then compared to MVAC in a ran-
by polymerases involved in repair.373 However, RR is an domized phase III trial and found to produce equivalent
S-phase specific, potentially rate-limiting enzyme for response and survival with better quality of life.375 De
DNA synthesis. The activity of gemcitabine is cell cycle Mulder et al.385 noted response rate of 8.1% or 3 of 39
specific with blockade at the G1/S phase transition, per- patients who could be evaluated in a phase II study of
haps suggesting that RR inhibition is an important con- gemcitabine in renal cell carcinoma, with durable
tributor to gemcitabine antineoplastic effect. responses exceeding 12 months in 2 patients.
In the initial phase I clinical trial, the drug was given Subsequently, researchers at the University of Chicago
over a 30-minute infusion weekly for 3 weeks every 4 have reported on combination of gemcitabine with 5-FU
weeks. The maximum tolerated dose was 790 mg/m2 in RCC with possible improved survival over historical
with myelosuppression, predominantly thrombocytope- controls and response rate of 17%.386 This study is cur-
nia and anemia, being the dose-limiting toxicity.374,375 rently being replicated with dose-scheduling variation
Other toxicities have been relatively mild. However, sub- and the incorporation of capecitabine instead of 5-FU.
sequent studies have shown that considerably higher Phase II studies in PC are ongoing, but highly prelimi-
doses can be given safely, particularly to patients with lit- nary data suggest that gemcitabine may have some pallia-
tle or no prior therapy. In patients with nonsmall cell tive benefit in the absence of major falls in serum PSA
lung cancer and no prior therapy, O’Rourke et al.376 concentration,387 Gemcitabine has some activity in
found that 2500 mg/m2 given over 4 hours every 2 weeks chemo-refractory testicular cancer388,389 and this is being
was below the MTD. Initial pharmacokinetic studies pursued in further clinical trials.
showed a rapid elimination of the drug with a median
half-life of 8 minutes. The drug was rapidly converted to
the corresponding uracil metabolite 2′,2′-difluo- CAMPTOTHECINS
rodeoxyuridine, which had a longer half-life, with a An important group of drugs under active clinical devel-
median of 14 hours. The active metabolite 2′,2′-difluo- opment are analogs of camptothecin. Camptothecin was
rodeoxycytidine triphosphate was analyzed in circulating isolated from extracts of the Japanese tree Camptotheca
mononuclear cells. A peak was observed within 30 min- acuminata by Wall et al.390 and shown to be active in
utes of the end of the infusion and increased with dose up experimental leukemia and some solid tumors. The
to a dose of 350 mg/m2. Beyond this dose, there was no structure of the camptothecins incorporates five rings
increase in the active metabolite indicating saturation of (Figure 4-6). The fifth, or E, ring can exist in a closed
the activation to the triphosphate.366 Subsequent study ring lactone or an open ring hydroxy acid form. The
suggests that infusion of gemcitabine at a fixed dose rate closed ring form of camptothecin is highly water insolu-
of 10 mg/m2/minute produces an optimal cellular level of ble; however, the sodium salt of the open ring hydroxy
active metabolite.377,378 Infusion rates faster than this acid form is water soluble. It was in this form that
may result in a lower cellular exposure (area under the the compound was introduced into early clinical trials in
concentration curve concentration) of 2′,2′-difluo- the late 1960s. Phase I trials showed that the compound
rodeoxycytidine triphosphate by exceeding the rate at gave rise to substantial gastrointestinal toxicity and
which the nucleoside transporter system and/or which myelosuppression and also had a tendency to produce
the enzymes involved in producing this active metabolite hemorrhagic cystitis.391,392 It showed antitumor activity
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 73

R2 R3

R1 O
N

CH3CH2
OH O

R1 R2 R3
O

Irinotecan N N C O H CH3CH2

Topotecan OH (CH3)2NCH2 H

Figure 4-6 Structures of irinotecan (CPT-11), topotecan.

in the phase I studies, but a subsequent phase II study in


Irinotecan
gastrointestinal malignancies proved to be negative,393
and development of the compound was halted. Work Irinotecan is 7 ethyl 10[4-(1 pyridino)-1-pyridino] car-
continued in the laboratory on the mode of action of this bonoxy camptothecin. It is the most extensively evaluated
compound, which proved to be unique.394–398 The com- of the newer camptothecins. In phase I studies, the drug
pound combines with a cleavable complex formed showed major toxicities of leukopenia, nausea, and vom-
between DNA and the enzyme topoisomerase I, an iting and diarrhea.402 On a weekly schedule the recom-
enzyme important in relieving the torsion that develops mended dose for phase II studies was 100 mg/m2/week,403
as the strands of DNA unwind for replication and tran- and when intermittent doses were administered every
scription. The role of topoisomerase I is to cleave one of 3 weeks,404 the recommended dose was 240 mg/m2. With
the strands of the DNA, thus allowing the supercoiled intensive treatment of diarrhea with loperamide, dosing
DNA to unwind. The combination of camptothecin with up to 750 mg/m2 every 3 weeks has been reported.405,406
the cleavable complex prevents the resealing of the DNA A variety of other schedules have been reported, includ-
and thus causes single-strand DNA breaks.397,399 ing a single dose every 4 weeks, 5-day continuous infu-
Mutations in topoisomerase I mutations alter both DNA sion every 3 to 4 weeks, and daily 3× every 3 weeks.402
cleavage and unwinding, as well as the interaction with CPT-11 is virtually inactive in vitro. To be activated, it
camptothecin.400 The identification of this unique mode must be hydrolyzed to 7-ethyl-10-hydroxy camptothecin
of antitumor action stimulated the development of (SN38), which is 3 orders of magnitude more active than
analogs of camptothecin that would be more soluble and the parent drug.407 Consequently, pharmacokinetic stud-
more active. It was shown that the closed ring form of the ies of the drug require the measurement of the total and
E ring is essential for activity, being several orders of the lactone forms of both CPT-11 and SN38. No rela-
magnitude more active than the hydroxy acid form.401 tionship between nonmyeloid toxicity and any pharma-
This may account for the variable activity of camp- cokinetic parameter was found in the study of Rowinsky
tothecin, which as noted above, was given as a sodium et al.404 A relationship between AUC of total SN38 and
salt of the open ring hydroxy acid form. Several com- percent decrease in absolute neutrophil count (ANC) was
pounds were developed as analogs of camptothecin; to found using a sigmoidal Emax model.
date, the two most important are irinotecan (CPT-11) Irinotecan is still undergoing early phase trials in the
and topotecan. The structures of these are shown in genitourinary malignancies. The Southwest Oncology
Figure 4-6. Group is conducting a phase II trial for patients with
74 Part I Principles of Urologic Oncology

advanced bladder cancer, and other studies in PC are advance or at worst a further option in patients with this
ongoing. We are not aware of published data with respect stage of disease.
to the utility of this agent in renal and other genitouri- By contrast, randomized trials have not proved a role
nary cancers. for combination chemotherapy in renal carcinoma and
clinical practice in this areas is still in an early stage of
evolution. At present, one of the major investigational
Topotecan
emphases is the combination of conventional and novel
Topotecan is a semisynthetic water soluble camptothecin cytotoxic agents with biochemical and biologic modula-
analog, which in preclinical systems is active against tors that target the effectors of multidrug resistance, cel-
experimental tumors and against human xenografts. In lular regulation, and immunologic function. However,
early phase I trials, the drug was given daily for 5 days these studies are beyond the scope of this chapter on
every 3 or 4 weeks.408,409 On this schedule, the dose- principles and applications of conventional chemother-
limiting toxicity was neutropenia, and the recommended apy. Further clinical trials to evaluate new approaches are
dose for phase II studies was 1.25 to 1.5 mg/m2. required in the genitourinary cancers, and in particular
Subsequent studies have evaluated infusions of from 24 those tumors that fail to respond to conventional first-
hours up to 21 days.410–412 With the 24-hour infusion, a line therapy. In addition to the need for new treatment
dose of 1.5 mg/m2/week was recommended for phase II strategies, careful definition of endpoints and appropriate
evaluation. Thrombocytopenia was more marked than design of clinical trials is essential if outcomes are to
neutropenia in the 21-day infusion study (MTD of improve.
0.53–0.6 mg/m2/day in patients previously treated with
cytotoxic therapy and 0.8 mg/m2/day in chemotherapy
naïve patients). Of interest, granulocyte-colony stimulat- REFERENCES
ing factor (G-CSF) did not permit dose intensification in
one study because dosing was limited by fatigue and 1. Burchenal JH: The historical development of cancer
thrombocytopenia.409 In pharmacodynamic studies, dose chemotherapy. Semin Oncol 1977; 4:135–146.
but not the AUC of the topotecan lactone could be 2. Steel GG, Lamerton LF: Cell population kinetics and
related to mean percentage change in ANC by a sig- chemotherapy. I. The kinetics of tumor cell populations.
moidal Emax model. The dose required to produce a 50% Natl Cancer Inst Monogr 1969; 30:29–42.
3. Bassukas ID: Comparative Gompertzian analysis of
decrease in ANC was 0.86 mg/m2/day, given daily for 5
alterations of tumor growth patterns. Cancer Res 1994;
days.408 54:4385–4392.
Data on the activity of topotecan against genitouri- 4. Ferrante L, Bompadre S, Possati L, Leone L: Parameter
nary tumors is scant, but evaluation is ongoing. estimation in a Gompertzian stochastic model for tumor
growth. Biometrics 2000; 56:1076–1081.
5. Hanahan D, Folkman J: Patterns and emerging
SUMMARY
mechanisms of the angiogenic switch during
There is no current consensus on the optimal strategy for tumorigenesis. Cell 1996; 86:353–364.
the clinical use of chemotherapy against cancers of the 6. Hanahan D, Weinberg RA: The hallmarks of cancer. Cell
genitourinary tract. The most common approach has 2000; 100:57–70.
been to use combination regimens in preference to single 7. Shackney SE, McCormack GW, Cuchural GJ, Jr: Growth
rate patterns of solid tumors and their relation to
agents, predicated largely on the success of combination
responsiveness to therapy: an analytical review. Ann Intern
chemotherapy for a variety of other solid tumors. There Med 1978; 89:107–121.
is no doubt that the introduction of the combination of 8. Nurse P: A long twentieth century of the cell cycle and
cisplatin, etoposide, and bleomycin revolutionized the beyond. Cell 2000; 100:71–78.
management of advanced germ cell tumors, providing a 9. Nurse P: Cyclin dependent kinases and cell cycle control
cure rate of up to 90%, compared to a cure rate of less (Nobel lecture). Chembiochem 2002; 3:596–603.
than 40% with single agent treatment. Similarly, after 10. Weinberg RA: The retinoblastoma protein and cell cycle
several years of clinical development of combination reg- control. Cell 1995; 81:323–330.
imens for bladder cancer, a seminal randomized trial 11. Sherr CJ: The Pezcoller lecture: cancer cell cycles
revealed a survival benefit from the combination of revisited. Cancer Res 2000; 60:3689–3695.
MVAC compared to cisplatin alone in this context. A fur- 12. Ishida S, Huang E, Zuzan H, et al: Role for E2F in
control of both DNA replication and mitotic functions as
ther landmark trial demonstrated that the combination of
revealed from DNA microarray analysis. Mol Cell Biol
gemcitabine and cisplatin was equivalent in efficacy to 2001; 21:4684–4699.
MVAC but less toxic. Mitoxantrone has an established 13. Wu L, Timmers C, Maiti B, et al: The E2F1-3
role in improving symptoms and quality of life in HRPC, transcription factors are essential for cellular proliferation.
while taxane-based combinations may provide a further Nature 2001; 414:457–462.
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 75

14. Massague J, Blain SW, Lo RS: TGFbeta signaling in 31. Giannakakou P, Gussio R, Nogales E, et al: A common
growth control, cancer, and heritable disorders. Cell 2000; pharmacophore for epothilone and taxanes: molecular
103:295–309. basis for drug resistance conferred by tubulin mutations in
15. Reynisdottir I, Massague, J: The subcellular locations of human cancer cells. Proc Natl Acad Sci U S A 2000;
p15(Ink4b) and p27(Kip1) coordinate their inhibitory 97:2904–2909.
interactions with cdk4 and cdk2. Genes Dev 1997; 32. Jiang H Yang LY: Cell cycle checkpoint abrogator UCN-
11:492–503. 01 inhibits DNA repair: association with attenuation of
16. Cordon-Cardo C: Mutations of cell cycle regulators. the interaction of XPA and ERCC1 nucleotide excision
Biological and clinical implications for human neoplasia. repair proteins. Cancer Res 1999; 59:4529–4534.
Am J Pathol 1995; 147:545–560. 33. Carlson BA, Dubay MM, Sausville EA, Brizuela
17. Sherr CJ, Roberts, JM: CDK inhibitors: positive and L,Worland PJ: Flavopiridol induces G1 arrest with
negative regulators of G1-phase progression. Genes Dev inhibition of cyclin-dependent kinase (CDK) 2 and CDK4
1999; 13:1501–1512. in human breast carcinoma cells. Cancer Res 1996;
18. Serrano M, Hannon GJ, Beach D: A new regulatory motif 56:2973–2978.
in cell-cycle control causing specific inhibition of cyclin 34. Cartee L, Wang Z, Decker RH, et al: The cyclin-
D/CDK4. Nature 1993; 366:704–707. dependent kinase inhibitor (CDKI) flavopiridol disrupts
19. Toyoshima H, Hunter T: p27, a novel inhibitor of G1 phorbol 12-myristate 13-acetate-induced differentiation
cyclin-Cdk protein kinase activity, is related to p21. Cell and CDKI expression while enhancing apoptosis in
1994; 78:67–74. human myeloid leukemia cells. Cancer Res 2001;
20. Alevizopoulos K, Vlach J, Hennecke S, Amati B: Cyclin E 61:2583–2591.
and c-myc promote cell proliferation in the presence of 35. Senderowicz AM: The cell cycle as a target for cancer
p16INK4a and of hypophosphorylated retinoblastoma therapy: basic and clinical findings with the small
family proteins. EMBO J 1997; 16:5322–5333. molecule inhibitors flavopiridol and UCN-01. Oncologist
21. Prall OW, Rogan EM, Musgrove EA, Watts CK, 2002; 7(Suppl 3):12–19.
Sutherland RL: c-myc or cyclin D1 mimics estrogen 36. deVere White RW, Deitch AD, Jackson AG, et al: Racial
effects on cyclin E-Cdk2 activation and cell cycle reentry. differences in clinically localized prostate cancers of black
Mol Cell Biol 1998; 18:4499–4508. and white men, J Urol 1998; 159:1979–1982. [Discussion
22. Fan Z, Shang BY, LuY, Chou JL. Mendelsohn J: 1982–1983.]
Reciprocal changes in p27(Kip1) and p21(Cip1) in growth 37. Khoo VS, Pollack A, Cowen D, et al: Relationship of Ki-
inhibition mediated by blockade or overstimulation of 67 labeling index to DNA-ploidy, S-phase fraction, and
epidermal growth factor receptors. Clin Cancer Res 1997; outcome in prostate cancer treated with radiotherapy.
3:1943–1948. Prostate 1999; 41:166–172.
23. Peng D, Fan Z, Lu Y, et al: Anti-epidermal growth factor 38. Agarwal ML, Taylor WR, Chernov MV, Chernova OB,
receptor monoclonal antibody 225 up-regulates p27KIP1 Stark GR: The p53 network. J Biol Chem 1998; 273:1–4.
and induces G1 arrest in prostatic cancer cell line DU145, 39. Lane DP: Cancer p53, guardian of the genome. Nature
Cancer Res 1996; 56:3666–3669. 1992; 358:15–16.
24. Li Q, Dang CV: c-myc overexpression uncouples DNA 40. Haldar S, Basu A, Croce CM: Bcl2 is the guardian of
replication from mitosis. Mol Cell Biol 1999; microtubule integrity, Cancer Res 1997; 57:229–233.
19:5339–5351. 41. Sionov RV, Haupt Y: The cellular response to p53: the
25. Suzuki N, Del Villar K, Tamanoi F: Farnesyltransferase decision between life and death. Oncogene 1999;
inhibitors induce dramatic morphological changes of 18:6145–6157.
KNRK cells that are blocked by microtubule 42. Oren M: Decision making by p53: life, death and cancer,
interfering agents, Proc Natl Acad Sci U S A 1998; Cell Death Differ 2003; 10:431–442.
95:10499–10504. 43. Cory S, Adams JM: The Bcl2 family: regulators of the
26. Moasser MM, Srethapakdi M, Sachar KS, Kraker AJ, cellular life-or-death switch, Nat Rev Cancer 2002;
Rosen N: Inhibition of Src kinases by a selective tyrosine 2:647–656.
kinase inhibitor causes mitotic arrest. Cancer Res 1999; 44. Berges RR, Vukanovic J, Epstein JI, et al: Implication of
59:6145–6152. cell kinetic changes during the progression of human
27. Hartwell LH, Weinert TA: Checkpoints: controls that prostatic cancer. Clin Cancer Res 1995; 1:473–480.
ensure the order of cell cycle events. Science 1989; 45. Westin P, Stattin P, Damber JE, Bergh A: Castration
246:629–634. therapy rapidly induces apoptosis in a minority and
28. Hartwell L H, Kastan MB: Cell cycle control and cancer. decreases cell proliferation in a majority of human
Science 1994; 266:1821–1828. prostatic tumors. Am J Pathol 1995; 146:1368–1375.
29. Park DS, Morris EJ, Greene LA, Geller HM: G1/S cell 46. Chang J, Ormerod M, Powles TJ, et al: Apoptosis and
cycle blockers and inhibitors of cyclin-dependent kinases proliferation as predictors of chemotherapy response in
suppress camptothecin-induced neuronal apoptosis. patients with breast carcinoma, Cancer 2000;
J Neurosci 1997; 17:1256–1270. 89:2145–2152.
30. Chen JG, Horwitz, SB: Differential mitotic responses to 47. Kulkarni RP, Reynolds KW, Newlands ES, et al:
microtubule-stabilizing and microtubule-destabilizing Cytoreductive surgery in disseminated non-seminomatous
drugs, Cancer Res 2002; 62:1935–1938. germ cell tumours of testis, Br J Surg 1991; 78:226–229.
76 Part I Principles of Urologic Oncology

48. Baniel J, Foster RS, Gonin R, et al: Late relapse of of initiated human prostatic epithelium. Cancer Res 1999;
testicular cancer. J Clin Oncol 1995; 13:1170–1176. 59:5002–5011.
49. Steyerberg EW, Keizer HJ, Fossa SD, et al: Prediction of 63. Henshall SM, Quinn DI, Lee CS, et al: Altered expression
residual retroperitoneal mass histology after of androgen receptor in the malignant epithelium and
chemotherapy for metastatic nonseminomatous germ adjacent stroma is associated with early relapse in prostate
cell tumor: multivariate analysis of individual patient cancer. Cancer Res 2001; 61:423–427.
data from six study groups. J Clin Oncol 1995; 64. Raghavan D, Cox K, Pearson BS, et al: Oral
13:1177–1187. cyclophosphamide for the management of hormone-
50. Brenner PC, Herr HW, Morse MJ, et al: Simultaneous refractory prostate cancer. Br J Urol 1993; 72:625–628.
retroperitoneal, thoracic, and cervical resection of 65. Beckley S, Wajsman Z, Slack N, Mittelman A, Murphy
postchemotherapy residual masses in patients with GP: The chemotherapy of prostatic carcinoma. Scand J
metastatic nonseminomatous germ cell tumors of the Urol Nephrol Suppl 1980; 55:151–162.
testis, J Clin Oncol 1996; 14:1765–1769. 66. Raghavan D, Tannock IF: Clinical trials in genitourinary
51. Ulbright TM, Loehrer PJ, Roth LM, et al: The oncology: what have they achieved? In Smith PH,
development of non-germ cell malignancies within germ Chisholm GD (eds): Combination Therapy in Urological
cell tumors. A clinicopathologic study of 11 cases. Cancer Malignancy, pp 226–253. London, Springer Verlag, 1989.
1994; 54:1824–1833. 67. Richie JP: Intravesical chemotherapy. Treatment selection,
52. Orazi A, Neiman RS, Ulbright TM, et al: Hematopoietic techniques, and results. Urol Clin North Am 1992;
precursor cells within the yolk sac tumor component are 19:521–527.
the source of secondary hematopoietic malignancies in 68. Doll DC, Weiss RB, Issell BF: Mitomycin: ten years
patients with mediastinal germ cell tumors. Cancer 1993; after approval for marketing. J Clin Oncol 1985;
71:3873–3881. 3:276–286.
53. Ledermann JA, Holden L, Newlands ES, et al: The long- 69. Highley MS, van Oosterom AT, Maes RA, De Bruijn
term outcome of patients who relapse after chemotherapy EA: Intravesical drug delivery. Pharmacokinetic and
for non-seminomatous germ cell tumours, Br J Urol 1994; clinical considerations. Clin Pharmacokinet 1999;
74:225–230. 37:59–73.
54. Toner GC, Panicek DM, Heelan RT, et al: Adjunctive 70. Hudes G: Estramustine-based chemotherapy. Semin Urol
surgery after chemotherapy for nonseminomatous germ Oncol 1997; 15:13–19.
cell tumors: recommendations for patient selection. J Clin 71. Gerl A, Clemm C, Kohl P, Schalhorn A, Wilmanns W:
Oncol 1990; 8:1683–1694. Central nervous system as sanctuary site of relapse in
55. Steyerberg EW, Donohue JP, Gerl A, et al: Residual patients treated with chemotherapy for metastatic
masses after chemotherapy for metastatic testicular testicular cancer. Clin Exp Metastasis 1994;
cancer: the clinical implications of the association between 12:226–230.
retroperitoneal and pulmonary histology. Re-analysis of 72. Reddel RR, Thompson JF, Raghavan D, et al: Surgery in
Histology in Testicular Cancer (ReHiT) Study Group. patients with advanced germ cell malignancy following a
J Urol 1997; 158:474–478. clinical partial response to chemotherapy. J Surg Oncol
56. Mayer F, Stoop H, Scheffer GL, et al: Molecular 1983; 23:223–227.
determinants of treatment response in human germ cell 73. Pinedo HM, Peters GF: Fluorouracil: biochemistry and
tumors. Clin Cancer Res 2003; 9:767–773. pharmacology. J Clin Oncol 1988; 6:1653–1664.
57. Brown JL, Russell PJ, Philips J, Wotherspoon J, Raghavan 74. Eliopoulos N, Cournoyer D, Momparler RL: Drug
D: Clonal analysis of a bladder cancer cell line: an resistance to 5-aza-2′-deoxycytidine, 2′,2′-difluoro-
experimental model of tumour heterogeneity. Br J Cancer deoxycytidine, and cytosine arabinoside conferred by
1990; 61:369–376. retroviral-mediated transfer of human cytidine deaminase
58. Quinn DI, Henshall SM, Head DR, et al: Prognostic cDNA into murine cells. Cancer Chemother Pharmacol
significance of p53 nuclear accumulation in localized 1998; 42:373–378.
prostate cancer treated with radical prostatectomy. Cancer 75. Merriman RL, Hertel LW, Schultz RM, et al:
Res 2000; 60:1585–1594. Comparison of the antitumor activity of gemcitabine and
59. Suzuki H. Freije D, Nusskern DR, et al: Interfocal ara-C in a panel of human breast, colon, lung and
heterogeneity of PTEN/MMAC1 gene alterations in pancreatic xenograft models., Invest New Drugs 1996;
multiple metastatic prostate cancer tissues. Cancer Res 14:243–247.
1998; 58:204–209. 76. Reigner B, Blesch K, Weidekamm E: Clinical
60. Craft N, Chhor C, Tran C, et al: Evidence for clonal pharmacokinetics of capecitabine. Clin Pharmacokinet
outgrowth of androgen-independent prostate cancer cells 2001; 40:85–104.
from androgen-dependent tumors through a two-step 77. Twelves C: Vision of the future: capecitabine. Oncologist
process. Cancer Res 1999; 59:5030–5036. 2001; 6(Suppl 4):35–39.
61. Xu X, Stower MJ, Reid IN, Garner RC, Burns PA: 78. Ishikawa T, Sekiguchi F, Fukase Y, Sawada N, Ishitsuka
Molecular screening of multifocal transitional cell H: Positive correlation between the efficacy of
carcinoma of the bladder using p53 mutations as capecitabine and doxifluridine and the ratio of thymidine
biomarkers. Clin Cancer Res 1996; 2:1795–800. phosphorylase to dihydropyrimidine dehydrogenase
62. Olumi AF, Grossfeld GD, Hayward SW, et al: activities in tumors in human cancer xenografts. Cancer
Carcinoma-associated fibroblasts direct tumor progression Res 1998; 58:685–690.
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 77

79. Murray M: P450 enzymes. Inhibition mechanisms, 98. Quinn DI, Day RO: Drug interactions of clinical
genetic regulation and effects of liver disease. Clin importance. An updated guide. Drug Saf 1995;
Pharmacokinet 1992; 23:132–146. 12:393–452.
80. Slaughter RL, Edwards DJ: Recent advances: the 99. Wagner JG: History of pharmacokinetics. Pharmacol
cytochrome P450 enzymes, Annals of Pharmacotherapy Ther 1981; 12: 537–562.
1995; 29:619–624. 100. Rodvold KA, Rushing DA, Tewksbury DA: Doxorubicin
81. Creaven PJ, Allen LM, Cohen MH, Nelson RL: Studies clearance in the obese. J Clin Oncol 1988; 6:
on the clinical pharmacology and toxicology of 1321–1327.
isophosphamide (NSC-109724). Cancer Treat Rep 1976; 101. Lind, MJ, Margison JM, Cerny T, Thatcher N,
60:445–449. Wilkinson PM: Prolongation of ifosfamide elimination
82. Allen LM, Creaven PJ: Pharmacokinetics of ifosfamide. half-life in obese patients due to altered drug
Clin Pharmacol Ther 1975; 17:492–498. distribution. Cancer Chemother Pharmacol 1989;
83. Colvin M: The comparative pharmacology of 25:139–142.
cyclophosphamide and ifosfamide. Semin Oncol 1982; 102. RobertJ, Hoerni B: Age dependence of the early-phase
9:2–7. pharmacokinetics of doxorubicin. Cancer Res 1983;
84. Lind MJ, Margison JM, Cerny T, Thatcher N, Wilkinson 43:4467–4469.
P M: Comparative pharmacokinetics and alkylating 103. Testa B, Jenner P: Inhibitors of Cytochrome P-450s and
activity of fractionated intravenous and oral ifosfamide in their mechanism of action. Drug Metab Rev 1981;
patients with bronchogenic carcinoma. Cancer Res 1989; 12:1–117.
49:753–757. 104. Tardiff RG, Dubois KP: Inhibition of hepatic
85. Powis G: Effect of human renal and hepatic disease on the microsomal enzymes by alkylating agents. Arch Int
pharmacokinetics of anticancer drugs. Cancer Treat Rev Pharmacodyn Ther 1969; 177:445–456.
1982; 9:85–124. 105. Drayer DE: Pathways of drug metabolism in man. Med
86. Benjamin RS, Riggs CE, Jr, Bachur NR: Plasma Clin North Am 1974; 58:927–944.
pharmacokinetics of adriamycin and its metabolites in 106. Liegler DG, Henderson ES, Hahn MA, Oliverio VT:
humans with normal hepatic and renal function. Cancer The effect of organic acids on renal clearance of
Res 1977; 37:1416–1420. methotrexate in man. Clin Pharmacol Ther 1969;
87. Riggs CE, Jr, Benjamin RS, Serpick AA, Bachur NR: 10:849–857.
Bilary disposition of adriamycin. Clin Pharmacol Ther 107. Sand TE, Jacobsen S: Effect of urine pH and flow on
1977; 22:234–241. renal clearance of methotrexate. Eur J Clin Pharmacol
88. Grochow LB, Colvin M: Clinical pharmacokinetics 1981; 19:453–456.
of cyclophosphamide. Clin Pharmacokinet 1979; 4: 108. Relling MV, Fairclough D, Ayers D, et al: Patient
380–394. characteristics associated with high-risk methotrexate
89. Colvin M, Hilton J: Pharmacology of cyclophosphamide concentrations and toxicity. J Clin Oncol 1994;
and metabolites. Cancer Treat Rep 1981; 65(Suppl 3):89–95. 12:1667–1672.
90. Owellen RJ, Root MA, Hains FO: Pharmacokinetics of 109. Valverde MA, Diaz M, Sepulveda FV, et al: Volume-
vindesine and vincristine in humans. Cancer Res 1977; regulated chloride channels associated with the human
37:2603–2607. multidrug-resistance P-glycoprotein. Nature 1992;
91. Bruno R, Hille D, Riva A, et al: Population 355:830–833.
pharmacokinetics/pharmacodynamics of docetaxel in 110. Juranka PF, Zastawny RL, Ling V: P-glycoprotein:
phase II studies in patients with cancer. J Clin Oncol multidrug-resistance and a superfamily of membrane-
1998; 16:187–196. associated transport proteins. FASEB J 1989;
92. Gormley PE, Bull JM, LeRoy AF, Cysyk R: Kinetics of 3:2583–2592.
cis-dichlorodiammineplatinum. Clin Pharmacol Ther 111. Abraham EH, Prat AG, Gerweck L, et al: The
1979; 25:351–357. multidrug resistance (mdr1) gene product functions as
93. van der Vijgh WJ: Clinical pharmacokinetics of an ATP channel. Proc Natl Acad Sci U S A 1993;
carboplatin. Clin Pharmacokinet 1991; 21: 90:312–316.
242–261. 112. Speicher LA, Barone LR, Chapman AE, et al:
94. Chabner BA, Donehower RC, Schilsky RL: Clinical P-glycoprotein binding and modulation of the
pharmacology of methotrexate. Cancer Treat Rep 1981; multidrug-resistant phenotype by estramustine, J Natl
65(Suppl 1):51–54. Cancer Inst 1994; 86:688–694.
95. Alberts DS, Chen HS, Liu R, et al: Bleomycin 113. Abbaszadegan MR, Cress AE, Futscher BW, Bellamy
pharmacokinetics in man. I. Intravenous administration. WT, Dalton WS: Evidence for cytoplasmic
Cancer Chemother Pharmacol 1978; 1:177–181. P-glycoprotein location associated with increased
96. Fleisher D, Li C, Zhou Y, Pao LH, Karim A: Drug, meal multidrug resistance and resistance to chemosensitizers.
and formulation interactions influencing drug absorption Cancer Res 1996; 56:5435–5442.
after oral administration. Clinical implications. Clin 114. Hazlehurst LA, Foley NE, Gleason-Guzman MC:
Pharmacokinet 1999; 36:233–254. Multiple mechanisms confer drug resistance to
97. Prescott LF: Gastrointestinal absorption of drugs. Med mitoxantrone in the human 8226 myeloma cell line.
Clin North Am 1974; 58:907–916. Cancer Res 1999; 59:1021–1028.
78 Part I Principles of Urologic Oncology

115. Goldstein LJ, Galski H, Fojo A, et al: Expression of a 131. Brandes LJ, Simons KJ, Bracken SP, Warrington RC:
multidrug resistance gene in human cancers. J Natl Results of a clinical trial in humans with refractory
Cancer Inst 1989; 81:116–124. cancer of the intracellular histamine antagonist, N,
116. Chevillard S, Pouillart P, Beldjord C, et al: Sequential N-diethyl-2-[4-(phenylmethyl)phenoxy]ethanamine-HCl,
assessment of multidrug resistance phenotype and in combination with various single antineoplastic agents.
measurement of S-phase fraction as predictive markers of J Clin Oncol 1994; 12:1281–1290.
breast cancer response to neoadjuvant chemotherapy. 132. Brandes LJ, Bracken SP, Ramsey EW: N,N-diethyl-2-
Cancer 1996; 77:292–300. [4-(phenylmethyl)phenoxy]ethanamine in combination
117. Benet LZ, Cummins CL: The drug efflux-metabolism with cyclophosphamide: an active, low-toxicity regimen
alliance: biochemical aspects. Adv Drug Deliv Rev 2001; for metastatic hormonally unresponsive prostate cancer.
50(Suppl 1):S3-S11. J Clin Oncol 1995; 13:1398–1403.
118. Cummins CL, Jacobsen W, Benet LZ: Unmasking 133. Raghavan D, Brandes LJ, Klapp K, et al: Phase II trial of
the dynamic interplay between intestinal P-glycoprotein mitoxantrone plus tesmilifene (DPPE) in hormone-
and CYP3A4. J Pharmacol Exp Ther 2002; refractory prostate cancer: high subjective and objective
300:1036–1045. response in patients with symptomatic bone metastases.
119. Fricker G, Miller DS: Relevance of multidrug resistance J Clin Oncol (in press).
proteins for intestinal drug absorption in vitro and in 134. Niedner H, Christen R, Lin X, Kondo A. Howell SB:
vivo, Pharmacol Toxicol 2002; 90:5–13. Identification of genes that mediate sensitivity to
120. Reinecke P, Schmitz M, Schneider EM, Gabbert HE, cisplatin. Mol Pharmacol 2001; 60:1153–1160.
Gerharz CD: Multidrug resistance phenotype and 135. Guminski AD, Harnett PR, deFazio A: Scientists and
paclitaxel (Taxol) sensitivity in human renal carcinoma clinicians test their metal-back to the future with
cell lines of different histologic types. Cancer Invest platinum compounds. Lancet Oncol 2002; 3:
2000; 18:614–625. 312–318.
121. Sugawara I: Expression and functions of P-glycoprotein 136. Hospers GA, Meijer C, de Leij L, et al: A study of
(mdr1 gene product) in normal and malignant tissues, human small-cell lung carcinoma (hSCLC) cell lines
Acta Pathol Jpn 1990; 40:545–553. with different sensitivities to detect relevant mechanisms
122. Shinohara N, Nonomura K, Takakura F, et al: of cisplatin (CDDP) resistance. Int J Cancer 1990;
Expression of multidrug resistance gene product 46:138–144.
(P-glycoprotein) in transitional cell carcinomas of the 137. Liebmann JE, Hahn SM, Cook JA, et al: Glutathione
upper urinary tract. Eur Urol 1994; 26:327–333. depletion by L-buthionine sulfoximine antagonizes taxol
123. Shinohara N, Liebert M, Wedemeyer G, Chang JH, cytotoxicity. Cancer Res 1993; 53:2066–2070.
Grossman HB: Evaluation of multiple drug resistance in 138. Pendyala L, Creaven PJ, Perez R, Zdanowicz JR,
human bladder cancer cell lines. J Urol 1993; Raghavan D: Intracellular glutathione and cytotoxicity of
150:505–509. platinum complexes. Cancer Chemother Pharmacol
124. Park J, Shinohara N, Liebert M, et al: P-glycoprotein 1995; 36:271–278.
expression in bladder cancer. J Urol 1994; 151:43–46. 139. Christen RD, Hom DK, Porter DC, et al: Epidermal
125. Petrylak DP, Scher HI, Reuter V, O’Brien JP, Cordon- growth factor regulates the in vitro sensitivity of human
Cardo C: P-glycoprotein expression in primary and ovarian carcinoma cells to cisplatin. J Clin Invest 1990;
metastatic transitional cell carcinoma of the bladder. Ann 86:1632–1640.
Oncol 1994; 5:835–840. 140. Cenni B, Aebi S, Nehme A, Christen RD: Epidermal
126. Barrand MA, Heppell-Parton AC, Wright KA, Rabbitts growth factor enhances cisplatin-induced apoptosis by a
PH, Twentyman PR: A 190-kilodalton protein caspase 3 independent pathway. Cancer Chemother
overexpressed in non-P-glycoprotein-containing Pharmacol 2001; 47:397–403.
multidrug-resistant cells and its relationship to the MRP 141. Fan Z, Masui H, Altas I, Mendelsohn J: Blockade of
gene. J Natl Cancer Inst 1994; 86:110–117. epidermal growth factor receptor function by bivalent
127. Lum BL, Fisher GA, Brophy NA, et al: Clinical trials of and monovalent fragments of 225 anti-epidermal growth
modulation of multidrug resistance. Pharmacokinetic factor receptor monoclonal antibodies. Cancer Res 1993;
and pharmacodynamic considerations. Cancer 1993; 53:4322–4328.
72:3502–3514. 142. Fan Z, Baselga J, Masui H, Mendelsohn J: Antitumor
128. Dalton WS, Grogan TM, Meltzer PS, et al: Drug- effect of anti-epidermal growth factor receptor
resistance in multiple myeloma and non-Hodgkin’s monoclonal antibodies plus cis-diamminedichloroplatinum
lymphoma: detection of P-glycoprotein and potential on well established A431 cell xenografts. Cancer Res
circumvention by addition of verapamil to 1993; 53:4637–4642.
chemotherapy, J Clin Oncol 1989; 7:415–424. 143. Lipponen P, Eskelinen M: Expression of epidermal
129. Ramirez LH, Munck JN, Zhao Z, et al: Verapamil- growth factor receptor in bladder cancer as related to
reversing concentrations induce blood flow changes that established prognostic factors, oncoprotein (c-erbB-2,
could counteract in vivo the MDR-1-modulating effects, p53) expression and long-term prognosis. Br J Cancer
Cancer 1994; 74:810–816. 1994; 69:1120–1125.
130. Russell PJ, Palavidis Z, Rozinova E, et al: 144. Gorgoulis VG, Barbatis C, Poulias I, Karameris AM:
Characterization of a new human bladder cancer cell Molecular and immunohistochemical evaluation of
line. UCRU-BL-28. J Urol 1993; 150:1038–1044. epidermal growth factor receptor and c-erb-B-2 gene
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 79

product in transitional cell carcinomas of the urinary 161. Ratain MJ, Mick R, Schilsky RL, Siegler M: Statistical
bladder: a study in Greek patients. Mod Pathol 1995; and ethical issues in the design and conduct of phases I
8:758–764. and II clinical trials of new anticancer agents. J Natl
145. Chow NH, Chan SH, Tzai TS, Ho CL, Liu HS: Cancer Inst 1993; 85:1637–1643.
Expression profiles of ErbB family receptors and 162. Creaven PJ, Mihich E: The clinical toxicity of anticancer
prognosis in primary transitional cell carcinoma of the drugs and its prediction. Semin Oncol 1977; 4:147–163.
urinary bladder. Clin Cancer Res 2001; 7:1957–1962. 163. Staquet M, Sylvester R: A decision theory approach to
146. Hollstein M, Sidransky D, Vogelstein B, Harris CC: phase II clinical trials. Biomedicine 1977; 26:262–266.
p53 mutations in human cancers. Science 1991; 164. Lee YJ, Staquet M, Simon R, Catane R, Muggia F: Two-
253:49–53. stage plans for patient accrual in phase II cancer clinical
147. Fritsche M, Haessler C, Brandner G: Induction of trials. Cancer Treat Rep 1979; 63:1721–1726.
nuclear accumulation of the tumor-suppressor protein 165. Simon R: Optimal two-stage designs for phase II clinical
p53 by DNA-damaging agents. Oncogene 1993; trials. Control Clin Trials 1989; 10:1–10.
8:307–318. 166. Korn EL, Arbuck SG, Pluda JM, et al: Clinical trial
148. Aas T, Borresen AL, Geisler S, et al: Specific P53 designs for cytostatic agents: are new approaches
mutations are associated with de novo resistance to needed? J Clin Oncol 2001; 19:265–272.
doxorubicin in breast cancer patients. Nat Med 1996; 167. Rosenberg B, VanCamp L, Trosko JE, Mansour VH:
2:811–814. Platinum compounds: a new class of potent antitumour
149. Lin X, Ramamurthi K, Mishima M, et al: p53 modulates agents. Nature 1969; 222:385–386.
the effect of loss of DNA mismatch repair on the 168. Gottesman MM: Mechanisms of cancer drug resistance,
sensitivity of human colon cancer cells to the cytotoxic Annu. Rev. Med 2002; 53:615–627.
and mutagenic effects of cisplatin. Cancer Res 2001; 169. Hospers GA, Mulder NH, de Jong B, et al: Characterization
61:1508–1516. of a human small cell lung carcinoma cell line with acquired
150. Achanta G, Pelicano H, Feng L, Plunkett W, Huang P: resistance to cis-diamminedichloroplatinum(II) in vitro.
Interaction of p53 and DNA-PK in response to Cancer Res 1988; 48: 6803–6807.
nucleoside analogues: potential role as a sensor complex 170. Meijer C, Mulder NH, Hospers GA, Uges DR, de Vries
for DNA damage. Cancer Res 2001; 61:8723–8729. EG: The role of glutathione in resistance to cisplatin in a
151. Cassinelli G, Supino R, Perego P, et al: A role for loss of human small cell lung cancer cell line. Br J Cancer 1990;
p53 function in sensitivity of ovarian carcinoma cells to 62:72–77.
taxanes. Int J Cancer 2001; 92:738–747. 171. Reed E: Platinum-DNA adduct, nucleotide excision
152. Esrig D, Elmajian D, Groshen S, et al: Accumulation of repair and platinum based anti-cancer chemotherapy.
nuclear p53 and tumor progression in bladder cancer. Cancer Treat Rev 1998; 24:331–344.
N Engl J Med 1994; 331:1259–1264. 172. Metzger R, Leichman CG, Danenberg KD, et al: ERCC1
153. Sarkis AS, Bajorin DF, Reuter VE, et al: Prognostic value mRNA levels complement thymidylate synthase mRNA
of p53 nuclear overexpression in patients with invasive levels in predicting response and survival for gastric cancer
bladder cancer treated with neoadjuvant MVAC. J Clin patients receiving combination cisplatin and fluorouracil
Oncol 1995; 13:1384–1390. chemotherapy. J Clin Oncol 1998; 16:309–316.
154. Goldie JH, Coldman AJ: A mathematic model for 173. Lord RV, Brabender J, Gandara D, et al: Low
relating the drug sensitivity of tumors to their ERCC1 expression correlates with prolonged survival
spontaneous mutation rate. Cancer Treat Rep 1979; after cisplatin plus gemcitabine chemotherapy in
63:1727–1733. non-small cell lung cancer. Clin Cancer Res 2002;
155. Day RS: Treatment sequencing, asymmetry, and 8:2286–2291.
uncertainty: protocol strategies for combination 174. Ferry KV, Hamilton TC, Johnson SW: Increased
chemotherapy. Cancer Res 1986; 46:3876–3885. nucleotide excision repair in cisplatin-resistant ovarian
156. Schwartz D, Lellouch J: Explanatory and pragmatic cancer cells: role of ERCC1-XPF. Biochem Pharmacol
attitudes in therapeutical trials. J Chronic Dis 1967; 2000; 60:1305–1313.
20:637–648. 175. DeConti RC, Toftness BR, Lange RC, Creasey WA:
157. Crawford ED, Wood DP, Petrylak DP, et al: Southwest Clinical and pharmacological studies with cis-
Oncology Group studies in bladder cancer. Cancer 2003; diamminedichloroplatinum (II). Cancer Res 1973;
97:2099–2108. 33:1310–1315.
158. de Wit R: Overview of bladder cancer trials in the 176. Himmelstein KJ, Patton TF, Belt RJ, et al: Clinical
European Organization for Research and Treatment. kinetics on intact cisplatin and some related species. Clin
Cancer 2003; 97:2120–2126. Pharmacol Ther 1981; 29:658–664.
159. Dreicer R, Roth B, Wilding G: Overview of advanced 177. Safirstein R, Winston J, Goldstein M, et al: Cisplatin
urothelial cancer trials of the Eastern Cooperative nephrotoxicity. Am J Kidney Dis 1986; 8:356–367.
Oncology Group. Cancer 2003; 97:2109–2114. 178. Kelsen DP, Alcock N, Young CW: Cisplatin
160. Daugherty C, Ratain MJ, Grochowski E, et al: nephrotoxicity. Correlation with plasma platinum
Perceptions of cancer patients and their physicians concentrations. Am J Clin Oncol 1985; 8:77–80.
involved in phase I trials. J Clin Oncol 1995; 179. Boyer M, Raghavan D, Harris PJ, et al: Lack of late
13:1062–1072. toxicity in patients treated with cisplatin-containing
80 Part I Principles of Urologic Oncology

combination chemotherapy for metastatic testicular 194. Petrioli R, Frediani B, Manganelli A, et al: Comparison
cancer. J Clin Oncol 1990; 8:21–26. between a cisplatin-containing regimen and a
180. Boyer M Raghavan D: Toxicity of treatment of germ cell carboplatin-containing regimen for recurrent or
tumors. Semin Oncol 1992; 19:128–142. metastatic bladder cancer patients. A randomized phase
181. Calvert H, Judson I, van der Vijgh WJ: Platinum II study. Cancer 1996; 77:344–351.
complexes in cancer medicine: pharmacokinetics and 195. Bajorin DF, Sarosdy MF, Pfister DG, et al: Randomized
pharmacodynamics in relation to toxicity and therapeutic trial of etoposide and cisplatin versus etoposide and
activity. Cancer Surv 1993; 17:189–217. carboplatin in patients with good-risk germ cell tumors:
182. Harland SJ, Newell DR, Siddik ZH, et al: a multiinstitutional study. J Clin Oncol 1993;
Pharmacokinetics of cis-diammine-1,1-cyclobutane 11:598–606.
dicarboxylate platinum(II) in patients with normal and 196. Horwich A, Sleijfer DT, Fossa SD, et al: Randomized
impaired renal function. Cancer Res 1984; trial of bleomycin, etoposide, and cisplatin compared
44:1693–1697. with bleomycin, etoposide, and carboplatin in good-
183. Egorin MJ, Van Echo DA, Tipping SJ, et al: prognosis metastatic nonseminomatous germ cell cancer:
Pharmacokinetics and dosage reduction of cis- a Multiinstitutional Medical Research Council/European
diammine(1,1-cyclobutanedicarboxylato)platinum in Organization for Research and Treatment of Cancer
patients with impaired renal function. Cancer Res 1984; Trial. J Clin Oncol 1997; 15:1844–1852.
44:5432–5438. 197. Horwich A, Oliver RT, Wilkinson PM, et al: A medical
184. Calvert AH, Newell DR, Gumbrell LA, et al: research council randomized trial of single agent
Carboplatin dosage: prospective evaluation of a simple carboplatin versus etoposide and cisplatin for advanced
formula based on renal function. J Clin Oncol 1989; metastatic seminoma. MRC Testicular Tumour Working
7:1748–1756. Party. Br J Cancer 2000; 83:1623–1629.
185. Kelly WK, Curley T, Slovin S, et al: Paclitaxel, 198. Kidani Y, Noji M, Tashiro T: Antitumor activity of
estramustine phosphate, and carboplatin in patients with platinum(II) complexes of 1,2-diamino-cyclohexane
advanced prostate cancer. J Clin Oncol 2001; 19:44–53. isomers. Gann 1980; 71:637–643.
186. Jungi WF, Bernhard J, Hurny C, et al: Effect of 199. Pendyala L, Creaven PJ: In vitro cytotoxicity, protein
carboplatin on response and palliation in hormone- binding, red blood cell partitioning, and
refractory prostate cancer. Swiss Group for Clinical biotransformation of oxaliplatin. Cancer Res 1993;
Cancer Research (SAKK). Support Care Cancer 1998; 53:5970–5976.
6:462–468. 200. Caussanel JP, Levi F, Brienza S, et al: Phase I trial of
187. Bellmunt J, Albanell J, Gallego OS, et al: Carboplatin, 5-day continuous venous infusion of oxaliplatin at
methotrexate, and vinblastine in patients with bladder circadian rhythm-modulated rate compared with
cancer who were ineligible for cisplatin-based constant rate. J Natl Cancer Inst 1990; 82:1046–1050.
chemotherapy. Cancer 1992; 70:1974–1979. 201. Machover D, Diaz-Rubio E, de Gramont A, et al: Two
188. Skarlos DV, Samantas E, Kosmidis P, et al: Randomized consecutive phase II studies of oxaliplatin (L-OHP) for
comparison of etoposide-cisplatin vs. etoposide- treatment of patients with advanced colorectal carcinoma
carboplatin and irradiation in small-cell lung cancer. A who were resistant to previous treatment with
Hellenic Co-operative Oncology Group study. Ann fluoropyrimidines. Ann Oncol 1996; 7:95–98.
Oncol 1994; 5:601–607. 202. Extra JM, Espie M, Calvo F, et al: Phase I study of
189. Scagliotti GV, De Marinis F, Rinaldi M, et al: Phase III oxaliplatin in patients with advanced cancer. Cancer
randomized trial comparing three platinum-based Chemother Pharmacol 1990; 25:299–303.
doublets in advanced non-small-cell lung cancer. J Clin 203. Shirota Y, Stoehlmacher J, Brabender J, et al: ERCC1
Oncol 2002; 20:4285–4291. and thymidylate synthase mRNA levels predict survival
190. Swenerton K, Jeffrey J, Stuart G, et al: Cisplatin- for colorectal cancer patients receiving combination
cyclophosphamide versus carboplatin-cyclophosphamide oxaliplatin and fluorouracil chemotherapy. J Clin Oncol
in advanced ovarian cancer: a randomized phase III study 2001; 19:4298–4304.
of the National Cancer Institute of Canada Clinical 204. Bokemeyer C, Kollmannsberger C, Harstrick A, et al:
Trials Group. J Clin Oncol 1992; 10:718–726. Treatment of patients with cisplatin-refractory testicular
191. Alberts DS, Green S, Hannigan EV, et al: Improved germ-cell cancer. German Testicular Cancer Study
therapeutic index of carboplatin plus cyclophosphamide Group (GTCSG). Int J Cancer 1999; 83:848–851.
versus cisplatin plus cyclophosphamide: final report by 205. Kollmannsberger C, Rick O, Derigs HG, et al: Activity
the Southwest Oncology Group of a phase III of oxaliplatin in patients with relapsed or cisplatin-
randomized trial in stages III and IV ovarian cancer. refractory germ cell cancer: a study of the German
J Clin Oncol 1992; 10:706–717. Testicular Cancer Study Group. J Clin Oncol 2002;
192. Lokich J, Anderson N: Carboplatin versus cisplatin in 20:2031–2037.
solid tumors: an analysis of the literature. Ann Oncol 206. Barranco SC, Humphrey RM: The effects of bleomycin
1998; 9:13–21. on survival and cell progression in Chinese hamster cells
193. Go RS, Adjei AA: Review of the comparative in vitro. Cancer Res 1971; 31:1218–1223.
pharmacology and clinical activity of cisplatin and 207. Barlogie B, Drewinko B, Schumann J, Freireich EJ:
carboplatin. J Clin Oncol 1999; 17:409–422. Pulse cytophotometric analysis of cell cycle perturbation
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 81

with bleomycin in vitro. Cancer Res 1976; 225. Greco FA, Brereton HD, Kent H, et al: Adriamycin and
36:1182–1187. enhanced radiation reaction in normal esophagus and
208. Lazo JS, Boland CJ, Schwartz PE: Bleomycin hydrolase skin. Ann Intern Med 1976; 85:294–298.
activity and cytotoxicity in human tumors. Cancer Res 226. Berruti A, Terzolo M, Pia A, Angeli A, Dogliotti L:
1982; 42:4026–4031. Mitotane associated with etoposide, doxorubicin, and
209. Levi JA, Raghavan D, Harvey V, et al: The importance cisplatin in the treatment of advanced adrenocortical
of bleomycin in combination chemotherapy for good- carcinoma. Italian Group for the Study of Adrenal
prognosis germ cell carcinoma. Australasian Germ Cell Cancer. Cancer 1998; 83:2194–2200.
Trial Group. J Clin Oncol 1993; 11:1300–1305. 227. Decker RA, Elson P, Hogan TF, et al: Eastern
210. Loehrer PJ, Sr, Johnson D, Elson P, Einhorn Cooperative Oncology Group study 1879: mitotane and
LH,Trump D: Importance of bleomycin in favorable- adriamycin in patients with advanced adrenocortical
prognosis disseminated germ cell tumors: an Eastern carcinoma. Surgery 1991; 110:1006–1013.
Cooperative Oncology Group trial. J Clin Oncol 1995; 228. Law TM, Mencel P, Motzer RJ: Phase II trial of
13:470–476. liposomal encapsulated doxorubicin in patients with
211. Crooke ST, Bradner WT: Mitomycin C: a review. advanced renal cell carcinoma. Invest New Drugs 1994;
Cancer Treat Rev 1976; 3:121–139. 12:323–325.
212. Dusre L, Rajagopalan S, Eliot HM, Covey JM, Sinha 229. Koeller J, Eble M: Mitoxantrone: a novel anthracycline
BK: DNA interstrand cross-link and free radical derivative. Clin Pharm 1988; 7:574–581.
formation in a human multidrug-resistant cell line from 230. Faulds D, Balfour JA, Chrisp P, Langtry HD:
mitomycin C and its analogues. Cancer Res 1990; Mitoxantrone. A review of its pharmacodynamic and
50:648–652. pharmacokinetic properties, and therapeutic potential in
213. Dusre L, Covey JM, Collins C, Sinha BK: DNA damage, the chemotherapy of cancer. Drugs 1991; 41:400–449.
cytotoxicity and free radical formation by mitomycin C 231. Smyth JF, Macpherson JS, Warrington PS, Leonard RC,
in human cells. Chem Biol Interact 1989; 71:63–78. Wolf CR: The clinical pharmacology of mitozantrone.
214. van Hazel GA, Kovach JS: Pharmacokinetics of Cancer Chemother Pharmacol 1986; 17:149–152.
mitomycin C in rabbit and human. Cancer Chemother 232. Launay MC, Iliadis A, Richard B: Population
Pharmacol 1982; 8:189–192. pharmacokinetics of mitoxantrone performed by a
215. van Hazel GA, Scott M, Rubin J, et al: Pharmacokinetics NONMEM method. J Pharm Sci 1989; 78:877–880.
of mitomycin C in patients receiving the drug alone or 233. Batra VK, Morrison JA, Woodward DL, Siverd NS,
in combination. Cancer Treat Rep 1983; 67:805–810. Yacobi A: Pharmacokinetics of mitoxantrone in man
216. Hanna WT, Krauss S, Regester RF, Murphy WM: Renal and laboratory animals. Drug Metab Rev 1986;
disease after mitomycin C therapy. Cancer 1981; 17:311–329.
48:2583–2588. 234. Hu OY, Chang SP, Law CK, Jian JM, Chen KY:
217. Valavaara R, Nordman E: Renal complications of Pharmacokinetic and pharmacodynamic studies with
mitomycin C therapy with special reference to the total mitoxantrone in the treatment of patients with
dose. Cancer 1985; 55:47–50. nasopharyngeal carcinoma. Cancer 1992; 69:847–853.
218. Chang AY, Kuebler JP, Pandya KJ, et al: Pulmonary 235. Ehninger G, Schuler U, Proksch B, Zeller KP, Blanz J:
toxicity induced by mitomycin C is highly responsive to Pharmacokinetics and metabolism of mitoxantrone. A
glucocorticoids. Cancer 1986; 57:2285–2290. review. Clin Pharmacokinet 1990; 18:365–380.
219. Stewart CF, Ratain MJ: Topoisomerase interactive 236. Alberts DS, Peng YM, Bowden GT, Dalton WS,
agents. In DeVita VTJ, Hellman S, Rosenberg, SA (eds): Mackel C: Pharmacology of mitoxantrone: mode of
Cancer: Principles and Practice of Oncology, 6th edition, action and pharmacokinetics. Invest New Drugs 1985;
pp 415–430. Philadelphia, Lippincott, Williams and 3:101–107.
Wilkins, 2001. 237. Alberts DS, Peng YM, Leigh S, Davis TP, Woodward
220. Tewey KM, Rowe TC, Yang L, Halligan BD, Liu LF: DL: Disposition of mitoxantrone in cancer patients.
Adriamycin-induced DNA damage mediated by Cancer Res 1985; 45:1879–1884.
mammalian DNA topoisomerase II. Science 1984; 238. Van Belle SJ, de Planque MM, Smith IE, et al:
226:466–468. Pharmacokinetics of mitoxantrone in humans following
221. Benjamin RS: Pharmacokinetics of adriamycin (NSC- single-agent infusion or intra-arterial injection therapy
123127) in patients with sarcomas. Cancer Chemother or combined-agent infusion therapy. Cancer Chemother
Rep 1974; 58:271–273. Pharmacol 1986; 18:27–32.
222. Chan KK, Chlebowski RT, Tong M, et al: Clinical 239. Weiss RB: Mitoxantrone: its development and role in
pharmacokinetics of adriamycin in hepatoma patients clinical practice. Oncology 1989; 3:135–141.
with cirrhosis. Cancer Res 1980; 40:1263–1268. 240. Raghavan D, Coorey G, Rosen M, Page J, Farebrother
223. Von Hoff DD, Layard MW, Basa P, et al: Risk factors for T: Management of hormone-resistant prostate cancer: an
doxorubicin-induced congestive heart failure. Ann Intern Australian trial. Semin Oncol 1996; 23:20–23.
Med 1979; 91:710–717. 241. Kantoff PW, Halabi S, Conaway M, et al:
224. Hortobagyi GN, Yap HY, Kau SW, et al: A comparative Hydrocortisone with or without mitoxantrone in men
study of doxorubicin and epirubicin in patients with with hormone-refractory prostate cancer: results of the
metastatic breast cancer. Am J Clin Oncol 1989; cancer and leukemia group B 9182 study [see
12:57–62. comments]. J Clin Oncol 1999; 17:2506–2513.
82 Part I Principles of Urologic Oncology

242. Tannock IF, Osoba D, Stockler MR, et al: 258. Rowinsky EK, Noe DA, Trump DL, et al:
Chemotherapy with mitoxantrone plus prednisone or Pharmacokinetic, bioavailability, and feasibility study of
prednisone alone for symptomatic hormone-resistant oral vinorelbine in patients with solid tumors. J Clin
prostate cancer: a Canadian randomized trial with Oncol 1994; 12:1754–1763.
palliative end points. J Clin Oncol 1996; 14:1756–1764. 259. Carles J, Domenech M, Gelabert-Mas A, et al: Phase II
243. Osoba D, Tannock IF, Ernst DS, Neville AJ: Health- study of estramustine and vinorelbine in hormone-
related quality of life in men with metastatic prostate refractory prostate carcinoma patients. Acta Oncol 1998;
cancer treated with prednisone alone or mitoxantrone 37:187–191.
and prednisone. J Clin Oncol 1999; 17:1654–1663. 260. Schmidinger M, Steger GG, Budinsky AC, et al:
244. Wiseman LR, Spencer CM: Mitoxantrone. A review of Vinorelbine and interferon-alpha2c as second-line
its pharmacology and clinical efficacy in the management therapy in metastatic renal cell carcinoma. Anticancer
of hormone-resistant advanced prostate cancer. Drugs Drugs 2000; 11:175–179.
Aging 1997; 10:473–485. 261. Sweeney CJ, Monaco FJ, Jung SH, et al: A phase II
245. DiPaola RS, Chenven ES, Shih WJ, et al: Mitoxantrone Hoosier Oncology Group study of vinorelbine and
in patients with prostate specific antigen progression estramustine phosphate in hormone-refractory prostate
after local therapy for prostate carcinoma. Cancer 2001; cancer. Ann Oncol 2002; 13:435–440.
92:2065–2071. 262. Oudard S, Caty A, Humblet Y, et al: Phase II study of
246. Wang J, Halford S, Rigg A, et al: Adjuvant mitozantrone vinorelbine in patients with androgen-independent
chemotherapy in advanced prostate cancer. BJU Int prostate cancer. Ann Oncol 2001; 12:847–852.
2000; 86:675–680. 263. Smith MR, Kaufman D, Oh W, et al: Vinorelbine and
247. Nelson RL: The comparative clinical pharmacology and estramustine in androgen-independent metastatic
pharmacokinetics of vindesine, vincristine, and prostate cancer: a phase II study. Cancer 2000;
vinblastine in human patients with cancer. Med Pediatr 89:1824–1828.
Oncol 1982; 10:115–127. 264. Wani MC, Taylor HL, Wall ME, Coggon P, McPhail
248. Owellen RJ, Hartke CA, Hains FO: Pharmacokinetics AT: Plant antitumor agents. VI. The isolation and
and metabolism of vinblastine in humans. Cancer Res structure of taxol, a novel antileukemic and antitumor
1977; 37:2597–2602. agent from Taxus brevifolia. J Am Chem Soc 1971;
249. Jackson DV Jr, Castle MC, Bender RA: Biliary 93:2325–2327.
excretion of vincristine. Clin Pharmacol Ther 1978; 265. Manfredi JJ, Parness J, Horwitz SB: Taxol binds to
24:101–107. cellular microtubules. J Cell Biol 1982; 94: 688–696.
250. Newlands ES, Begent RH, Rustin GJ, Parker D, 266. Parness J, Horwitz SB: Taxol binds to polymerized
Bagshawe KD: Further advances in the management of tubulin in vitro. J Cell Biol 1981; 91:479–487.
malignant teratomas of the testis and other sites. Lancet 267. Schiff PB, Horwitz SB: Taxol stabilizes microtubules in
1983; 1:948–951. mouse fibroblast cells. Proc Natl Acad Sci U S A 1980;
251. Williams SD, Birch R, Einhorn LH, et al: Treatment of 77:1561–1565.
disseminated germ-cell tumors with cisplatin, bleomycin, 268. Schiff PB, FantJ, Horwitz SB: Promotion of
and either vinblastine or etoposide. N Engl J Med 1987; microtubule assembly in vitro by taxol. Nature 1979;
316:1435–1440. 277:665–667.
252. Logothetis CJ, Dexeus FH, Finn L, et al: A prospective 269. Manfredi JJ, Horwitz SB: Taxol: an antimitotic agent
randomized trial comparing MVAC and CISCA with a new mechanism of action. Pharmacol Ther 1984;
chemotherapy for patients with metastatic urothelial 25:83–125.
tumors. J Clin Oncol 1990; 8:1050–1055. 270. McGuire WP, Rowinsky EK, Rosenshein NB, et al:
253. Fellous A, Ohayon R, Vacassin T, et al: Biochemical Taxol: a unique antineoplastic agent with significant
effects of Navelbine on tubulin and associated proteins. activity in advanced ovarian epithelial neoplasms. Ann
Semin Oncol 1989; 16:9–14. Intern Med 1989; 111:273–279.
254. Cros S, Wright M, Morimoto M, et al: Experimental 271. Arbuck SG, Christian MC, Fisherman JS, et al:. Clinical
antitumor activity of Navelbine. Semin Oncol 1989; development of Taxol. J Natl Cancer Inst Monogr
16:15–20. 1993:11–24.
255. Photiou A, Sheikh MN, Bafaloukos D, Retsas S: 272. Donehower RC, Rowinsky EK, Grochow LB,
Antiproliferative activity of vinorelbine (Navelbine) Longnecker SM, Ettinger DS: Phase I trial of taxol in
against six human melanoma cell lines. J Cancer Res patients with advanced cancer. Cancer Treat Rep 1987;
Clin Oncol 1992; 118:249–254. 71:1171–1177.
256. Mathe G, Reizenstein P: Phase I pharmacologic study of 273. Arbuck SG: Paclitaxel: what schedule? What dose?.
a new Vinca alkaloid: navelbine. Cancer Lett 1985; J Clin Oncol 1994; 12:233–236.
27:285–293. 274. Rowinsky EK, Onetto N, Canetta RM, Arbuck SG:
257. Bore P, Rahmani R, van Cantfort J, Focan C, Cano JP: Taxol: the first of the taxanes, an important new class of
Pharmacokinetics of a new anticancer drug, navelbine, in antitumor agents. Semin Oncol 1992; 19:646–662.
patients. Comparative study of radioimmunologic and 275. Rowinsky EK, Burke PJ, Karp JE, et al: Phase I and
radioactive determination methods. Cancer Chemother pharmacodynamic study of taxol in refractory acute
Pharmacol 1989; 23:247–251. leukemias. Cancer Res 1989; 49:4640–4647.
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 83

276. Rowinsky EK, McGuire WP, Guarnieri T, et al: Cardiac 293. Bissett D, Setanoians A, Cassidy J, et al: Phase I and
disturbances during the administration of taxol. J Clin pharmacokinetic study of taxotere (RP 56976) administered
Oncol 1991; 9:1704–1712. as a 24-hour infusion. Cancer Res 1993; 53:523–527.
277. Arbuck SG, Strauss H, Rowinsky E,et al: A reassessment 294. Pazdur R, Newman RA, Newman BM, et al: Phase I trial
of cardiac toxicity associated with Taxol. J Natl Cancer of Taxotere: five-day schedule. J Natl Cancer Inst 1992;
Inst Monogr 1993:117–130. 84:1781–1788.
278. Schiller JH, Storer B, Tutsch K, et al: Phase I trial of 295. Beer TM, Eilers KM, Garzotto M, et al: Weekly
3-hour infusion of paclitaxel with or without granulocyte high-dose calcitriol and docetaxel in metastatic
colony-stimulating factor in patients with advanced androgen-independent prostate cancer. J Clin Oncol
cancer. J Clin Oncol 1994; 12:241–248. 2003; 21:123–128.
279. Eisenhauer EA, ten Bokkel Huinink WW, Swenerton 296. Petrylak DP, Macarthur RB, O’Connor J, et al:
KD, et al: European-Canadian randomized trial of Phase I trial of docetaxel with estramustine in
paclitaxel in relapsed ovarian cancer: high-dose versus androgen-independent prostate cancer. J Clin Oncol
low-dose and long versus short infusion. J Clin Oncol 1999; 17:958–967.
1994; 12:2654–2666. 297. Savarese D, Taplin ME, Halabi S, et al: A phase II study
280. Rowinsky EK, Wright M, Monsarrat B, Lesser GJ, of docetaxel (Taxotere), estramustine, and low-dose
Donehower RC: Taxol: pharmacology, metabolism and hydrocortisone in men with hormone-refractory prostate
clinical implications. Cancer Surv 1993; 17:283–304. cancer: preliminary results of cancer and leukemia group
281. Huizing MT, Keung AC, Rosing H, et al: Pharmaco- B Trial 9780. Semin Oncol 1999; 26:39–44.
kinetics of paclitaxel and metabolites in a randomized 298. Petrylak DP: Docetaxel (Taxotere) in hormone-
comparative study in platinum-pretreated ovarian cancer refractory prostate cancer. Semin Oncol 2000; 27:24–29.
patients. J Clin Oncol 1993; 11:2127–2135. 299. Calabro F, Sternberg CN: High-risk metastatic
282. Monsarrat B, Alvinerie P, Wright M, et al: Hepatic urothelial cancer: chances for cure?, Curr Opin Urol
metabolism and biliary excretion of Taxol in rats and 2002; 12:441–448.
humans. J Natl Cancer Inst Monogr 1993:39–46. 300. Bruntsch U, Heinrich B, Kaye SB, et al: Docetaxel
283. Rowinsky EK, Gilbert MR, McGuire WP, et al: (Taxotere) in advanced renal cell cancer. A phase II trial
Sequences of taxol and cisplatin: a phase I and of the EORTC Early Clinical Trials Group. Eur J
pharmacologic study. J Clin Oncol 1991; 9:1692–1703. Cancer.1994; 30A:1064–1067.
284. Gianni L, Kearns CM, Giani A, et al: Nonlinear 301. Gunnarsson PO, Andersson SB, Johansson SA, Nilsson
pharmacokinetics and metabolism of paclitaxel and its T, Plym-Forshell G: Pharmacokinetics of estramustine
pharmacokinetic/pharmacodynamic relationships in phosphate (Estracyt) in prostatic cancer patients. Eur J
humans. J Clin Oncol 1995; 13:180–190. Clin Pharmacol.1984; 26:113–119.
285. Motzer RJ, Bajorin DF, Schwartz LH, et al: Phase II trial 302. Gunnarsson PO, Davidsson T, Andersson SB, Backman
of paclitaxel shows antitumor activity in patients with C, Johansson SA: Impairment of estramustine phosphate
previously treated germ cell tumors. J Clin Oncol 1994; absorption by concurrent intake of milk and food. Eur J
12:2277–2283. Clin Pharmacol 1990; 38:189–193.
286. Roth BJ, Dreicer R, Einhorn LH, et al: Significant 303. Walz PH, Bjork P, Gunnarsson PO, Edman K, Hartley-
activity of paclitaxel in advanced transitional-cell Asp B: Differential uptake of estramustine phosphate
carcinoma of the urothelium: a phase II trial of the metabolites and its correlation with the levels of
Eastern Cooperative Oncology Group. J Clin Oncol estramustine binding protein in prostate tumor tissue.
1994; 12:2264–2270. Clin Cancer Res 1998; 4:2079–2084.
287. Einzig AI, Gorowski E, Sasloff J, Wiernik PH: Phase II 304. Smith PH, Suciu S, Robinson MR, et al: A comparison
trial of taxol in patients with metastatic renal cell of the effect of diethylstilbestrol with low dose
carcinoma. Cancer Invest 1991; 9:133–136. estramustine phosphate in the treatment of advanced
288. Roth BJ, Yeap BY, Wilding G, et al: Taxol in advanced, prostatic cancer: final analysis of a phase III trial of the
hormone-refractory carcinoma of the prostate. A phase European Organization for Research on Treatment of
II trial of the Eastern Cooperative Oncology Group. Cancer. J Urol 1986; 136:619–623.
Cancer 1993; 72:2457–2460. 305. Hudes G, Einhorn L, Ross E, et al: Vinblastine versus
289. Kang MH, Figg WD, Dahut W: Taxanes in vinblastine plus oral estramustine phosphate for patients
hormone-refractory prostate cancer. Cancer Pract with hormone-refractory prostate cancer: A Hoosier
1999; 7:270–272. Oncology Group and Fox Chase Network phase III trial.
290. Ringel I, Horwitz SB: Studies with RP 56976 (taxotere): J Clin Oncol 1999; 17:3160–3166.
a semisynthetic analogue of taxol. J Natl Cancer Inst 306. Bracarda S, Tonato M, Rosi P, et al: Oral estramustine
1991; 83:288–291. and cyclophosphamide in patients with metastatic
291. Tomiak E, Piccart MJ, Kerger J, et al: Phase I study of hormone refractory prostate carcinoma: a phase II study.
docetaxel administered as a 1-hour intravenous infusion Cancer 2000; 88:1438–1444.
on a weekly basis. J Clin Oncol 1994; 12:1458–1467. 307. Sumiyoshi Y, Hashine K, Nakatsuzi H, Yamashita Y,
292. Burris H, Irvin R, Kuhn J, et al: Phase I clinical trial of Karashima T: Oral estramustine phosphate and oral
taxotere administered as either a 2-hour or 6-hour etoposide for the treatment of hormone-refractory
intravenous infusion. J Clin Oncol 1993; 11:950–958. prostate cancer. Int J Urol 2000; 7:243–247.
84 Part I Principles of Urologic Oncology

308. Sangrajrang S, Denoulet P, Millot G, et al: Estramustine 324. Motzer RJ, Sheinfeld J, Mazumdar M, et al: Paclitaxel,
resistance correlates with tau over-expression in human ifosfamide, and cisplatin second-line therapy for patients
prostatic carcinoma cells. Int J Cancer 1998; 77:626–631. with relapsed testicular germ cell cancer. J Clin Oncol
309. Williams JF, Muenchen HJ, Kamradt JM, Korenchuk S, 2000; 18:2413–2418.
Pienta KJ: Treatment of androgen-independent prostate 325. Loehrer PJ, Williams SD, Nichols CR, Einhorn LH:
cancer using antimicrotubule agents docetaxel and Clinical trials with ifosfamide: the Indiana University
estramustine in combination: an experimental study. experience. Semin Oncol 1992; 19:35–39.
Prostate 2000; 44:275–278. 326. Roth BJ: Ifosfamide in the treatment of bladder cancer.
310. Stein CA: Mechanisms of action of taxanes in prostate Semin Oncol 1996; 23:50–55.
cancer. Semin Oncol 1999; 26:3–7. 327. Bajorin DF, McCaffrey JA, Hilton S, et al: Treatment of
311. Weitzman A, Shelton G, Zuech N, et al: Phase II study patients with transitional-cell carcinoma of the urothelial
of estramustine combined with docetaxel in patients with tract with ifosfamide, paclitaxel, and cisplatin: a phase II
androgen-independent prostate cancer. Proc Am Soc trial. J Clin Oncol 1998; 16:2722–2727.
Clin Oncol, Atlanta, GA, 1999, p A1369. 328. Bajorin DF, McCaffrey JA, Dodd PM, et al: Ifosfamide,
312. Kreis W Budman D:. Daily oral estramustine and paclitaxel, and cisplatin for patients with advanced
intermittent intravenous docetaxel (Taxotere) as transitional cell carcinoma of the urothelial tract: final
chemotherapeutic treatment for metastatic, hormone- report of a phase II trial evaluating two dosing schedules.
refractory prostate cancer. Semin Oncol 1999; 26:34–38. Cancer 2000; 88:1671–1678.
313. Smith DC, Esper P, Strawderman M, Redman B,Pienta 329. Cohen NA, Egorin MJ, Snyder SW, et al: Interaction of
KJ: Phase II trial of oral estramustine, oral etoposide, N,N′,N′ ′-triethylenethiophosphoramide and N,N′,
and intravenous paclitaxel in hormone-refractory N′ ′-triethylenephosphoramide with cellular DNA.
prostate cancer. J Clin Oncol 1999; 17:1664–1671. Cancer Res 1991; 51:4360–4366.
314. Weitzman AL, Shelton G, Zuech N, et al: 330. Cohen BE, Egorin MJ, Kohlhepp EA, Aisner J,
Dexamethasone does not significantly contribute Gutierrez PL: Human plasma pharmacokinetics and
to the response rate of docetaxel and estramustine in urinary excretion of thiotepa and its metabolites. Cancer
androgen independent prostate cancer. J Urol 2000; Treat Rep 1986; 70:859–864.
163:834–837. 331. Wolff SN, Herzig RH, Fay JW, et al: High-dose
315. Kosty MP, Ferreira A, Bryntesen T, Grossman J: Weekly thiotepa with autologous bone marrow transplantation
docetaxel and low-dose estramustine phosphate in for metastatic malignant melanoma: results of phase I
hormone refractory prostate cancer: a phase II study. and II studies of the North American Bone Marrow
Proc Am Soc Clin Oncol, New Orleans, LA, 2000, Transplantation Group. J Clin Oncol 1989; 7:245–249.
p A1442. 332. Vogelzang NJ, Raghavan D, Kennedy BJ: VP-16-213
316. Connors TA, Cox PJ, Farmer PB, Foster AB, Jarman M: (etoposide): the mandrake root from Issyk-Kul. Am J
Some studies of the active intermediates formed in the Med 1982; 72:136–144.
microsomal metabolism of cyclophosphamide and 333. Creaven PJ: The clinical pharmacology of VM26 and
isophosphamide. Biochem Pharmacol 1974; 23: VP16-213. A brief overview. Cancer Chemother
115–129. Pharmacol 1982; 7:133–140.
317. Dorr RT, Alberts DS: Cimetidine enhancement of 334. Slevin ML:. The clinical pharmacology of etoposide.
cyclophosphamide antitumour activity. Br J Cancer 1982; Cancer 1991; 67:319–329.
45:35–43. 335. Krishan A, Paika K, Frei E, III: Cytofluorometric studies
318. Dorr RT, Soble MJ, Alberts DS: Interaction of on the action of podophyllotoxin and
cimetidine but not ranitidine with cyclophosphamide in epipodophyllotoxins (VM-26, VP-16-213) on the cell
mice. Cancer Res 1986; 46:1795–1799. cycle traverse of human lymphoblasts. J Cell Biol 1975;
319. Motzer RJ, Bajorin DF, Bosl GJ:. “Poor-risk” germ cell 66:521–530.
tumors: current progress and future directions. Semin 336. Yang L, Rowe TC, Liu LF: Identification of DNA
Oncol 1992; 19:206–214. topoisomerase II as an intracellular target of antitumor
320. Cerny T, Margison JM, Thatcher N, Wilkinson PM: epipodophyllotoxins in simian virus 40-infected monkey
Bioavailability of ifosfamide in patients with bronchial cells. Cancer Res 1985; 45:5872–5876.
carcinoma. Cancer Chemother Pharmacol 1986; 337. Jolivet J, Schilsky RL, Bailey BD, Drake JC, Chabner
18:261–264. BA: Synthesis, retention, and biological activity of
321. Anderson NR, Tandon DS: Ifosfamide extrapyramidal methotrexate polyglutamates in cultured human breast
neurotoxicity. Cancer 1991; 68:72–75. cancer cells. J Clin Invest 1982; 70:351–360.
322. Skinner R, Pearson AD, English MW, et al: Risk factors 338. Chu E, Drake JC, Boarman D, Baram J, Allegra CJ:
for ifosfamide nephrotoxicity in children. Lancet 1996; Mechanism of thymidylate synthase inhibition by
348:578–580. methotrexate in human neoplastic cell lines and normal
323. McCaffrey JA, Mazumdar M, Bajorin DF, et al: human myeloid progenitor cells. J Biol Chem 1990;
Ifosfamide- and cisplatin-containing chemotherapy as 265:8470–8478.
first-line salvage therapy in germ cell tumors: response 339. Borchers AH, Kennedy KA, Straw JA: Inhibition of
and survival. J Clin Oncol 1997; 15:2559–2563. DNA excision repair by methotrexate in Chinese
hamster ovary cells following exposure to ultraviolet
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 85

irradiation or ethylmethanesulfonate. Cancer Res 1990; of action of 5-fluorouracil. Cancer Res 1981;
50:1786–1789. 41:3288–3295.
340. Chu E, Mota AC, Fogarasi MC: Antimetabolites. In 356. Baker SD, Diasio RB, O’Reilly S, et al: Phase I and
DeVita VTJ, Hellman S, Rosenberg SA (eds): Cancer: pharmacologic study of oral fluorouracil on a chronic
Principles and Practice of Oncology, 6th edition, pp daily schedule in combination with the
388–415. Philadelphia, Lippincott, Williams and dihydropyrimidine dehydrogenase inactivator eniluracil.
Wilkins, 2001. J Clin Oncol 2000; 18:915–926.
341. Huffman DH, Wan SH, Azarnoff DL, Hogstraten B:. 357. Khor SP, Amyx H, Davis ST, et al: Dihydropyrimidine
Pharmacokinetics of methotrexate. Clin Pharmacol Ther dehydrogenase inactivation and 5-fluorouracil
1973; 14:572–579. pharmacokinetics: allometric scaling of animal data,
342. Stoller RG, Hande KR, Jacobs SA, Rosenberg SA, pharmacokinetics and toxicodynamics of 5-fluorouracil
Chabner BA: Use of plasma pharmacokinetics to predict in humans. Cancer Chemother Pharmacol 1997;
and prevent methotrexate toxicity. N Engl J Med 1977; 39:233–238.
297:630–634. 358. Schilsky RL, Hohneker J, Ratain MJ, et al: Phase I
343. Stoller RG, Kaplan HG, Cummings FJ, Calabresi P: A clinical and pharmacologic study of eniluracil plus
clinical and pharmacological study of high-dose fluorouracil in patients with advanced cancer. J Clin
methotrexate with minimal leucovorin rescue. Cancer Oncol 1998; 16:1450–1457.
Res 1979; 39:908–912. 359. McGavin JK, Goa KL: Capecitabine: a review of its use
344. Conaghan PG, Quinn DI, Brooks PM, Day RO: in the treatment of advanced or metastatic colorectal
Hazards of low dose methotrexate. Aust N Z J Med cancer. Drugs 2001; 61:2309–2326.
1995; 25:670–673. 360. Van Cutsem E, Twelves C, Cassidy J, et al: Oral
345. Heidelberger C, Chaudhuri NK, Dannenberg P, et al: capecitabine compared with intravenous fluorouracil plus
Fluorinated pyrimidines: a new class of tumor inhibitory leucovorin in patients with metastatic colorectal cancer:
compounds. Nature 1957; 179:663. results of a large phase III study. J Clin Oncol 2001;
346. Diasio RB, Harris BE: Clinical pharmacology of 19:4097–4106.
5-fluorouracil. Clin Pharmacokinet 1989; 16:215–237. 361. Hoff PM, Cassidy J, Schmoll HJ: The evolution of
347. Salonga D, Danenberg KD, Johnson M, et al: Colorectal fluoropyrimidine therapy: from intravenous to oral.
tumors responding to 5-fluorouracil have low gene Oncologist 2001; 6(Suppl 4):3–11.
expression levels of dihydropyrimidine dehydrogenase, 362. Wenzel C, Locker GJ, Schmidinger M, et al:
thymidylate synthase, and thymidine phosphorylase. Clin Capecitabine in the treatment of metastatic renal cell
Cancer Res 2000; 6:1322–1327. carcinoma failing immunotherapy. Am J Kidney Dis
348. Mizutani Y, Wada H, Ogawa O, et al: Prognostic 2002; 39:48–54.
significance of thymidylate synthase activity in bladder 363. Oevermann K, Buer J, Hoffmann R, et al: Capecitabine
carcinoma. Cancer 2001; 92:510–518. in the treatment of metastatic renal cell carcinoma. Br J
349. Bruckner HW, Creasey WA: The administration of Cancer 2000; 83:583–587.
5-fluorouracil by mouth. Cancer 1974; 33:14–18. 364. Keefe DL, Roistacher N, Pierri MK: Clinical
350. Cano JP, Rigault JP, Aubert C, Carcassonne Y, Seitz JF: cardiotoxicity of 5-fluorouracil. J Clin Pharmacol 1993;
Determination of 5-fluorouracil in plasma by GC/MS 33:1060–1070.
using an internal standard. Applications to 365. Hertel LW, Boder GB, Kroin JS, et al: Evaluation of the
pharmacokinetics. Bull Cancer 1979; 66:67–74. antitumor activity of gemcitabine (2′,2′-difluoro-2′-
351. Myers CE, Young RC, Chabner BA: Biochemical deoxycytidine). Cancer Res 1990; 50:4417–4422.
determinants of 5-fluorouracil response in vivo. The role 366. Heinemann V, Hertel LW, Grindey GB, Plunkett W:
of deoxyuridylate pool expansion. J Clin Invest 1975; Comparison of the cellular pharmacokinetics and toxicity
56:1231–1238. of 2′,2′ -difluorodeoxycytidine and 1-beta-D-
352. Creaven PJ, Rustum YM, Petrelli NJ, et al: Phase I and arabinofuranosylcytosine. Cancer Res 1988;
pharmacokinetic evaluation of floxuridine/leucovorin 48:4024–4031.
given on the Roswell Park weekly regimen. Cancer 367. Heinemann V, Xu YZ, Chubb S, et al: Inhibition of
Chemother Pharmacol 1994; 34:261–265. ribonucleotide reduction in CCRF-CEM cells by 2′,2′-
353. Meropol NJ, Creaven PJ, Petrelli NJ: Metastatic difluorodeoxycytidine. Mol Pharmacol 1990; 38:567-572.
colorectal cancer: advances in biochemical modulation 368. Heinemann V, Schulz L, Issels RD, Plunkett W:
and new drug development. Semin Oncol 1995; Gemcitabine: a modulator of intracellular nucleotide and
22:509–524. deoxynucleotide metabolism. Semin Oncol 1995;
354. Petrelli N, Herrera L, Rustum Y, et al: A prospective 22:11–18.
randomized trial of 5-fluorouracil versus 5-fluorouracil 369. Neff T, Blau CA: Forced expression of cytidine
and high-dose leucovorin versus 5-fluorouracil and deaminase confers resistance to cytosine arabinoside and
methotrexate in previously untreated patients with gemcitabine. Exp Hematol 1996; 24:1340–1346.
advanced colorectal carcinoma. J Clin Oncol 1987; 370. Goan YG, Zhou B, Hu E, Mi S, Yen Y: Overexpression
5:1559–1565. of ribonucleotide reductase as a mechanism of resistance
355. Evans RM, Laskin JD, Hakala MT: Effect of excess to 2,2-difluorodeoxycytidine in the human KB cancer
folates and deoxyinosine on the activity and site cell line. Cancer Res 1999; 59:4204–4207.
86 Part I Principles of Urologic Oncology

371. Dumontet C, Fabianowska-Majewska K, Mantincic D, advanced renal cancer. Cancer Chemother Pharmacol
et al: Common resistance mechanisms to deoxynucleoside 1996; 37:491–495.
analogues in variants of the human erythroleukaemic line 386. Rini BI, Vogelzang NJ, Dumas MC, et al: Phase II trial
K562. Br J Haematol 1999; 106:78–85. of weekly intravenous gemcitabine with continuous
372. Ruiz van Haperen VW, Peters GJ: New targets for infusion fluorouracil in patients with metastatic renal cell
pyrimidine antimetabolites for the treatment of solid cancer. J Clin Oncol 2000; 18:2419–2426.
tumours. 2: Deoxycytidine kinase. Pharm World Sci 387. Morant R, Bernhard J, Maibach R, et al: Response and
1994; 16:104–112. palliation in a phase II trial of gemcitabine in hormone-
373. Gandhi V, Legha J, Chen F, Hertel LW, Plunkett W: refractory metastatic prostatic carcinoma. Swiss Group
Excision of 2′,2′ -difluorodeoxycytidine (gemcitabine) for Clinical Cancer Research (SAKK). Ann Oncol 2000;
monophosphate residues from DNA. Cancer Res 1996; 11:183–188.
56:4453–4459. 388. Hinton S, Catalano P, Einhorn LH, et al: Phase II study
374. Abbruzzese JL, Grunewald R, Weeks EA, et al: A phase I of paclitaxel plus gemcitabine in refractory germ cell
clinical, plasma, and cellular pharmacology study of tumors (E9897): a trial of the Eastern Cooperative
gemcitabine. J Clin Oncol 1991; 9:491–498. Oncology Group. J Clin Oncol 2002; 20:1859–1863.
375. von der Maase H, Hansen SW, Roberts JT, et al: 389. Bokemeyer C, Gerl A, Schoffski P, et al: Gemcitabine in
Gemcitabine and cisplatin versus methotrexate, patients with relapsed or cisplatin-refractory testicular
vinblastine, doxorubicin, and cisplatin in advanced or cancer. J Clin Oncol 1999; 17:512–516.
metastatic bladder cancer: results of a large, randomized, 390. Wall ME, Wani MC, Taylor H: Isolation and chemical
multinational, multicenter, phase III study,. J Clin Oncol characterization of antitumor agents from plants. Cancer
2000; 18:3068–3077. Treat Rep 1976; 60:1011–1030.
376. O’Rourke TJ, Brown TD, Havlin K, et al: Phase I 391. Gottlieb JA, Guarino AM, Call JB, Oliverio VT, Block
clinical trial of gemcitabine given as an intravenous bolus JB: Preliminary pharmacologic and clinical evaluation of
on 5 consecutive days. Eur J Cancer 1994; 30A:417–418. camptothecin sodium (NSC-100880). Cancer
377. Touroutoglou N, Gravel D, Raber MN, Plunkett W, Chemother Rep 1970; 54:461–470.
Abbruzzese JL: Clinical results of a 392. Muggia FM, Creaven PJ, Hansen HH, Cohen MH,
pharmacodynamically-based strategy for higher dosing of Selawry OS: Phase I clinical trial of weekly and daily
gemcitabine in patients with solid tumors. Ann Oncol treatment with camptothecin (NSC-100880): correlation
1998; 9:1003–1008. with preclinical studies. Cancer Chemother Rep 1972;
378. Gandhi V, Plunkett W, Du M, Ayres M, Estey EH: 56:515–521.
Prolonged infusion of gemcitabine: clinical and 393. Moertel CG, Schutt AJ, Reitemeier RJ, Hahn RG: Phase
pharmacodynamic studies during a phase I trial in II study of camptothecin (NSC-100880) in the treatment
relapsed acute myelogenous leukemia. J Clin Oncol of advanced gastrointestinal cancer. Cancer Chemother
2002; 20:665–673. Rep 1972; 56:95–101.
379. Grunewald R, Abbruzzese JL, Tarassoff P, Plunkett W: 394. Hsiang YH, Hertzberg R, Hecht S, Liu LF:
Saturation of 2′ ,2′-difluorodeoxycytidine 5′-triphosphate Camptothecin induces protein-linked DNA breaks via
accumulation by mononuclear cells during a phase I trial mammalian DNA topoisomerase I. J Biol Chem 1985;
of gemcitabine. Cancer Chemother Pharmacol 1991; 260:14873–14878.
27:258–262. 395. Hsiang YH, Liu LF: Identification of mammalian DNA
380. Grunewald R, Kantarjian H, Du M, et al: Gemcitabine topoisomerase I as an intracellular target of the
in leukemia: a phase I clinical, plasma, and cellular anticancer drug camptothecin. Cancer Res 1988;
pharmacology study. J Clin Oncol 1992; 10:406–413. 48:1722–1726.
381. Patel SR, Gandhi V, Jenkins J, et al: Phase II clinical 396. Hsiang YH, Lihou MG, Liu LF: Arrest of replication
investigation of gemcitabine in advanced soft tissue forks by drug-stabilized topoisomerase I-DNA cleavable
sarcomas and window evaluation of dose rate on complexes as a mechanism of cell killing by
gemcitabine triphosphate accumulation. J Clin Oncol camptothecin. Cancer Res 1989; 49:5077–5082.
2001; 19:3483–3489. 397. Hsiang YH, Liu LF, Wall ME, et al: DNA
382. Pollera CF, Ceribelli A, Crecco M, Calabresi F: Weekly topoisomerase I-mediated DNA cleavage and
gemcitabine in advanced bladder cancer: a preliminary cytotoxicity of camptothecin analogues. Cancer Res
report from a phase I study. Ann Oncol 1994; 5:182–184. 1989; 49:4385–4389.
383. Stadler WM, Kuzel T, Roth B, Raghavan D, Dorr FA: 398. Liu LF: DNA topoisomerase poisons as antitumor drugs.
Phase II study of single-agent gemcitabine in previously Annu Rev Biochem 1989; 58:351–375.
untreated patients with metastatic urothelial cancer. 399. Hertzberg RP, Busby RW, Caranfa MJ, et al: Irreversible
J Clin Oncol 1997; 15:3394–3398. trapping of the DNA-topoisomerase I covalent complex.
384. Kaufman D, Raghavan D, Carducci M, et al: Phase II Affinity labeling of the camptothecin binding site. J Biol
trial of gemcitabine plus cisplatin in patients with Chem 1990; 265:19287–19295.
metastatic urothelial cancer. J Clin Oncol 2000; 400. Li XG, Haluska P, Jr, Hsiang YH, et al: Identification of
18:1921–1927. topoisomerase I mutations affecting both DNA cleavage
385. De Mulder PH, Weissbach L, Jakse G, Osieka R, Blatter and interaction with camptothecin. Ann N Y Acad Sci
J: Gemcitabine: a phase II study in patients with 1996; 803:111–127.
Chapter 4 Principles of Chemotherapy for Genitourinary Cancer 87

401. Hertzberg RP, Caranfa MJ, Holden KG, et al: 407. Kaneda N, Nagata H, Furuta T, Yokokura T:
Modification of the hydroxy lactone ring of Metabolism and pharmacokinetics of the camptothecin
camptothecin: inhibition of mammalian topoisomerase analogue CPT-11 in the mouse. Cancer Res 1990;
I and biological activity. J Med Chem 1989; 32: 50:1715–1720.
715–720. 408. Rowinsky EK, Grochow LB, Hendricks CB, et al: Phase
402. Slichenmyer WJ, Rowinsky EK, Donehower RC, I and pharmacologic study of topotecan: a novel
Kaufmann SH: The current status of camptothecin topoisomerase I inhibitor. J Clin Oncol 1992;
analogues as antitumor agents. J Natl Cancer Inst 1993; 10:647–656.
85:271–291. 409. Saltz L, Sirott M, Young C, et al: Phase I clinical and
403. Negoro S, Fukuoka M, Masuda N, et al: Phase I study of pharmacology study of topotecan given daily for 5
weekly intravenous infusions of CPT-11, a new consecutive days to patients with advanced solid tumors,
derivative of camptothecin, in the treatment of advanced with attempt at dose intensification using recombinant
non-small-cell lung cancer. J Natl Cancer Inst 1991; granulocyte colony-stimulating factor. J Natl Cancer Inst
83:1164–1168. 1993; 85:1499–1507.
404. Rowinsky EK, Grochow LB, Ettinger DS, et al: Phase I 410. Haas NB, LaCreta FP, Walczak J, et al: Phase
and pharmacological study of the novel topoisomerase I I/pharmacokinetic study of topotecan by 24-hour
inhibitor 7-ethyl-10-[4-(1-piperidino)-1- continuous infusion weekly. Cancer Res 1994;
piperidino]carbonyloxycamptothecin (CPT-11) 54:1220–1226.
administered as a ninety-minute infusion every 3 weeks. 411. Hochster H, Liebes L, Speyer J, et al: Phase I trial of
Cancer Res 1994; 54:427–436. low-dose continuous topotecan infusion in patients with
405. Abigerges D, Armand JP, Chabot GG, et al: Irinotecan cancer: an active and well-tolerated regimen. J Clin
(CPT-11) high-dose escalation using intensive high-dose Oncol 1994; 12:553–559.
loperamide to control diarrhea. J Natl Cancer Inst 1994; 412. Hochster H, Liebes L, Speyer J, et al: Effect of
86:446–449. prolonged topotecan infusion on topoisomerase 1 levels:
406. Abigerges D, Chabot GG, Armand JP, et al: Phase I and a phase I and pharmacodynamic study. Clin Cancer Res
pharmacologic studies of the camptothecin analog 1997; 3: 1245–1252.
irinotecan administered every 3 weeks in cancer patients.
J Clin Oncol 1995; 13:210–221.
C H A P T E R

5 Immunotherapy: Basic Guidelines


Jason B. Wynberg, MD, FRCSC, W. Marston Linehan, MD,
and Richard Childs, MD

Harnessing the power of the immune system against binds to IL-2 receptors expressed on their cell surfaces.
malignant cells is often thought of as a relatively modern Administration of IL-2 at pharmacologic doses, as is
treatment modality for patients with advanced cancer. done in the treatment of metastatic renal cell carcinoma
However, more than 100 years have passed since the first (RCC), appears to directly interrupt this autocrine
report was published by Dr. W. Coley documenting pathway.6,7
immune-mediated disease regression against cancer fol- The field of cytokine therapeutics has grown dramat-
lowing injections of bacterial toxins directly into tumor ically since the development of recombinant DNA tech-
lesions.1 Since then, investigators have gained tremen- nology in the 1980s, which enabled cytokines to be
dous insight into the mechanisms by which tumors are produced in large quantities.8–10 A discussion of the
able to evade the innate immune system2–4 (Table 5-1) cytokines commonly administered in the setting of
and remain resistant to “conventional” cancer advanced urologic malignancy is presented below.
immunotherapies. The steady expansion in our knowl-
edge of tumor biology over the past few decades is grad-
Interleukin-2 and Interferon-a
ually being translated into improvements in the efficacy
Background
and safety of cancer immunotherapeutics.
In 1992, the Food and Drug Administration (FDA)
approved high-dose IL-2 for the treatment of metastatic
CYTOKINE THERAPY
RCC.11 Although the precise mechanism accounting for
Cytokines are protein molecules produced and secreted tumor regression in RCC patients treated with IL-2 is
by immune and inflammatory cells that bind to comple- not known,12 many in vitro effects of IL-2 on immune
mentary cytokine receptors resulting in either the stimu- effector populations have been characterized (Table
lation or inhibition of immune cells. Cytokines usually 5-2).13,14 Whereas IL-2 and resting lymphocytes sepa-
act in an autocrine or paracrine fashion, in contrast to rately fail to kill RCC tumor cells, prior incubation of the
hormones, which typically act at a distance from their same lymphocytes with IL-2 significantly augments their
cells of origin. Following the binding of the cytokine to in vitro tumor cytotoxicity. These observations provided
its receptor target, cell signaling is initiated through some of the first preclinical evidence that the immune
intracellular pathways, such as the Jak-STAT tyrosine system could mediate antitumor effects, providing theo-
kinase, Src, Zap70 and related proteins, phosphatidyli- retical grounds for the pursuit of immune-based cancer
nositol 3-kinase, IRS-1, IRIS-2, and phosphatases.5 therapy in humans. Unlike the interleukins, IFNs are a
Cytokines that typically up-regulate immune responses family of proteins secreted by leukocytes in response to
include interleukin-2 (IL-2) and interferon (IFN), whereas viral infection and other antigenic stimuli. IFNs have
cytokines that typically down-regulate immune responses powerful antiproliferative and immunoregulatory activity
include transforming growth factor β, interleukin-6, and (Table 5-3),13 such as up-regulation of class I and class II
interleukin-10. Through their secretion of immunosup- major histocompatibility complex (MHC) molecules.15
pressive cytokines (e.g., transforming growth factor β), When administered in a therapeutic setting, IFN-α
certain tumors are able to directly impair the hosts’ is typically given as a subcutaneous injection. Due to its
immune defenses. Interestingly, some tumors can stimu- relatively short half-life, IFN-α is most commonly
late their own proliferation by secreting IL-2, which administered at least three times per week. Recently,

88
Chapter 5 Immunotherapy: Basic Guidelines 89

Table 5-1 Mechanisms of Tumor Escape from Immune System


Tumor-related Host-related

Decreased tumor Ag expression Antigen-specific suppressor T cells


Decreased MHC class I expression Deficient presentation of tumor antigens by host antigen-presenting cells
Failure to express immune costimulatory Failure of host effectors to reach the tumor (e.g., stromal barrier)
molecules (e.g., B7.1)
Production of immune inhibitors (e.g., TGFβ, Immune dysfunction due to carcinogen, infections, age
IL-6, IL-10, free tumor Ag)
Tumor antigens weakly immunogenic
Induction of T cell apoptosis by tumor Development of T cell anergy or tolerance to tumor antigens
expression of Fas ligand (FasL)

MHC, major histocompatibility complex; TGFβ, transforming growth factor β; Ag, antigen.

Table 5-2 Effects of Interleukin-2 Cytokine Therapy as Treatment of Metastatic


Proliferation of T cells, B cells, NK cells, and monocytes
Renal Cell Carcinoma
Despite early enthusiasm based on favorable outcome of
Potentiation of Fas-mediated apoptosis of T cells to
pilot clinical trials, neither IL-2 nor IFN-α has proven to
prevent clonal persistence
be a panacea for the treatment of metastatic RCC (Table
Induction of antibody synthesis by B cells 5-4).17–30 Unfortunately, the great majority of patients
treated with either cytokine fail to respond, with median
No direct anti-tumor activity survival typically being 2 years or less.
Despite their overall low response rate, however, some
patients clearly benefit from treatment with IL-2 and
IFN-α. Partial responses, defined as a ≥50% reduction in
Table 5-3 Effects of Interferon-alfa the sum of the products of maximal perpendicular diam-
Direct antiproliferative effects on tumor and other tissues
eters of all measurable metastatic lesions, occur in
approximately 15% of patients treated with either IL-2
Antiviral activity or IFN-α (see Table 5-4). Although partial responses can
be associated with significant disease palliation, long-
Occasionally promotes partial reversal of the malignant term survival in partial responders is a rare event.
phenotype Among patients who achieve a complete response
Increases expression of MHC molecules and tumor- (defined as the complete disappearance of all evidence of
associated Ags metastatic disease for at least 1 month) to high-dose IL-
2 therapy, 60% to 90% remain alive and free of disease
Activates T cells, augments NK cell function for 8–10 years after treatment. In contrast, virtually no
evidence exists of long-term survival following IFN-
Upregulates macrophage antigen presentation
based therapy (see Table 5-4). It is, therefore, not sur-
Increases macrophage production of angiogenesis-inhibitor prising that many urologists and oncologists consider
high-dose IL-2 to be superior to IFN-α in the treatment
MHC, major histocompatibility complex; NK, natural killer; Ag, of metastatic RCC.
antigen.
Nonetheless, many oncologists continue to treat
metastatic RCC patients with IFN-α, largely due to con-
cerns regarding the substantial toxicities associated with
high-dose IL-2. Toxicities associated with high-dose
investigators have shown that the serum half-life of IFN- IL-2 therapy are primarily the consequence of a cascade
α-2b can be extended dramatically by covalently linking of cytokines being released from circulating leukocytes
polyethylene glycol (PEG) to its histidine-34 moiety, following drug exposure. This cytokine shower can sig-
thus making dosing schedules more convenient16. IFNs nificantly increase capillary permeability, leading to dra-
may have direct antitumor effects against a variety of matic fluid shifts, reduced peripheral vascular resistance,
malignancies, including hairy cell leukemia, cervical and sometimes profound hypotension, renal failure, and
intraepithelial neoplasia, basal cell cancer, Kaposi’s sar- pulmonary edema. The severity of these symptoms is
coma, melanoma, renal cell carcinoma, multiple dose dependent. Toxicities related to IFN therapy are
myeloma, and chronic myelogenous leukemia.13 also dose and schedule dependent, and include primarily
90

Table 5-4 Treatment with Il-2 or IFN-α in Metastatic Clear Cell RCC
Prior Median
Nephrectomy Survival
References (%) N (Months) PR (%) CR (%) Long-Term Survival of CRs

High-dose IV IL-2 Fisher et al.17 85 255 16.3 8 7 >60% of CRs alive, NED at 10 years

Yang et al.18 93 156 18 14 7 73% of CRs alive, NED at median 9.3 years

Dutcher et al.19 86 71 15.5 11 7 90% of CRs alive, NED at >8 years20


Part I Principles of Urologic Oncology

Subcutaneous IL-2 Yang et al.18 93 150 18 9 4 50% of CRs alive, NED at median 10.1 years

Subcutaneous IL-2/IFN-α McDermott et al.21 75 94 NA 10 2 No long-term data available

Dutcher et al.22 87 47 20 13 4 One CR alive at 49 (+) months

Rogers et al.23 82 33 10 9 0 No long-term data available

Negrier et al.24 79 70 13 2 0 Not applicable—no CRs

Subcutaneous IFN Mickisch et al.25 100 42 17 7 12 No long-term data available

Flanigan et al.26 100 92 11.1 3 0 Not applicable—no CRs

Negrier et al.27 92 147 13 5 1 One CR alive, NED at 10 years28

Motzer et al.29 52 145 15 6 1 No long term data available

Gleave et al.30* 86 91 12 1 3 No long-term data available

IL-2, interleukin-2; IFN-α, interferon-alpha; RCC, renal cell carcinoma; N, number of patients; CR, complete response; PR, partial response; IV, intravenous; mos., months; NED, no evidence
of disease; NA, not available.
Note: No treatment-related mortalities were experienced in any of the above studies, except for Fisher et al.17, in which 4% of patients receiving high-dose IL-2 died of treatment-related
complications.
*IFN-γ rather than IFN-α was used in this study.
Chapter 5 Immunotherapy: Basic Guidelines 91

hematologic effects (i.e., bone marrow suppression and disease-free following a resection of limited recurrent
cytopenias) and flu-like symptoms.13,15,25 A large prelim- disease = total 91% alive and disease-free at a median of
inary series of patients given high-dose IL-2 at the 9.3 years) compared to the low-dose arm (50% alive and
National Cancer Institute reported a 4% treatment- disease-free at a median of 10.1 years) (Figure 5-1A).
related mortality rate.17 Because this mortality rate However, no difference in overall survival was observed
approached the complete response rate with high-dose between the high-dose and low-dose cohorts (Figure
IL-2, many oncologists still consider it unacceptable to 5-1B), again reflecting the unfortunate fact that complete
have one regimen-related death for every patient cured responders to any form of IL-2 therapy are in the signif-
with this therapy. icant minority.
Since its initial use, the mortality rate related to high- Another prospective randomized study compared low-
dose IL-2 therapy has dropped dramatically at the dose intravenous IL-2 (group 1) versus subcutaneous
National Cancer Institute, with no deaths among the last IFN-α-2a (group 2) versus both drugs combined
800+ patients treated. This improvement in outcome is (group 3) in patients with metastatic RCC.27 A total of
likely multi-factorial, owing to alterations in eligibility 425 patients were randomized between 1992 and 1995
criteria, the treatment regimen, and improvements in into one of three treatment groups. As with the previous
supportive care.31 Other groups similarly contend that trial, the majority of patients (>90%) enrolled in the
high-dose IL-2 can be given safely when patients are study had undergone a prior nephrectomy. Toxicities
carefully selected and treatment is given in a well- were most evident among those receiving IL-2 (groups 1
monitored setting.32,33 and 3), including 67% who became hypotensive and 50%
Despite improvements in morbidity related to high- who experienced high fevers. Importantly, all patients
dose IL-2 therapy, there has been considerable interest in ultimately recovered from these adverse events and
the development of low-dose IL-2 regimens in the hope returned to their pretreatment status. Response rates
of reducing drug-related complications. A randomized were 6.5%, 7.5%, and 18.6% (p < 0.01) for patients
study comparing high-dose versus low-dose IL-2 in the receiving IL-2, IFN-α-2a, and IL-2 plus IFN-α-2a,
setting of metastatic RCC has recently been completed.18 respectively. Although the response rate was higher
The vast majority of patients enrolled on this trial (93%) among those receiving both IL-2 and IFN-α, no long-
had previously undergone cytoreductive nephrectomy. term survival benefit was observed in this group
Although no regimen-related mortalities occurred in this (Figure 5-2).
study, toxicities were greater in the high-dose IL-2 arm Biochemotherapy is another field of research wherein
compared to the low-dose IL-2 arm, especially in terms chemotherapeutic drugs are administered concomitant
of clinically significant hypotensive episodes (36.4% ver- with biologic agents, such as IL-2, in hopes of improving
sus 2.9%, respectively). The overall response rate (CR + therapeutic indices. 5-Flurouracil (5-FU) has been the
PR) was higher in the high-dose versus the low-dose arm most widely used chemotherapeutic agent that has been
(21% versus 13%, p = 0.048). Importantly, the durability combined with cytokines in the setting of advanced
of responses among complete responders was superior in RCC, with response rates up to 30% in several small
the high-dose arm (73% disease-free + an additional 18% studies. However, the vast majority of responses are par-

1.0 1.0
0.9 0.9
High dose
0.8 0.8
Proportion surviving

Proportion surviving

0.7 0.7 High-dose versus low-dose


0.6 0.6 (p = 0.41)
0.5 0.5
0.4 Low dose
0.4
0.3 p2 = 0.04 0.3
High dose (fail/total = 117/155)
0.2 0.2
0.1 0.1
Low dose (fail/total = 121/150)
0.0 0.0
0 12 24 36 48 60 72 84 96 108 120 132 0 12 24 36 48 60 72 84 96 108 120 132
A Survival time in months B Survival time in months

Figure 5-1 A, Disease-free survival in patients achieving a complete response with high-dose
or low-dose IL-2. B, Survival by treatment received—high-dose or low-dose IL-2. (From Yang
JC, Sherry RM, Steinberg SM, et al: J Clin Oncol 2003; 21(16): 3127-3132, with permission.)
92 Part I Principles of Urologic Oncology

100 Interferon alfa-2a


90 Interleukin-2 + interferon alfa-2a
80 Interleukin-2

Overall survival (%)


70
60
50
40
30
20
10
0
0 6 12 18 24 30 36
Months after randomization
Kaplan-Meier curves for overall survival among patients in the three treatment groups.
Tick marks represent censored data on patiients who were alive or lost to follow-up. The results
shown are from an intention-to-treat analysis. p = 0.55 for the comparison among the groups.

Figure 5-2 Overall survival in patients treated with IFN-α-2a with or without IL-2. (Based on
Negrier S, Escudier B, Lasset C, et al: N Engl J Med 1998; 338(18):1272–1278, with
permission.)

tial only, with no data supporting long-term disease-free BCG-naïve patients and 53% for BCG-failure patients.
survival using this approach.34 Kaplan– Meier estimates for freedom-from-disease were
Reactive oxygen species (ROS) are released by tumor 71% and 61% for BCG-naïve patients and 53% and 40%
infiltrating monocytes and macrophages and inhibit for BCG-failure patients at 1 and 2 years, respectively.
cytokine-stimulated lymphocytes. Histamine dihy- The toxicity-related premature dropout rate among
drochloride is a biogenic amine that inhibits the formation BCG-naïve patients was 3.7% and among BCG-failure
of ROS and has been used in clinical trials as an adjuvant patients was 7.3%. This multicenter trial substantiates
to IL-2 and IFN-α with a view to reducing oxidative inhi- the earlier encouraging reports of the efficacy of combi-
bition of lymphocytes.35 Although clinical efficacy has nation BCG + IFN as upfront and salvage therapy for
been observed in melanoma patients,36 no benefit has yet patients with moderate to high-risk superficial bladder
been observed in the setting of advanced RCC.37 cancer.41–43 Longer follow-up will be needed to define
Tumor necrosis factor–related apoptosis-inducing lig- the ultimate role this cytokine will play in the treatment
and (TRAIL) is a trans-membrane protein expressed on of superficial bladder cancer.
immune cells that plays a primary role in natural killer
cell-mediated tumor surveillance. Based on its high
Colony-Stimulating Factors
specificity and cytotoxicity against tumor cells in vitro,
TRAIL remains as an active area of research.38–40 Both granulocyte colony-stimulating factor (G-CSF) and
granulocyte-macrophage CSF (GM-CSF) are recombi-
nant cytokines that stimulate the bone marrow to
Intravesical Interferon Therapy for the Treatment
increase production of neutrophils, monocytes,
of Superficial Bladder Cancer
eosinophils, and dendritic cells. Both cytokines are used
Intravesical IFN-α is currently being evaluated as an clinically to speed recovery from chemotherapy-induced
immunotherapeutic agent in patients with superficial neutropenia. Because GM-CSF also has immunomodu-
bladder cancer. Interim results from a national multicen- latory effects (Table 5-5),13,44 investigators have explored
ter phase II trial of combination Bacille Calmette-Guerin whether this agent has immunotherapeutic activity in
(BCG) plus IFN-α-2b suggest that this agent has bio- RCC patients. Unfortunately, to date no significant clin-
logic activity in this setting.41 A total of 337 patients who ical benefits have been attributable to this cytokine.45–48
could be evaluated with moderate to high-risk superficial
tumors (BCG naïve = 206 patients, BCG failures = 131
BACILLE CALMETTE-GUERIN
patients) received induction, followed by maintenance
courses of BCG + IFN-α. At a median follow-up of 24 Intravesical BCG for bladder cancer represents one of
months, the simple tumor recurrence rates were 35% for the most clinically successful applications of nonspecific
Chapter 5 Immunotherapy: Basic Guidelines 93

Table 5-5 Effects of GM-CSF retreatment in patients who had previously failed IL-2,
these data provided indirect evidence suggesting in vivo
Activates macrophages, monocytes, and dendritic cells
activity of TIL in humans. However, in a follow-up mul-
Enhances tumoricidal activity of macrophages and ticenter, randomized phase III trial of TILs with IL-2 in
monocytes against tumor the setting of metastatic RCC, patients who received
TILs + IL-2 failed to have a survival advantage or
Upregulates macrophage antigen presentation improved response rate compared to those receiving
Increases macrophage production of angiogenesis-inhibitor IL-2 alone (see Figure 5-3B).55
Although unselected TILs appear to be of limited
Enhances antibody-dependent cellular toxicity benefit in patients with metastatic RCC, a recent study
demonstrated significant clinical benefits when TILs,
Chemotactic for monocytes and polymorphonuclear cells prescreened in vitro for cytotoxicity against melanoma
GM-CSF, granulocyte-macrophage colony-stimulating factor. cells, were expanded and then given back to melanoma
patients following nonmyeloablative immunosuppression
(see Figure 5-3C).56 It has been hypothesized that
immunotherapy. The immunologic events associated immunodepleting chemotherapy given with this regimen
with the BCG-induced antitumor responses are not well may have created “immunologic space” or perhaps oblit-
understood, however, it is clear that stimulation of T erated T-suppressor cell populations, allowing for an
lymphocytes with subsequent T-cell cytokine secretion increased in vivo expansion of melanoma-toxic TIL.
are downstream effects of BCG that are critical to its
antitumor activity.49–51 The clinical results of intravesical
HEMATOPOIETIC CELL TRANSPLANTATION
BCG as treatment for patients with superficial bladder
cancer are discussed in detail in Chapter 18. Approximately 35 years ago, allogeneic hematopoietic
stem cell transplantation (HCT) was devised as a method
to maximize the dose of chemotherapy that could be
ADOPTIVE CELLULAR THERAPY
given to patients with advanced malignancies. It was
Lymphokine-Activated Killer Cells
hypothesized that patients with chemotherapy-resistant
Lymphokine-activated killer (LAK) cells are generated ex tumors might benefit from “dose-intensification,” largely
vivo by collecting circulating lymphocytes by large vol- based on evidence showing a dose–response relationship
ume of lymphocytapheresis and incubating them in IL-2- of some neoplasms to chemotherapy. The transplanta-
rich media for several days (Figure 5-3A). Although LAK tion of HLA-matched hematopoietic stem cells (the allo-
cells have shown dramatic MHC class I nonrestricted graft) is used as a means of regenerating hematopoiesis
cytotoxicity against RCC and other tumor cells in vitro, rendered defunct as a consequence of intensive
their clinical value in humans is questionable, as two chemotherapy (the conditioning regimen). The tradi-
phase III randomized trials failed to demonstrate a sur- tional method of harvesting hematopoietic stem cells is
vival advantage in metastatic RCC patients treated with via multiple bone marrow aspirations from the posterior
LAK cells + high-dose IL-2 compared to those who iliac crest under general anesthetic. Alternatively, G-CSF,
received high-dose IL-2 alone.52,53 GM-CSF, or even low-dose chemotherapy, can be used
to mobilize bone marrow stem cells, which are then col-
lected from the peripheral circulation by leukapheresis.
Tumor-Infiltrating Lymphocytes
The actual bone marrow transplant involves simply
Tumor-infiltrating lymphocytes (TILs) are lymphocytes infusing the allograft into one of the patient’s peripheral
(T cells + NK cells) that are extracted from fresh tumor or central veins. The infusion of the donor allograft
specimens (e.g., nephrectomies) and expanded in vitro in (referred to as “day 0”) is usually performed 1–2 days fol-
IL-2-rich media (see Figure 5-3B). By virtue of their hav- lowing the completion of the conditioning regimen.57
ing infiltrated into tumor tissue and their previously The allograft consists of not only donor hematopoietic
demonstrated antitumor activity in vitro (class I MHC stem cells but also immune cells (including NK, B, and
restricted), TILs are thought to contain a population of T-cells) that were eradicated in the patient by the condi-
T cells that recognize antigens specific to the tumor. tioning regimen. Patients typically receive immuno-
Following in vitro expansion, TILs are reinfused into the suppressive drugs, such as cyclosporine (CSA), or
patient along with other immunomodulators, such as tacrolimus, for the first 3–6 months following the proce-
IL-2. A trial evaluating TIL + IL-2 in patients who failed dure to prevent the donor immune cells from attacking
in previous IL-2-based therapy for metastatic melanoma normal host tissues, such as the liver, GI tract, or skin, a
or RCC demonstrated a 14% response rate.54 Given the complication known as graft-versus-host disease
historical data showing the ineffectiveness of IL-2 (GVHD).
94

Collection of lymphocytes Preparation of lymphocytes Single treatment cycle (days) Results

Lymphokine
A activated
killer cells Lymphocytes + IL-2
Lymphocytes isolated from circulating ⫻ 5 days
(LAK cells) 1 4 7 10 13 16 19 22 25 28 31
blood on days 8–10 ( ) (following IL-2) No improvement in objective
response rate over IL-2-alone
Part I Principles of Urologic Oncology

regimen in metastatic RCC


Tumor
patients52,53,55
infiltrating
B
lymphocytes Unselected TILs + IL-2
(TILs) Tumor infiltrating lymphocytes (TILs)
⫻ 5 days
extracted from renal cell carcinoma 1 4 7 10 13 16 19 22 25 28 31

Immunodepleting
C chemotherapy Partial response in 6/13
+ melanoma patients;
Tumoricidal TILs + IL-2
Tumoricidal Tumor infiltrating lymphocytes (TILs) no complete responses56
In vitro screening 14 days
extracted from melanoma tumor 1 4 7 10 13 16 19 22 25 28 31
TILs of TILs against
melanoma cells

Intravenous IL-2.
Leukapheresis (collection of lymphocytes from peripheral blood).
Lymphocyte infusion.
Tumor with infiltrating lymphocytes (TILs).
Immunodepleting chemotherapy.
RCC, Renal cell carcinoma.

Figure 5-3 Newer methods of adoptive immunotherapy. A, LAK cells. (Based on Law TM,
Motzer RJ, Mazumdar M, et al: Cancer 1995; 76(5):824–832, with permission.) B, TIL cell.
(Based on Figlin RA, Thompson JA, Bukowski RM, et al: J Clin Oncol 1999;
17(8):2521–2529, with permission.) C, Tumoricidal TILs. (Based on Dudley ME, Wunderlich
JR, Robbins PF, et al: Science 2002; 298(5594):850–854, with permission.)
Chapter 5 Immunotherapy: Basic Guidelines 95

In conventional myeloablative HCT, the primary anti- cells typically contain cells from both the patient and the
tumor effect comes from the high-dose chemotherapy donor, a condition referred to as mixed chimerism (in
and/or total body irradiation (conditioning regimen) Greek mythology, a Chimaira was a fire-spouting mon-
(Table 5-6). The term “myeloablative” indicates that the ster with a lion’s head, goat’s body, and serpent’s tail).59
patient’s bone marrow is totally obliterated as a result of Mixed chimerism results in a “tolerogenic state,” mean-
these highly cytotoxic interventions. It is now known that ing that the immune cells that develop from the donor’s
powerful antitumor effects against those malignant cells engrafted cells are unable to kill the patient’s tumor. In
surviving high-dose conditioning are generated by donor order for these cells to acquire antitumor activity, the
lymphocytes transplanted with the allograft. This allo- hematopoietic environment needs to transition to “full
geneic immune-mediated antineoplastic effect, called donor chimerism,” in which patient’s immune system is
graft-versus-leukemia (GVL) or graft-versus-tumor completely replaced by donor cells. This transformation
(GVT), is capable of curing patients with a variety of is facilitated by the withdrawal of posttransplant
treatment-refractory hematologic malignancies. immunosuppression (e.g., tacrolimus or cyclosporine)
Knowledge of RCCs susceptibility to immune effec- and the administration of donor lymphocyte infusions.60
tors has recently led investigators to test allogeneic trans- Nonmyeloablative HCT is capable of inducing curative
plantation as a form of immunotherapy in patients with GVT effects against a number of hematologic malignan-
metastatic RCC. Pilot trials were based on the hypothe- cies. Equally important, preliminary data show the
sis that GVT effects, analogous to the GVL effect seen in approach is associated with a lower risk of regimen-related
leukemias and lymphomas, might be generated following mortality (7.5% to 18%) compared to conventional mye-
the transplantation of allogeneic donor T cells.58 loablative procedures (25% to 35%). The improved toxic-
ity profile observed with nonmyeloablative HCT has
provided the basis for exploring allogeneic immunother-
Nonmyeloablative Hematopoietic Cell
apy’s potential to induce GVT effects in patients with
Transplantation
treatment-refractory solid tumors. Recently, a few groups
“Mega-dose” conditioning is largely responsible for the have published the results of pilot trials investigating
25% to 35% regimen-related mortality associated with nonmyeloablative HCT for the treatment of cytokine-
traditional myeloablative HCT. The powerful and cura- refractory metastatic RCC. The strategy used at the
tive capacity of the GVT effect has recently brought into National Institutes of Health involves nonmyeloablative
question the need for toxic myeloablative conditioning conditioning with a cyclophosphamide and fludarabine-
regimens. Reduced intensity conditioning regimens or based regimen, intended to induce host immunosuppres-
nonmyeloablative transplants were proposed as a poten- sion, followed by infusion of a G-CSF mobilized
tially less toxic alternative to conventional HCT. In con- peripheral blood stem cell graft from an HLA-identical
trast to a myeloablative HCT, the primary antitumor sibling donor (Figure 5-4). Ten of the first nineteen61 and
effect in a nonmyeloablative HCT results from the immune subsequently 22 of the first 55 patients undergoing this
cells that are transferred to the patient from the immuno- approach had evidence for a GVT effect, including 6 patients
competent HLA-matched donor. In this setting, the con- who had a complete response and 15 with a partial response.
ditioning regimen serves primarily to suppress the Many of the responses were durable, including the first
patient’s immune system just enough to prevent host patient treated who remains in complete remission 512 years
rejection of the donor allograft (see Table 5-6). after transplantation. GVHD was the most common com-
Accordingly, the dose of chemotherapy and toxicities plication, with approximately two-thirds of the patients
related to the conditioning regimen are less following experiencing acute grade II–IV GVHD. However,
nonmyeloablative compared to myeloablative HCT. patients developing this complication were more likely to
After donor cell engraftment, the patient’s hematopoietic have a disease response than those who never developed

Table 5-6 Allogeneic Hematopoietic Cell Transplantation (HCT)


Conditioning Source of Mechanism of Tumor
Regimen Transplanted Cells Regression

Myeloablative allogeneic HCT High-dose chemotherapy +/− HLA-compatible donor Conditioning regimen and
(Conventional HCT) Total Body XRT graft-versus-tumor effects

Non-myeloablative HCT Low-dose chemotherapy +/− HLA-compatible donor Immune mediated via
(“Mini-transplant”) Low-dose total body XRT graft-versus-tumor effects

HCT, Hematopoietic cell transplantation; HLA, Human leukocyte antigen; XRT, External beam radiation therapy.
96 Part I Principles of Urologic Oncology

Transfuse donor
Nonmyeloablative T-cells (DLI)
chemotherapy
Transplant day

–7 –6 –5 –4 –3 –2 –1 30 45 60 100

GVT effect
Transfuse the
allograft
Taper dose of cyclosporine
Cyclosporine

DLI, Donor lymphocyte infusion; GVT, Graft-versus-tumor.

Figure 5-4 Nonmyeloablative hematopoietic cell transplantation.

GVHD.61 Disease regression was typically delayed by 4–6 was refractory to high-dose IL-2. Preliminary experience
months following the procedure, and occurred after would suggest that certain patient and tumor characteris-
cyclosporine had been tapered and after T-cell chimerism tics herald a better outcome, such as small tumor burden,
had converted to predominantly donor in origin. Despite lung-only disease, slow rate of tumor growth, and good
the advanced disease status of patients enrolled on this patient performance status. Several other groups have
pilot trial, transplant-related mortality was relatively low, recently reported graft-versus-RCC effects following non-
with 6 patients succumbing to nonrelapse-related mortal- myeloablative HCT. Using cyclophosphamide and flu-
ity. Figure 5-5 shows a clinical response following non- darabine for pretransplant conditioning, investigators at
myeloablative HCT in a patient with metastatic RCC that the University of Chicago achieved complete donor

Prior to graft-versus-tumor effect


Day 100 Posttransplant Day 100 Posttransplant

A-1 B-1
Following graft-versus-tumor effect
Day 189 Posttransplant Day 189 Posttransplant

A-2 B-2
Figure 5-5 Clinical response following nonmyeloablative hematopoietic cell transplantation
in a patient with metastatic refractory RCC.
Chapter 5 Immunotherapy: Basic Guidelines 97

engraftment in 12 of 15 metastatic RCC patients undergo- success of antiCD20 (Rituximab) for low-grade lym-
ing an HLA-matched sibling transplant, four of whom had phomas and antiHER-2 (Herceptin) for breast carci-
a partial response. None of the patients who rejected the noma, monoclonal antibodies (mAbs) for treating cancer
allograft demonstrated a disease response.62 Investigators remain a very active area of research.
from Milan, Italy, reported 4 of 7 patients having a disease Therapeutic monoclonal antibodies were originally
response following a nonmyeloablative transplant that used derived from rodents. One of the major problems with
Thiotepa and fludarabine-based conditioning regimen.63 murine antibodies is their immunogenicity; the patient’s
Although these pilot results are encouraging, this immune system recognizes them as being foreign (Figure
approach remains experimental and should be reserved 5-6A), leading to an antibody attack against the mouse
for patients with metastatic RCC who have previously antibody (human antimouse antibody, HAMA). This
failed cytokine therapy, especially considering the risk of host immune response not only neutralized the thera-
fatal complications (mostly severe GVHD) associated peutic potential of foreign antibody but also occasionally
with the procedure. The primary outcome of these trials caused life-threatening anaphylactoid reactions.64 Two
has been to establish evidence of GVT effects against this antibody design strategies were subsequently developed
malignancy. Larger trials and refinements in the trans- to overcome this problem.64,65 Chimeric antibodies are
plant technique to decrease the risk of GVHD are rodent–human antibody constructs, wherein the variable
needed to define the role transplantation will ultimately (antigen binding) region is of rodent origin and the con-
play in their management of these patients. stant (effector) region is of human origin (Figure 5-6B).
Humanized antibodies are antibody constructs in which
rodent gene segments coding for the antigen binding
MONOCLONAL ANTIBODIES
loops are grafted onto human antibodies (Figure 5-6C).
The potential of antibodies to function as “magic bullets” More recently, phage display technology and transgenic
for the treatment of cancer has great appeal due to their approaches have enabled the production of entirely
tremendous target specificity. Based on the preliminary human recombinant antibodies.64,65

“Naked” antibody Chimeric antibody

Rodent constant regions (heavy, light chains) Human constant regions (heavy, light chains)
Rodent variable region (heavy, light-chain) Rodent variable region (heavy, light-chain)
Rodent CDR (complementarity-determining Rodent CDR (complementarity-determining
region) region)
A B
Humanized antibody

Human constant regions (heavy, light chains)


Rodent variable region (heavy, light-chain)
Rodent CDR (complementarity-determining
region)
C
Figure 5-6 Therapeutic rodent monoclonal antibodies (mAbs). A, Naked rodent mAb.
B, Chimeric rodent-human mAb. C, Humanized mAb.
98 Part I Principles of Urologic Oncology

The first two antibodies to be approved by the FDA involves the conjugation of effector moieties to the anti-
were Rituximab, a chimeric antiCD20 monoclonal body to improve tumor killing (Table 5-8).64,65
antibody for treatment of low-grade non-Hodgkin’s lym- Vascular endothelial growth factor (VEGF) promotes
phoma, as well as Herceptin, a humanized monoclonal endothelial cell proliferation and is often secreted by
antibody against HER-2 for the treatment of metastatic tumors in order to establish and/or increase their blood
breast cancer. Both drugs have shown individual activity in supply. Clear-cell RCCs often produce and secrete high
cancer patients with chemotherapy refractory tumors. levels of VEGF due to mutations in the von Hippel-
Research efforts are underway to develop effective anti- Lindau tumor suppressor gene.66–68 A recent randomized,
bodies against prostate-specific membrane antigen (PSMA) phase II trial was conducted to assess the efficacy of
in prostate cancer and G250, a cell surface antigen in RCC Bevacizumab, an anti-VEGF humanized monoclonal
(clear cell type). However, besides HAMA, a number of antibody, in the setting of advanced clear-cell RCC.69
other factors limit the efficacy of antibody therapy includ- One-hundred and sixteen patients were randomized to
ing their ability to mediate cell death once they have bound receive either placebo, low-dose Bevacizumab (3 mg/kg
to their tumor target (Table 5-7).64,65 Strategies are cur- every 2 weeks), or high-dose Bevacizumab (10 mg/kg every
rently being developed to overcome these barriers in hopes 2 weeks). The vast majority (>89%) had previously under-
of enhancing antibody efficacy. One such approach gone nephrectomy and received (and failed) IL-2 therapy.
At a second interim evaluation (at a median follow-up
time of 27 months from study entry), the National Cancer
Institute data safety and monitoring board recommended
Table 5-7 Barriers to Effective Antibody Cancer Therapy closure of accrual on the basis of the difference between
Mouse antibodies are immunogenic, provoking human the placebo and high-dose Bevacizumab in the time to
anti-mouse antibody (HAMA) response progression of disease (2.5 months and 4.8 months,
respectively, p < 0.001 by log-rank test). Only 4 of 116
Difficulty in identifying antigens that are expressed on patients had objective responses (all of which were partial
tumor cells and not on normal cells responses), and all of these had received high-dose
Bevacizumab. In the high-dose cohort, the response rate
Antigen-loss on tumor cell surface
was 10% (4 of 39 patients). At the last analysis, there were
Adequate delivery of antibody to tumor depends on good no significant differences in overall survival between
vascularity of tumor groups ( p > 0.20 for all comparisons).
Bevacizumab has further demonstrated antitumor
Short half-life of some antibodies requires repeated activity in preclinical studies against hormone-refractory
administrations
prostate cancer70 and in a phase II randomized trial
Antibodies are usually species-specific, limiting preclinical (Bevacizumab + Fluorouracil/Leucovorin versus
animal toxicology studies Fluorouracil/Leucovorin) in the setting of metastatic col-
orectal carcinoma.71

Table 5-8 Antibody Strategies in Cancer Therapy


Ab-Conjugate Mechanism of Tumor Cell Killing

“Naked” Ab (unconjugated) CDC, ADCC, direct antiproliferative effects, idiotype/anti-idiotype network

Ab-radioisotope Direct radiation-induced cytotoxicity

Ab-immunotoxin Once inside the cell, toxin irreversibly blocks an essential metabolic process

Ab-drug Tumoricidal drug internalized by tumor cell

Ab-photosensitizer Photosensitizer-moiety produces oxygen free radical when exposed to light

Ab-Ab (bispecific Ab’s) Unlike direct conjugations of effector agents to a single antibody, the effector and
antigen-binding domains are bound to separate, covalently-linked antibodies

Ab-enzyme, then prodrug (ADEPT) Enzyme-Ab conjugate administered first, followed by prodrug. Enzyme converts
prodrug into active cytotoxic agent at tumor site only

Ab, antibody; CDC, complement-dependent cytotoxicity; ADCC, antibody-dependent cell-mediated cytotoxicity; ADEPT, antibody-dependent
enzyme-mediated cytotoxicity
Chapter 5 Immunotherapy: Basic Guidelines 99

REFERENCES Working Group experience, 1989–1997. Cancer J Sci Am


1997; 3 (Suppl 1):S73–S78.
1. Coley W: Further observations on the treatment of 20. Dutcher J: Personal Communication, 2003.
malignant tumors with the toxins of erysipelas and bacillus 21. McDermott D: A randomized phase III trial of high-dose
prodigious with a report of 160 cases. Johns Hopkins interleukin-2 vs. subcutaneous IL2/interferon in patients
Hospital Bulletin 1896; 7:157–162. with metastatic renal cell carcinoma. Paper presented at
2. Schreiber H: Tumor immunology. In Paul WE (ed): the American Society of Clinical Oncology, May 12–15,
Fundamental Immunology, 4th edition, pp 1237–1270. 2001, San Francisco, CA.
Philadelphia, Lippencott-Raven Publishers, 1999. 22. Dutcher JP, Fisher RI, Weiss G, et al: Outpatient
3. Abbas AK, Lichtman AH, Pober JS: Immunity to tumors. subcutaneous interleukin-2 and interferon-alpha for
In Abbas AK (ed): Cellular and Molecular Immunology, metastatic renal cell cancer: five-year follow-up of the
4th edition, pp 384–403. Philadelphia, WB Saunders Co, Cytokine Working Group Study. Cancer J Sci Am 1997;
2000. 3(3):157–162.
4. Shu S, Plautz GE, Krauss JC, Chang AE: Tumor 23. Rogers E, Bredin H, Butler M, et al: Combined
immunology. JAMA 1997; 278(22):1972–1981. subcutaneous recombinant alpha-interferon and
5. Leonard WJ: Type I cytokines and interferons and their interleukin-2 in metastatic renal cell cancer: results of the
receptors. In Paul WE (ed): Fundamental Immunology, Multicentre All Ireland Immunotherapy Study Group.
4th edition, pp 741–774. Philadelphia, Lippencott-Raven Eur Urol 2000; 37(3):261–266.
Publishers, 1999. 24. Negrier S, Caty A, Lesimple T, et al: Treatment of
6. Lin WC, Yasumura S, Suminami Y, et al: Constitutive patients with metastatic renal carcinoma with a
production of IL-2 by human carcinoma cells, expression combination of subcutaneous interleukin-2 and interferon
of IL-2 receptor, and tumor cell growth. J Immunol 1995; alfa with or without fluorouracil. Groupe Francais
155(10):4805–4816. d’Immunotherapie, Federation Nationale des Centres de
7. Oppenheim J, Fujiwara H: The role of cytokines in Lutte Contre le Cancer. J Clin Oncol 2000;
cancer. Cytokine Growth Factor Rev 1996; 7(3): 18(24):4009–4015.
279–288. 25. Mickisch GH, Garin A, van Poppel H, de Prijck L,
8. Society for Leukocyte Biology— Sylvester R: Radical nephrectomy plus
www.leukocytebiology.org. interferon-alfa-based immunotherapy compared with
9. International Society for Interferon and Cytokine interferon alfa alone in metastatic renal-cell carcinoma: a
Research—http://bioinformatics.weizmann.ac.il/ISICR/. randomised trial. Lancet 2001; 358(9286):966–970.
10. International Cytokine Society— 26. Flanigan RC, Salmon SE, Blumenstein BA, et al:
http://bioinformatics.weizmann.ac.il/cytokine/. Nephrectomy followed by interferon alfa-2b compared
11. Proleukin (registered trademark), Aldesleukin for injection with interferon alfa-2b alone for metastatic renal-cell
[package insert]. cancer. N Engl J Med 2001; 345(23):1655–1659.
12. Whittington R, Faulds D: Interleukin-2. A review of its 27. Negrier S, Escudier B, Lasset C, et al: Recombinant
pharmacological properties and therapeutic use in patients human interleukin-2, recombinant human interferon alfa-
with cancer. Drugs 1993; 46(3):446–514. 2a, or both in metastatic renal-cell carcinoma. Groupe
13. Rosenberg SA: Principles of cancer management: biologic Francais d’Immunotherapie. N Engl J Med 1998;
therapy. In Vincent T, DeVita J, Hellman S, Rosenberg 338(18):1272–1278.
SA (eds): Cancer: Principles & Practice of Oncology, 6th 28. Negrier S: Personal communication, 2003.
edition, pp 307–333. Philadelphia, Lippencott Williams & 29. Motzer RJ, Murphy BA, Bacik J, et al: Phase III trial of
Wilkins, 2001. interferon alfa-2a with or without 13-cis-retinoic acid for
14. Abbas AK: Cellular and Molecular Immunology, 4th patients with advanced renal cell carcinoma. J Clin Oncol
edition. Philadelphia, WB Saunders Co, 2000. 2000; 18(16):2972–2980.
15. Kirkwood J: Cancer immunotherapy: the interferon-alpha 30. Gleave ME, Elhilali M, Fradet Y, et al: Interferon
experience. Semin Oncol 2002; 29(3 Suppl 7):18–26. gamma-1b compared with placebo in metastatic renal-cell
16. Bukowski RM, Tendler C, Cutler D, et al: Treating cancer carcinoma. Canadian Urologic Oncology Group. N Engl
with PEG intron: pharmacokinetic profile and dosing J Med 1998; 338(18):1265–1271.
guidelines for an improved interferon-alpha-2b 31. Kammula US, White DE, Rosenberg SA: Trends in the
formulation. Cancer 2002; 95(2):389–396. safety of high dose bolus interleukin-2 administration in
17. Fisher RI, Rosenberg SA, Fyfe G: Long-term survival patients with metastatic cancer. Cancer 1998;
update for high-dose recombinant interleukin-2 in 83(4):797–805.
patients with renal cell carcinoma. Cancer J Sci Am 2000; 32. Schwartz RN, Stover L, Dutcher J: Managing toxicities of
6(Suppl 1):S55–S57. high-dose interleukin-2. Oncology (Huntingt) 2002;
18. Yang JC, Sherry RM, Steinberg SM, et al: A 16(11 Suppl 13):11–20.
randomized study of high and low-dose interleukin-2 in 33. Dutcher J, Atkins MB, Margolin K, et al: Kidney
patients with metastatic renal cancer. J Clin Oncol 2003 cancer: the Cytokine Working Group experience
(in press). (1986–2001): part II. Management of IL-2 toxicity and
19. Dutcher JP, Atkins M, Fisher R, et al: Interleukin-2-based studies with other cytokines. Med Oncol 2001;
therapy for metastatic renal cell cancer: the Cytokine 18(3):209–219.
100 Part I Principles of Urologic Oncology

34. Dutcher J: Current status of interleukin-2 therapy for metastatic renal cell carcinoma: results of two phase II
metastatic renal cell carcinoma and metastatic melanoma. clinical trials. Cancer 2000; 88(6):1317–1324.
Oncology (Huntingt) 2002; 16(11 Suppl 13):4–10. 48. Wos E, Olencki T, Tuason L, et al: Phase II trial of
35. Jorkov AS, Donskov F, Steiniche T, et al: Immune subcutaneously administered granulocyte-macrophage
response in blood and tumour tissue in patients with colony-stimulating factor in patients with metastatic renal
metastatic malignant melanoma treated with IL-2, IFN cell carcinoma. Cancer 1996; 77(6):1149–1153.
alpha and histamine dihydrochloride. Anticancer Res 49. Magno C, Melloni D, Gali A, et al: The anti-tumor
2003; 23(1B):537–542. activity of bacillus Calmette-Guerin in bladder cancer is
36. Agarwala SS, Glaspy J, O’Day SJ, et al: Results from a associated with an increase in the circulating level of
randomized phase III study comparing combined interleukin-2. Immunol Lett 2002; 81(3):235–238.
treatment with histamine dihydrochloride plus 50. Luo Y, Chen X, O’Donnell MA: Role of Th1 and Th2
interleukin-2 versus interleukin-2 alone in patients with cytokines in BCG-induced IFN-gamma production:
metastatic melanoma. J Clin Oncol 2002; 20(1):125–133. cytokine promotion and simulation of BCG effect.
37. Donskov F, von der Maase H, Henriksson R, et al: Cytokine 2003; 21(1):17–26.
Outpatient treatment with subcutaneous histamine 51. Broghammer EL, Ratliff TL: Immunotherapy of urologic
dihydrochloride in combination with interleukin-2 and tumors: principles and progress. Urol Oncol 2002;
interferon-alpha in patients with metastatic renal cell 7(2):45–56.
carcinoma: results of an open single-armed multicenter 52. Law TM, Motzer RJ, Mazumdar M, et al: Phase III
phase II study. Ann Oncol 2002; 13(3):441–449. randomized trial of interleukin-2 with or without
38. Smyth MJ, Takeda K, Hayakawa Y, Nature’s TRAIL—on a lymphokine-activated killer cells in the treatment of
path to cancer immunotherapy. Immunity 2003; 18(1):1–6. patients with advanced renal cell carcinoma. Cancer 1995;
39. Schmaltz C, Alpdogan O, Kappel BJ, et al: T cells require 76(5):824–832.
TRAIL for optimal graft-versus-tumor activity. Nat Med 53. Rosenberg SA, Lotze MT, Yang JC, et al: Prospective
2002; 8(12):1433–1437. randomized trial of high-dose interleukin-2 alone or in
40. Smyth MJ, Cretney E, Takeda K, et al: Tumor necrosis conjunction with lymphokine-activated killer cells for the
factor-related apoptosis-inducing ligand (TRAIL) treatment of patients with advanced cancer. J Natl Cancer
contributes to interferon gamma-dependent natural killer Inst 1993; 85(8):622–632.
cell protection from tumor metastasis. J Exp Med 2001; 54. Lee DS, White DE, Hurst R, Rosenberg SA, Yang JC:
193(6):661–670. Patterns of relapse and response to retreatment in patients
41. O’Donnell MA, Lilli K, Leopold C: Interim results from with metastatic melanoma or renal cell carcinoma who
a national multicenter phase II trial of combination BCG responded to interleukin-2-based immunotherapy. Cancer
plus interferon-alfa-2B for superficial bladder cancer. J Sci Am 1998; 4(2):86–93.
Paper presented at the American Urological Association 55. Figlin RA, Thompson JA, Bukowski RM, et al:
Meeting, April 28, 2003, Chicago, IL. Multicenter, randomized, phase III trial of CD8(+) tumor-
42. Bazarbashi S, Raja MA, El Sayed A, et al: Prospective infiltrating lymphocytes in combination with recombinant
phase II trial of alternating intravesical Bacillus Calmette- interleukin-2 in metastatic renal cell carcinoma. J Clin
Guerin (BCG) and interferon alpha IIB in the treatment Oncol 1999; 17(8):2521–2529.
and prevention of superficial transitional cell carcinoma of 56. Dudley ME, Wunderlich JR, Robbins PF, et al: Cancer
the urinary bladder: preliminary results. J Surg Oncol regression and autoimmunity in patients after clonal
2000; 74(3):181–184. repopulation with antitumor lymphocytes. Science 2002;
43. Stricker P, Pryor K, Nicholson T, et al: Bacillus Calmette- 298(5594):850–854.
Guerin plus intravesical interferon alpha-2b in patients 57. Vose JM, et al: Bone marrow transplantation.
with superficial bladder cancer. Urology 1996; In Abeloff MD, Armitage JO, Lichter AS, Niederhuber
48(6):957–961. [Discussion 961-962.] JE (eds): Abeloff: Clinical Oncology, 2nd edition,
44. Spitler LE: Adjuvant therapy of melanoma. Oncology p 482. Philadelphia, Churchill Livingstone
(Huntingt) 2002; 16(1 Suppl 1):40–48. Inc, 2000.
45. Smith IJ, Kurt RA, Baher AG, et al: Immune effects of 58. Childs RW, Clave E, Tisdale J, et al: Successful treatment
escalating doses of granulocyte-macrophage colony- of metastatic renal cell carcinoma with a nonmyeloablative
stimulating factor added to a fixed, low-dose, inpatient allogeneic peripheral-blood progenitor-cell transplant:
interleukin-2 regimen: a randomized phase I trial in evidence for a graft-versus-tumor effect. J Clin Oncol
patients with metastatic melanoma and renal cell 1999; 17(7):2044–2049.
carcinoma. J Immunother 2003; 26(2):130–138. 59. Dorland’s On-Line Medical Dictionary.
46. Schmidinger M, Steger G, Wenzel C, et al: Sequential 60. Childs RW: Nonmyeloablative blood stem cell
administration of interferon-gamma, GM-CSF, and transplantation as adoptive allogeneic immunotherapy for
interleukin-2 in patients with metastatic renal cell metastatic renal cell carcinoma. Crit Rev Immunol 2001;
carcinoma: results of a phase II trial. J Immunother 2001; 21(1–3):191–203.
24(3):257–262. 61. Childs R, Chernoff A, Contentin N, et al: Regression of
47. Ryan CW, Vogelzang NJ, Dumas MC, Kuzel T, Stadler metastatic renal-cell carcinoma after nonmyeloablative
WM: Granulocyte-macrophage-colony stimulating factor allogeneic peripheral-blood stem-cell transplantation.
in combination immunotherapy for patients with N Engl J Med 2000; 343(11):750–758.
Chapter 5 Immunotherapy: Basic Guidelines 101

62. Rini BI, Zimmerman T, Stadler WM, Gajewski TF, 67. Mukhopadhyay D, Knebelmann B, Cohen HT, Ananth S,
Vogelzang NJ: Allogeneic stem-cell transplantation of Sukhatme VP: The von Hippel-Lindau tumor suppressor
renal cell cancer after nonmyeloablative chemotherapy: gene product interacts with Sp1 to repress vascular
feasibility, engraftment, and clinical results. J Clin Oncol endothelial growth factor promoter activity. Mol Cell Biol
2002; 20(8):2017–2024. 1997; 17(9):5629–5639.
63. Bregni M, Dodero A, Peccatori J, et al: Nonmyeloablative 68. Folkman J, Shing Y. Angiogenesis. J Biol Chem 1992;
conditioning followed by hematopoietic cell allografting 267(16):10931–10934.
and donor lymphocyte infusions for patients with 69. Yang JC, Haworth L, Sherry RM, et al: A randomized
metastatic renal and breast cancer. Blood 2002; trial of bevacizumab, an anti-vascular endothelial growth
99(11):4234–4236. factor antibody, for metastatic renal cancer. N Engl J Med
64. Waldmann H, Gilliland LK, Cobbold SP, Hale G: 2003; 349(5):427–434.
Immunotherapy. In Paul WE (ed): Fundamental 70. Fox WD, Higgins B, Maiese KM, et al: Antibody to
Immunology, 4th edition, pp 1511–1533. Philadelphia, vascular endothelial growth factor slows growth of an
Lippincott-Raven Publishers, 1999. androgen-independent xenograft model of prostate
65. Welschof M, Krauss J (eds): Recombinant Antibodies for cancer. Clin Cancer Res 2002; 8(10):3226–3231.
Cancer Therapy: Methods and Protocols, vol 207. 71. Kabbinavar F, Hurwitz HI, Fehrenbacher L, et al: Phase
Totowa, NJ, Humana Press, 2002. II, randomized trial comparing bevacizumab plus
66. Maxwell PH, Wiesener MS, Chang GW, et al: The fluorouracil (FU)/leucovorin (LV) with FU/LV alone in
tumour suppressor protein VHL targets hypoxia-inducible patients with metastatic colorectal cancer. J Clin Oncol
factors for oxygen-dependent proteolysis. Nature 1999; 2003; 21(1):60–65.
399(6733):271–275.
C H A P T E R

6 Health-Related Quality of Life Issues


in Urologic Oncology
David F. Penson, M.D., M.P.H., and Mark S. Litwin, M.D., M.P.H.

The primary goal of health care providers when treating HRQOL research and discuss various instruments avail-
patients with urologic malignancies has traditionally been able to assess HRQOL in genitourinary cancers. We
to prolong patient survival. However, as the past decade then briefly examine the current literature regarding the
has brought new screening modalities and improvements effect of urologic malignancies on quality of life.
in treatment, clinicians and researchers have begun to Necessarily, we focus primarily on prostate cancer, as this
focus on other outcomes as well. In particular, as we have has the largest body of literature. However, we touch on
recognized that cancer and its treatment affects both quality of life issues in bladder, kidney, and testicular can-
quantity and quality of life in our patients, it has become cers as well.
clear that various components of well-being must also be
addressed when treating individual patients with cancer or
THE METHODOLOGY OF HEALTH-RELATED
when conducting cancer clinical trials.1 While clinicians
QUALITY OF LIFE RESEARCH
must still focus on “traditional” outcomes, such as 5- or
HRQOL Instruments
10-year survival rates, complete and partial responses, or
serum tumor marker levels, the increased attention to It is difficult for the novice to understand the task of
patients’ overall well-being has generated interest in more objectively quantifying quality of life, which is an inher-
“refined” endpoints in urologic oncology. One such end- ently qualitative phenomenon.5 However, the principles
point is health-related quality of life (HRQOL). of psychometric test theory 6 may be applied to produce
HRQOL encompasses a wide range of human experi- accurate assessments of HRQOL. HRQOL data are col-
ence, including the daily necessities of life, intrapersonal lected using patient-centered surveys, called instruments.
and interpersonal responses to illness, and activities Instruments can be self-administered by the patient or
associated with professional fulfillment and personal hap- can be administered with the assistance of a neutral third-
piness.2 Importantly, HRQOL involves patients’ percep- party interviewer in a standardized fashion. Instruments
tions of their own health and ability to function in life. typically contain multiple questions, or items, that are
HRQOL is often confused with functional status. While organized into scales. Each scale measures a different
functional status is an important dimension of HRQOL, aspect, or domain, of HRQOL. Domains can be general
there are numerous other aspects of HRQOL, including or disease-specific. General HRQOL domains address
role function, vitality, mental health, pain, and psychoso- the components of overall well-being, while disease-
cial interactions, which are equally important. Despite specific domains focus on the impact of particular
the commonly held belief that this type of data cannot be organic dysfunctions that affect HRQOL.7 General
easily collected, patients’ compliance with HRQOL HRQOL domains typically address general health per-
questionnaires is usually high.3 The impact of HRQOL ceptions, sense of overall well-being, and function in the
in clinical oncology is now considered so important that physical, emotional, and social domains. Cancer-specific
a cancer trial is considered incomplete without the inclu- HRQOL domains focus on more directly relevant
sion of HRQOL outcomes.1,4 domains, such as anxiety about cancer recurrence, nausea
In this chapter, we review quality of life issues in uro- from chemotherapy, or urinary incontinence from
logic oncology. We initially review the methodology of sphincter damage.

102
Chapter 6 Health-Related Quality of Life Issues in Urologic Oncology 103

Creation and Testing of New Instruments Reliability


It is ill-advised to use casually constructed instruments Reliability refers to how reproducible the scale is. In other
in HRQOL research, as this can result in inaccurate words, what proportion of a patient’s test score is true and
data and flawed conclusions. Therefore, before an what proportion is due to random variation. Several types
instrument is used in a clinical setting, statistical and of reliability are typically assessed. Test–retest reliability is a
psychometric testing must be performed to measure the measure of response stability over time. It is assessed by
survey’s reliability and validity. Because psychometric administering scales to patients at two distinct time
testing is so rigorous and time-intensive, it is always points, typically 1 month apart. The correlation coeffi-
preferable to use an established, validated instrument cients between the two scores reflect the stability of
when available. Another advantage of using published responses. Test–retest reliability is most easily assessed
instruments in the collection of HRQOL data is that when the domain of interest is unlikely to change over
they allow researchers to compare new results to previ- short periods of time. When assessing test–retest reliabil-
ously studied populations. ity, one needs to ensure that the interval between admin-
If an appropriate, established HRQOL instrument is istrations is not too long, as real change can occur in the
not available for the disease process one is interested in variable, artificially deflating test–retest reliability coeffi-
studying, it may be reasonable to design a new instru- cients. Internal consistency reliability is a measure of the
ment. The first step in this difficult process is to pilot test similarity of an individual’s responses across several items,
the questionnaire to ensure that the patient population indicating the homogeneity of a scale.6 The statistic used
can easily understand and complete the survey. Pilot test- to quantify the internal consistency of a scale is called
ing often reveals problems that the researchers may not Cronbach’s coefficient alpha.8 Generally accepted stan-
notice, such as the inadvertent use of medical jargon that dards dictate that reliability statistics measured by these
patients do not understand (leading to unanswered ques- two methods should exceed 0.70.9 These and various
tions or inaccurate responses), use of diminutive print other forms of reliability are reviewed in Table 6-1.
size (causing difficulty reading the question), or unclear
wording (leading patients to misunderstand the ques-
Validity
tion). After pilot testing, new instruments are evaluated
for the two fundamental psychometric statistical proper- Validity refers to how well the scale or instrument measures
ties of reliability and validity. the attribute it is intended to measure. Content validity,

Table 6-1 Types of Reliability


Type of Reliability Characteristics Comments

Test–Retest Measures the stability of Requires the administration of survey to a sample


responses over time, typically in at two different and appropriate points in time.
the same group of respondents Time points that are too far apart may produce
diminished reliability estimates that reflect actual
change over time in the variable of interest

Intraobserver Measures the stability of responses Requires completion of a survey by an individual


over time in the same individual at two different and appropriate points in time.
respondent Time points that are too far apart may produce
diminished reliability estimates that reflect
actual change over time in the variable of interest

Alternate-form Uses differently worded stems or Requires two items in which the wording is different
response sets to obtain the same but aimed at the same specific variable and at
information about a specific topic the same vocabulary level

Internal consistency Measures how well several items Usually requires a computer to carry out calculations
in a scale vary together in a sample

Interobserver Measures how well two or more May be used to demonstrate reliability of a survey
respondents rate the same or may itself be the variable of interest in a study
phenomenon
104 Part I Principles of Urologic Oncology

sometimes incorrectly referred to as face validity, is the non- Many clinicians and researchers find the process and
quantitative assessment by experts of the scope and com- patient-centered data collection daunting and mistakenly
pleteness of a proposed items and scales. It is more believe that they can accurately estimate a patient’s qual-
superficial than other types of validity, and considered by ity of life. Numerous studies have documented that
some not to be a true measure of validity at all.10 physicians tend to underestimate both the degree of
Nevertheless, it is almost always included in the early stages symptoms and their negative effect on HRQOL.12–14
of instrument development, even if only as a general review Therefore, it is usually preferable to obtain HRQOL
of items by physicians or patients. Criterion validity requires data directly from patients; the treating physician should
the correlation of scale scores with other measurable health not attempt to estimate the patient’s HRQOL.
outcomes (predictive validity) and with results from other
established tests (concurrent validity). For example, the pre-
ESTABLISHED HRQOL INSTRUMENTS FOR USE
dictive validity of a new HRQOL scale for bony pain in
WITH UROLOGIC MALIGNANCIES
prostate cancer might be correlated with narcotic usage.
Likewise, the concurrent validity of a new urinary inconti- HRQOL instruments tend to be general or disease-
nence scale in prostate or bladder cancer could be correlated specific, depending on the domains addressed with the
with objective results on urodynamic testing. Construct survey. When studying urologic cancers, it is preferable
validity is a measure of how meaningful the scale or survey to use both general and disease-specific instruments, as
instrument is when in practical use. It is often requires years disease-specific symptoms can have profound effects on
of experience with a survey instrument to assess correctly. both disease-specific HRQOL and patients’ general well-
Often it is not calculated as a quantifiable statistic but as a being and overall functional status. The broad effect of
gestalt of how well a survey instrument performs in a mul- certain symptoms associated with urologic cancer may be
titude of settings and populations over time. An overview of overlooked if researchers do not use both general and
the various types of validity is presented in Table 6-2. disease-specific measures.15

Collection of HRQOL Data Established General HRQOL Instruments


In addition to using validated and reliable HRQOL instru- General HRQOL instruments focus on general domains
ments, clinicians and researchers must collect data in a of HRQOL and have been extensively researched and
manner that minimizes bias. For example, data regarding validated in many types of patients, including sick and
sexual dysfunction following radical prostatectomy should well. Examples include the RAND Medical Outcomes
not be collected directly by the operating surgeon, as Study 36-Item Health Survey (also known as the
patients have an unconscious desire to produce responses SF-36),16–20 the quality of well-being scale (QWB),21–25
that they think their physicians want to hear.11 This intro- the sickness impact profile (SIP),21,26,27 and the
duces measurement error. Therefore, it is always preferable Nottingham health profile (NHP).21,28–31 All of these
that data be gathered by disinterested third parties or that instruments assess the various general components of
instruments be self-administered by patients to avoid bias. HRQOL, including physical and emotional functioning,

Table 6-2 Types of Validity


Type of Validity Characteristics Comments

Face Casual review of how good an item Assessed by individuals with no formal training in the
or group of items appear subject under study

Content Formal expert review of how good Usually assessed by individuals with expertise in some
an item or series of items appear aspect of the subject under study

Criteron: Concurrent Measures how well the item or scale Requires the identification of an established, generally
correlates with “gold standard” accepted gold standard
measures of the same variable

Criterion: Predictive Measures how well the item or scale Used to predict outcomes or events of significance
predicts expected future that the item or scale might subsequently be used to
observations predict

Construct Theoretical measure or how Determined usually after years of experience by numerous
meaningful a survey instrument is investigators
Chapter 6 Health-Related Quality of Life Issues in Urologic Oncology 105

social functioning, and symptoms and have been thor- prostate cancer-specific HRQOL.41,42 It is a 20-item,
oughly validated and tested. self-administered tool that takes about 10 minutes to
complete. It is typically administered alongside the
SF-36, a general HRQOL instrument.19 The 20 items of
Cancer-Specific HRQOL Instruments for Use
the UCLA PCI are specific for prostate cancer and com-
in Urology
prise six scales (urinary function and bother, sexual func-
A number of cancer-specific instruments have long been tion and bother, and bowel function and bother) from 0
available. However, recently, many have had modules to 100, with higher scores representing better outcomes.
developed that are specific for urologic disease. For The UCLA PCI has now been used in several national
example, the European Organization for Research and and international studies and has recently been validated
Treatment of Cancer (EORTC) Core Quality of Life in Spanish.43 The UCLA PCI makes the important dis-
Questionnaire (QLQ-C30)32 is a 30-item questionnaire tinction between function and bother in the prostate-
that includes five scales (physical, role, emotional, cogni- specific domains. This feature is significant because the
tive, and social functioning), a global health scale, three bother experienced by patients does not necessarily cor-
symptom scales (fatigue, nausea/vomiting, and pain), and relate with the level of dysfunction.44
six single items concerning dyspnea, insomnia, appetite Although the UCLA PCI has been validated and is
loss, constipation, diarrhea, and financial difficulties due widely used, its focus on urinary incontinence does not
to disease. The EORTC QLQ-C30 can be used in attend to irritative voiding complaints. Hence,
patients with any type of cancer. However, a 20-item researchers at the University of Michigan developed the
prostate cancer module has recently been developed that Expanded Prostate Cancer Index-Composite (EPIC).
specifically includes a bowel symptom scale, urinary Building on the UCLA PCI, Wei et al.45 added 30 items
symptom scale and sexuality scale. This module has to the existing disease-specific domains, for a total of 50
been validated and shown to be reliable in men with items. The EPIC added additional items to the three
both localized33,34 and metastatic35 prostate cancer. existing bother domains (urinary, sexual, and bowel),
Unfortunately, this instrument does not distinguish developed hormonal function and bother domains, and
between function and bother in these domains. most importantly, expanded the urinary domain by
Another example of a cancer-specific HRQOL instru- adding items that capture irritative voiding symptoms.
ment that now has modules for various urologic malig- Therefore, the EPIC contains eight disease-specific
nancies is the Functional Assessment of Cancer Therapy domains: sexual function and bother, urinary function
(FACT).36–39 The main FACT instrument includes a set and bother, bowel function and bother, and hormonal
of 28 general items that pertain to all cancer patients function and bother. The urinary function domain con-
(FACT-G). Each item contains a statement that a patient tains two distinct subscales, urinary incontinence and uri-
may agree or disagree with across a five-point Likert nary irritation/obstruction, each with a separate
range. The FACT-G domains include well-being in four summary score. Because the disease-specific domains of
areas: physical, social-family, emotional, and functional. the EPIC instrument include significantly more items
The FACT-P, a new module to measure HRQOL in men than the original UCLA PCI, general HRQOL is typi-
with prostate cancer, was recently validated and may cally measured using the 12-item RAND SF-1246 rather
prove useful in the future.40 There is also a FACT mod- than the SF-36.
ule for bladder cancer, although this has not been widely
used or validated. The FACT may be accessed at
HEALTH-RELATED QUALITY OF LIFE IN SPECIFIC
http://www.facit.org/facit_questionnair.htm.
UROLOGIC MALIGNANCIES
Prostate Cancer
HRQOL Instruments Designed Specifically for Use
Of all genitourinary tumors, prostate cancer is the malig-
in Urologic Malignancies
nancy with the largest body of HRQOL research. There
A number of instruments developed for use specifically in are considerable data in both metastatic and localized dis-
urologic cancers. In the past, most of the research focus ease because both prostate cancer and its treatment affect
has been in prostate cancer, and, therefore, there are more quality of life.
established instruments available for use in this malig- In the case of metastatic prostate cancer, patients
nancy. Recently, new HRQOL instruments have been experience decrements in quality of life due to both
developed for use in bladder and kidney cancer as well. painful bony lesions and hormone ablation therapy.
In prostate cancer, the University of California, Los Investigators have shown that as metastatic prostate can-
Angeles Prostate Cancer Index (UCLA PCI) was the first cer progresses from hormone-sensitive to hormone
validated instrument specifically designed for use in this resistant disease, general HRQOL worsens accord-
condition and has been the gold standard for measuring ingly.47 Kim et al.48 also noted that among patients
106 Part I Principles of Urologic Oncology

treated for metastases, those with progressive disease randomized to orchiectomy plus placebo.53 However, the
appeared to have worse quality of life than those with sta- negative impact of androgen receptor blockade on
ble disease, particularly for pain, fatigue, sleep, and phys- HRQOL due to gastrointestinal side effects may be less
ical and role functioning. Studies have shown that in patients receiving bicalutamide.54
treatment of advanced prostate cancer improves Quality of life is of particular importance to men with
HRQOL. Albertsen et al.35 studied metastatic prostate hormone resistant disease where the goal of therapy is
cancer patients in remission on LHRH agonists and flu- often palliative in nature. A number of studies have doc-
tamide and found that their general HRQOL was indis- umented that various chemotherapeutic agents improve
tinguishable from an age-matched control population of HRQOL in men with hormone-resistant disease. For
men without prostate cancer. While treatment of example, Osoba et al.55 assessed HRQOL in 161 men
advanced prostate cancer appears to improve HRQOL, randomized to receive prednisone alone versus pred-
at least initially, the therapy itself can negatively affect nisone and mitoxantrone over a 26-week follow-up
HRQOL. This is of particular concern in men who pres- period. At 6 weeks, patients taking prednisone alone
ent with advanced but asymptomatic prostate cancer, as, showed no improvement in HRQOL scores, whereas
in these patients, the tumor itself probably has little those taking mitoxantrone plus prednisone showed sig-
impact on HRQOL. For example, Herr and O’Sullivan49 nificant improvements in global quality of life ( p = 0.009)
compared patients receiving hormonal therapy to those and four functional domains ( p < 0.01). In the cross-over
being observed for asymptomatic advanced disease. arm of the study, the addition of mitoxantrone to pred-
Patients who elected to observation until the develop- nisone after failure of prednisone alone was associated
ment of symptoms had better HRQOL than those who with improvements in pain, pain impact, pain relief,
opted for early intervention. In particular, men who insomnia, and global quality of life ( p < 0.003). After 18
received early therapy experienced significantly more weeks of therapy, those receiving mitoxantrone plus
fatigue and hot flashes that impacted quality of life than prednisone continued to improve in 11 of the 14 function
those who delayed treatment. Other studies note similar and symptom scales of the HRQOL measures used. This
findings. For example, Green et al.50 demonstrated that study and others56,57 demonstrate that palliative
early hormone ablation therapy was associated with chemotherapy can lessen the physical burden of prostate
worse sexual function and decreased role and social func- cancer in men with advanced hormone-resistant disease.
tioning. However, the patients who received early hor- Although these treatments may not prolong life
monal therapy did report better physical function than expectancy, a documented quality of life advantage for a
those who received deferred management. Although not given treatment will result in benefit to the patient and
a study of metastatic prostate cancer, researchers from should be strongly considered when choosing therapies.
the Prostate Cancer Outcomes Study (PCOS)51 com- In the case of localized prostate cancer, most of the
pared men with localized disease who received hormone research examining HRQOL has focused on the effect of
ablation therapy to men with localized disease who were treatment on quality of life. However, even in the case of
observed. Those who elected hormone ablation therapy localized disease, prostate cancer itself appears to have an
reported worse sexual function and more physical dis- effect on HRQOL. Bacon et al.58 compared general
comfort. Although all of these studies were observational HRQOL in 783 men from the Health Professionals
in nature and therefore may have been influenced by Follow-up Study cohort with incidental prostate cancer
selection bias, they all underscore the potential deleteri- to 1928 age-matched controls. They found that the men
ous effects of hormone therapy on HRQOL. with localized prostate cancer had worse general health,
Once patients elect to receive treatment for metastatic vitality, social function, and role limitations due to phys-
prostate cancer, studies demonstrate that there is little ical and emotional problems (all p values <0.004) than the
difference in HRQOL outcomes between men undergo- control subjects. Although the differences were small,
ing medical versus surgical castration. Litwin et al.52 this study demonstrates that localized prostate cancer
identified no differences in any of the general or disease- itself may affect quality of life.
specific domains of the RAND 36 Item Health Survey All of the treatments for localized prostate cancer are
(SF-36) or the UCLA Prostate Cancer Index when com- associated with side effects, such as sexual, urinary or
paring men who underwent orchiectomy to those receiv- bowel dysfunction, which can impact quality of life.
ing combined androgen blockade. Although the method Although there are differences in disease-specific
of castration does not appear to have a significant impact HRQOL outcomes between treatment, it appears there
on HRQOL, the presence or absence of additional is no difference in general HRQOL outcomes between
androgen blocker therapy does influence HRQOL. In a therapies, at least once the patient gets beyond the imme-
clinical trial of 739 men with stage M1 prostate cancer, diate posttreatment period. Penson et al.59 analyzed data
patients randomized to orchiectomy plus flutamide from PCOS, a population-based cohort of 5672 men
reported significantly more diarrhea than those who were with prostate cancer followed for 2 years after diagnosis
Chapter 6 Health-Related Quality of Life Issues in Urologic Oncology 107

of localized disease. Of the 2693 analyzed, no differences ing EBRT mentioned above, 65% reported sexual dys-
were seen comparing various treatments for localized function 8 years after EBRT.65 Similarly, another study
prostate cancer. Interestingly, men with sexual and uri- that noted actuarial potency rates of 96, 75, 59, and 53%
nary dysfunction had significantly worse outcomes in the at 1, 20, 40, and 60 months after EBRT.69 In summary,
bodily pain, mental health, role limitation, vitality, and EBRT also affects disease-specific HRQOL although
overall health status domains of general HRQOL, when somewhat differently than surgery.
controlling for baseline function, treatment, and 27 other While interstitial brachytherapy (IB) is similar to
co-variates. This underscores the importance of under- EBRT in that radiation is delivered to the prostate in
standing the effect of the various therapies for localized hopes of killing cancer cells, the effect of this treatment
prostate cancer on disease-specific HRQOL. on disease-specific HRQOL is different, given the fact
There is a significant body of literature examining that higher doses of radiation can be targeted to the
disease-specific HRQOL outcomes after treatment for prostate with IB. In particular, men undergoing IB are at
localized prostate cancer. A recent randomized, clinical increased risk for experiencing irritative voiding symp-
trial comparing radical prostatectomy (RP) to watchful toms. Lee et al.70,71 prospectively measured HRQOL in
waiting (WW), quality of life data were available in 189 31 men prior to IB and at 1, 3, 6, and 12 months
men who had RP and 187 who had WW (mean follow- posttreatment. At 3 months, patients were noted to have
up, 68 months). In the sexual domains, 55% of RP significant irritative voiding symptoms when compared
patients reported insufficient erections and 30% to baseline (15 versus 8, respectively), as measured by the
reported great distress due to their erectile dysfunction International Prostate Symptom Score (IPSS). By one
(as opposed to 30% and 17%, respectively, in the WW year, however, mean irritative voiding scores had
group). In urinary domains, 18% reported moderate- returned close to baseline (10 versus 8). Sexual and bowel
to-severe urinary leakage and 9% reported great dis- dysfunction can also occur after IB, although not to the
tress from this leakage (as opposed to 2% and 3%, same degree as is seen after EBRT. In one cross-sectional
respectively in the WW group). However, there were study, 35 patients who were potent pretreatment were
no differences between the two groups in overall dis- evaluated 6 months following IB. Eighty-seven percent
tress from urinary symptoms, presumably due to a higher retained their potency, while 3% reported rectal bleed-
incidence of obstructive symptoms in the WW patients.60 ing.72 With longer follow-up, Talcott et al.73 reported a
Stanford et al.61 studied the 1291 men in PCOS who 44% incidence of complete impotence, a 14% incidence
underwent RP for localized prostate cancer, noting that in rectal urgency and a 6% incidence of rectal bleeding.
8.4% of men were incontinent and 59.9% of men were Brandeis et al.74 compared 74 men who underwent radi-
impotent 2 years after surgery. Other studies have cal prostatectomy to 48 men who received brachytherapy
reported impotence rates ranging from 29%62 to 88.4%63 and 134 age-matched controls. They found that men
and incontinence rates from 4%64 to 33%63 after surgery. who underwent radical prostatectomy had significantly
In contrast to radical prostatectomy, patients undergo- worse outcomes in terms of urinary incontinence when
ing external beam radiotherapy (EBRT) tend to experi- compared to the brachytherapy patients. However,
ence less urinary incontinence and sexual dysfunction but patients who underwent brachytherapy had significantly
more bowel dysfunction and irritative voiding symptoms. more irritative voiding symptoms than those who under-
For example, researchers compared long-term HRQOL went prostatectomy. Interestingly, when asked about how
outcomes in 120 men who underwent EBRT to 125 much bother either type of urinary problem caused in the
age-matched controls.65 At 8 years after treatment, 54% patient’s life, there was no differences between the groups.
of patients and 31% of controls reported urinary prob- This study clearly demonstrates that men undergoing rad-
lems, most of which were irritative in nature. In another ical prostatectomy have more urinary incontinence than
study of 129 men undergoing EBRT, 15% reported uri- men undergoing brachytherapy, while those receiving
nary dysfunction to the extent that they had to modify brachytherapy report greater irritative voiding symptoms,
their activities of daily living and 2% felt virtually house- although both groups experience similar levels of bother.
bound by their irritative voiding symptoms.66 Crook et Recently, a number of cross-sectional studies have
al.67 studied bowel dysfunction in 192 men undergoing been published comparing disease-specific HRQOL
EBRT and noted that 25% reported moderate and 11% outcomes after surgery, external beam radiotherapy and
reported severe bowel changes. In another study of 200 interstitial brachytherapy. Although all three stud-
EBRT patients, 59% reported gastrointestinal problems, ies75–77 indicate that sexual dysfunction is common after
although 90% of these problems were classified as all treatments for localized prostate cancer, the results
minor.68 Finally, it is worth noting that, while the inci- in the urinary and bowel domains are somewhat contra-
dence of sexual dysfunction following EBRT appears to dictory. Wei et al.75 found no difference between IB and
be initially less than that following surgery, it is not RP in mean urinary function scores, while both Davis et
insignificant. In the study of long-term HRQOL follow- al.76 and Bacon et al.77 note significantly better urinary
108 Part I Principles of Urologic Oncology

function following IB when compared to surgery. This cer patients. Despite this, there are still a number of
may be related to differences in patient selection or reports examining HRQOL following treatment for
brachytherapy technique. Perhaps more interesting, muscle-invasive bladder cancer. As part of the rationale
however, are the results in the urinary bother domain, for performing orthotopic neobladder urinary diversion
where both Wei et al.75 and Bacon et al.77report less in these patients is to maintain normal body image and
bother in the surgery group than in the IB group. This quality of life, the majority of studies have compared
may be related to irritative voiding symptoms often HRQOL following differing types of urinary diversion.
seen after brachytherapy. Wei et al. compared HRQOL For example, Bjerre and colleagues78 compared men
outcomes in the urinary function:irritative domain and undergoing cystectomy followed by either ileal conduit
found men undergoing IB had significantly lower mean or orthotopic urethral Kock bladder substitution. They
scores (72 out of 100) when compared to men undergo- showed that urine leakage caused much less distress in
ing EBRT (84 out of 100) or surgery (90 out of 100). those who underwent bladder substitution, while body
Although these findings are useful when counseling image was surprisingly similar in both groups. Global
patients who are choosing therapy for localized prostate satisfaction with life was high and similar in both groups.
cancer, one must remember that all of these studies were Hunt Raleigh et al.79 compared HRQOL among bladder
observational in nature, which can introduce selection cancer patients treated with ileal loop or neobladder and
bias and may influence patient-centered outcomes, such found no significant differences noted in general
as HRQOL. Ultimately, a well-designed randomized HRQOL between the two groups, although patients
clinical trial is needed. (Table 6-3). with ileal loop diversion were more likely to report a neg-
ative impact on their social activity. Hara et al. also com-
pared HRQOL following orthotopic neobladder or ileal
Bladder Cancer
loop diversion and noted minimal differences in
There are considerably fewer reports studying HRQOL HRQOL outcomes.80 These studies, and others,81 imply
in bladder cancer patients. This is due, at least in part, to that the type of urinary diversion has little impact on
the lack of a validated HRQOL specific for bladder can- HRQOL outcomes in these patients.

Table 6-3 Results of Three Cross-sectional Studies Comparing Disease-specific Hrqol


after Treatment for Localized Prostate Cancer
Wei et al. study75 Bacon et al. study77 Davis et al. study76

Number of subjects

Surgery 570 421 220

EBRT 127 221 188

IB 61 69 103

Domains

Urinary function EBRT(93)*>IB(82)>RP(78) EBRT(89)*>IB(87)*>RP(76) IB(88)*>EBRT(87)*>RP(68)

Urinary bother RP(8%)>EBRT(10%)>IB(28%) EBRT(83)>RP(82)>IB(75)* EBRT(83)*>IB(77)>RP(74)

Bowel function RP(93)>EBRT(85)*>IB(76)*,† RP(86)>EBRT(81)*>IB(80)* RP(86)>IB(83)†>EBRT(77)*

Bowel bother RP(3%)>EBRT(7%)>IB(17%) RP(86)>EBRT(78)*>IB(72)* RP(83)>IB(79)>EBRT(72)*

Sexual function EBRT(39)>RP(34)>IB(27)† IB(36)*>EBRT(34)*>RP(26) IB(32)*>EBRT(26)*>RP(18)

Sexual bother EBRT(46%)>RP(50%)>IB(60%) IB(54)*>EBRT(51)*>RP(43) IB(40)*=EBRT(40)*>RP(25)

Note: Treatments are ranked by mean scores from best to worst, which are presented in the parenthesis. All HRQOL scores are 0 to 100 with
higher scores being better quality of life. The one exception to this is the results from the bother domains in the Wei et al. study. As these results
were not presented as summary scores, the numbers in parenthesis represent the percentage of patients who reported that symptoms in the
particular domain were a moderate or big problem. No statistical testing was performed on the bother domains from the Wei et al. study. External
beam radiotherapy and interstitial brachytherapy were not compared statistically in the Bacon et al. study.
IB, interstitial brachytherapy; EBRT, external beam radiotherapy; RP, radical prostatectomy.
*Statistically significantly different from radical prostatectomy at a p-value less than 0.05.
†Statistically significantly different from EBRT at a p-value less than 0.05.
Chapter 6 Health-Related Quality of Life Issues in Urologic Oncology 109

In contrast, others have noted better HRQOL follow- surgical technique used appear to influence short-term
ing continent urinary diversions, such as orthotopic HRQOL outcomes in kidney cancer.
neobladders. For example, Boyd et al.82 noted that self-
image was worse among patients with ileal conduits than
Testicular Cancer
in those with continent cutaneous Kock reservoirs,
although no differences were seen in mental or emotional There has been minimal HRQOL research in patients
health indices. Dutta et al.83 compared patients undergo- treated for testis cancer. Joly et al.93 compared long-term
ing orthotopic neobladder to those undergoing ileal loop HRQOL outcomes in testicular cancer survivors to age-
following cystectomy for bladder cancer. Although the matched controls and found no differences. In another
study was confounded by age and co-morbidity, the study, Fossa et al.94 studied HRQOL outcomes in men
patients undergoing neobladder were found to have mar- with good prognosis metastatic germ cell tumors. They
ginally better general HRQOL outcomes. Hardt et al.84 noted that two years after treatment, 36% of patients had
reported results from a prospective study of 44 patients improved general HRQOL, while general HRQOL had
undergoing radical cystectomy and either ileal loop deteriorated in 13% of patients. The remaining patients
(incontinent) diversion or continent diversion for bladder were effectively unchanged. Arai et al.95 used a Japanese
cancer. At 1 year after surgery, general HRQOL had translation of a questionnaire that had been validated in
returned to baseline in both groups, but general life sat- English to assess HRQOL in men treated for testicular
isfaction and social functioning were better in the conti- cancer. Patients treated with chemotherapy (with or
nent diversion group while they were decreased without retroperitoneal lymph node dissection), radio-
following incontinent diversion. Finally, McGuire et al.85 therapy, and surveillance were compared. Working abil-
compared HRQOL outcomes following incontinent or ity was better in the radiotherapy and chemotherapy
continent diversion and demonstrated that patients groups. These patients also reported a greater overall sat-
undergoing incontinent diversion had significantly isfaction with life than those in the surveillance group.
decreased mental health. While these studies must all be Weissbach et al.96 compared HRQOL outcomes in men
considered preliminary, they do provide some support undergoing retroperitoneal lymph node dissection
for the commonly held belief that continent diversions, (RPLND) or upfront chemotherapy for stage II non-
such as orthotopic neobladders, result in better quality of seminomatous germ cell tumors as part of a prospective,
life. More prospective data are needed to confirm this multicenter clinical trial. HRQOL outcomes were simi-
hypothesis. lar between the two groups, leading the authors to rec-
ommend surgery for these patients, as chemotherapy
could then be avoided in a considerable number of
Kidney Cancer
patients with little effect on quality of life.
There are surprisingly few reports on HRQOL in kidney
cancer patients. While a few of the reports of focused on
SUMMARY
HRQOL in patients with metastatic disease,86–89 recently
a number of reports have focused on HRQOL following HRQOL is an essential outcome for patients with geni-
various surgical techniques for removal of the primary tourinary malignancies. While patients are concerned
tumor. For example, Shinohara et al.90 compared with maximizing their life expectancy following a diag-
HRQOL outcomes following either radical or partial nosis of cancer, they are often just as concerned, if not
nephrectomy for renal cell carcinoma. They noted that, more so, with maintaining their quality of life after treat-
while there were no differences in long-term survival or ment. Clinicians and researchers must be sensitive to this
HRQOL, patients undergoing partial nephrectomy had and focus more attention of the effects of therapy on can-
better physical function in the immediate postoperative cer survivors’ quality of life.
period than those undergoing radical nephrectomy. Clark
et al.91 performed a similar study. While they noted no
difference in overall HRQOL between partial and radical REFERENCES
nephrectomy patients, they did find that patients with
more intact renal parenchyma reported better physical 1. Fayers PM, Jones DR: Measuring and analyzing quality of
life in cancer clinical trials: a review. Stat Med 1983; 2:429.
health. Finally, Pace et al.92 compared HRQOL following
2. Patrick DL, Erickson P: Assessing health-related quality of
laparoscopic or open radical nephrectomy. Again, while life for clinical decision-making. In Walker SR, Rosser RM
there were no differences in long-term general HRQOL (eds): Quality of Life Assessment: Key Issues in the 1990s,
outcomes, patients undergoing laparoscopic nephrectomy Chap. 19. Dordrecht, Kluwer Academic Publishers, 1993.
reported better HRQOL immediately postoperatively 3. Sadura A, Pater J, Osoba D, et al: Quality of life
and returned to their baseline HRQOL state quicker. assessment: patient compliance with questionnaire
These studies indicate that the type of operation and the completion. J Natl Cancer Inst 1992; 84:1023.
110 Part I Principles of Urologic Oncology

4. Altwein J, Ekman P, Barry M, et al: How is quality of life 22. Kaplan RM, Bush JW, Berry CC: Health status: types of
in prostate cancer patients influenced by modern validity and the index of well-being. Health Ser Res 1976;
treatment? The Wallenberg Symposium. Urology 1997; 11:478.
49:66. 23. Kaplan RM, Bush JW: Health-related quality of life
5. Litwin MS: Measuring health related quality of life in measurement for evaluation research and policy analysis.
men with prostate cancer. J Urol 1994; 152:1882. Health Psychol 1982; 1:61.
6. Tulsky DA: An introduction to test theory. Oncology 24. Kaplan RM, Anderson JP: A general health policy model:
1990; 4:43. update and applications. Health Ser Res 1988; 23.
7. Patrick DL, Deyo RA: Generic and disease-specific 25. Hays RM, Shapiro MF: An overview of generic health-
measures in assessing health care status and quality of life. related quality of life measures for HIV research. Quality
Med Care 1989; 27(Suppl):S217. Life Res 1992; 1: 91-97.
8. Cronbach LJ: Coefficient alpha and the internal structure 26. Deyo RA, Inui TS, Leininger JD, et al: Measuring
of tests. Psychometrika 1951; 16:297. functional outcomes in chronic disease: a comparison of
9. Nunnally JC: Psychometric Theory, 2nd edition New traditional scales and a self-administered health status
York, McGraw-Hill, 1978. questionnaire in patients with rheumatoid arthritis. Med
10. Messick S: The once and future issues of validity: Care 1983; 21:180.
assessing the meaning and consequences of measurement. 27. Bergner M, Bobbitt RA, Carter WB, et al: The sickness
In Wainer H, Braun HI (eds):Test Validity, Hillside, NJ, impact profile: development and final revision of a health
Lawrence Erlbaum Associates, 1988. status measure. Med Care 1981; 19:787.
11. Tannock IF: Management of breast and prostate cancer: 28. Moinpour CM, Feigl P, Metch B, et al: Quality of life end
how does quality of life enter the equation? Oncology points in cancer clinical trials: review and
1990; 4:149. recommendations. J Natl Cancer Inst 1989; 81:485.
12. Litwin MS, Lubeck DP, Henning JM, et al: Differences in 29. Hunt SM, McEwen J, McKenna SP: Measuring health
urologist and patient assessments of health related quality status: a new tool for clinicians and epidemiologists.
of life in men with prostate cancer: results of the J Royal College General Practitioners 1985; 35:185.
CaPSURE database. J Urol 1998; 159:1988. 30. McDowell IW, Martini CJM, Waugh W: A method for
13. Bennett CL, Chapman G, Elstein AS, et al: A comparison self-assessment of disability before and after hip
of perspectives on prostate cancer: analysis of utility replacement operations. Br Med 1978; J2:57.
assessments of patients and physicians. Eur Urol 1997; 31. Martini CJ, McDowell I: Health status: patient and
32:86. physician judgements. Health Ser Res 1976; 11:508.
14. Crawford ED, Bennett CL, Stone NN, et al: Comparison 32. Aaronson NK, Ahmedzai S, Bergman B, et al: The
of perspectives on prostate cancer: analyses of survey data. European Organization for Research and the treatment of
Urology 1997; 50:366. Cancer QLQ-C30: a quality of life instrument for use in
15. Parkerson GR Jr, Connis RT, Broadhead WE, et al: international clinical trials in oncology. J Natl Cancer Inst
Disease-specific versus generic measurement of health- 1993; 85:356.
related quality of life in insulin-dependent diabetic 33. Borghede G, Sullivan M: Measurement of quality of life
patients. Med Care 1993; 31:629. in localized prostatic cancer patients treated with
16. Tarlov AR, Ware JE Jr, Greenfield S, et al: The Medical radiotherapy. Development of a prostate cancer-specific
Outcomes Study. an application of methods for monitoring module supplementing the EORTC QLQ-C30. Quality
the results of medical care. JAMA 1989; 262:925. Life Res 1996; 5:212.
17. Ware JE, Sherbourne CD, Davies AR: Developing and 34. Borghede G, Karlsson J, Sullivan M: Quality of life in
testing the MOS 20-item short-form health survey: a patients with prostatic cancer: results from a Swedish
general population application. In Stewart AL, Ware JE population study. J Urol 1997; 158:1477.
(eds): Measuring Functioning and Well-Being: The 35. Albertsen PC, Aaronson NK, Muller MJ, et al: Health-
Medical Outcomes Study Approach, Durham, NC, Duke related quality of life among patients with metastatic
University Press, 1992 prostate cancer. Urology 1997; 49:207.
18. Stewart AL, Hays RD, Ware JE: The MOS short-form 36. Cella DF, Tulsky DS: Measuring quality of life today.
general health survey: reliability and validity in a patient Oncology 1990; 4:29.
population. Med Care 1988; 26:724. 37. Tulsky DS, Cella DF, Bonomi A, et al: Development and
19. Ware JE Jr, Sherbourne CD: The MOS 36-item short- validation of new quality of life measures for patients with
form health survey (SF-36). I. Conceptual framework and cancer. Proc Soc Behav Med 0990; 11:45.
item selection. Med Care 1992; 30:473. 38. Cella DF, Cherin EA: Quality of life during and after
20. McHorney CA, Ware JE Jr, Rogers W, et al: The validity cancer treatment. Compreh Therapy 1988; 14:68.
and relative precision of MOS short- and long-form 39. Cella DF, Orofiamma B, Holland JC, et al: Relationship of
health status scales and Dartmouth COOP charts. Results psychological distress, extent of disease, and performance
from the Medical Outcomes Study. Med Care 1992; status in patients with lung cancer. Cancer 1987; 60:239.
30:MS253. 40. Esper P, Mo F, Chodak G, et al: Measuring quality of life
21. McDowell I, Ewell C: Measuring Health: A Guide to in men with prostate cancer using the functional
Rating Scales and Questionnaires. New York, Oxford assessment of cancer therapy-prostate instrument.
University Press, 1987. Urology 1997; 50:920.
Chapter 6 Health-Related Quality of Life Issues in Urologic Oncology 111

41. Litwin MS, Hays RD, Fink A, et al: Quality-of-life 58. Bacon CG, Giovannucci E, Testa M, et al: The
outcomes in men treated for localized prostate cancer. association of treatment-related symptoms with quality-
JAMA 1995; 273:129. of-life outcomes for localized prostate carcinoma patients.
42. Litwin MS, Hays RD, Fink A, et al: The UCLA Prostate Cancer 2002; 94:862.
Cancer Index: development, reliability, and validity of a 59. Penson DF, Feng Z, Kuniyuki A, et al: General quality of
health-related quality of life measure. Med Care 1998; life 2 years following treatment for prostate cancer: what
36:1002. influences outcomes? Results from the Prostate Cancer
43. Krongrad A, Perczek RE, Burke MA, et al: Reliability of Outcomes Study. J Clin Oncol 2003; 21:1147.
Spanish translations of select urological quality of life 60. Steineck G, Helgesen F, Adolfsson J, et al: Quality of life
instruments. J Urol 1997; 158:493. after radical prostatectomy or watchful waiting. N Engl J
44. Litwin MS, Fink A, Hays RD, et al: Quality of life in men Med 2002; 347:790.
with prostate cancer: a pilot study. J Urol 1993; 149:494A. 61. Stanford JL, Feng Z, Hamilton AS, et al: Urinary and
45. Wei JT, Dunn RL, Litwin MS, et al: Development and sexual function after radical prostatectomy for clinically
validation of the expanded prostate cancer index localized prostate cancer: the Prostate Cancer Outcomes
composite (EPIC) for comprehensive assessment of Study. JAMA 2000; 283:354.
health-related quality of life in men with prostate cancer. 62. Steiner MS: Current results and patient selection for
Urology 2000; 56:899. nerve-sparing radical retropubic prostatectomy. Semin
46. Ware J Jr, Kosinski M, Keller SD: A 12-Item Short-Form Urologic Oncol 1995; 13:204.
Health Survey: construction of scales and preliminary 63. Kao T, Cruess D, Garner D, et al: Multicenter patient
tests of reliability and validity. Med Care 1996; 34:220. self-reporting questionnaire on impotence, incontinence
47. Curran D, Fossa S, Aaronson N, et al: Baseline quality of and stricture after radical prostectomy. J Urol 2000;
life of patients with advanced prostate cancer. European 163:858.
Organization for Research and Treatment of Cancer 64. Murphy G, Mettlin C, Menck H, et al: National patterns
(EORTC), Genito-Urinary Tract Cancer Cooperative of prostate cancer treatment by radical prostatectomy:
Group (GUT-CCG). Eur J Cancer 1997; 33:1809. results of a survey by the American College of Surgeons
48. Kim SP, Bennett CL, Chan C, et al: QOL and outcomes Commission on Cancer. J Urol 1994; 152:1817.
research in prostate cancer patients with low 65. Fransson P, Widmark A: Late side effects unchanged 4–8
socioeconomic status. Oncology 1999; 13:823. years after radiotherapy for prostate carcinoma: a
49. Herr HW, O’Sullivan M: Quality of life of asymptomatic comparison with age-matched controls. Cancer 1999;
men with nonmetastatic prostate cancer on androgen 85:678.
deprivation therapy. J Urol 2000; 163:1743. 66. Franklin CI, Parker CA, Morton KM: Late effects of
50. Green HJ, Pakenham KI, Headley BC, et al: Coping and radiation therapy for prostate carcinoma: the patient’s
health-related quality of life in men with prostate cancer perspective of bladder, bowel and sexual morbidity.
randomly assigned to hormonal medication or close Australas Radiol 1998; 42:58.
monitoring. Psychooncology 2001; 11:401. 67. Crook J, Esche B, Futter N: Effect of pelvic radiotherapy
51. Potosky AL, Reeve BB, Clegg LX, et al: Quality of life for prostate cancer on bowel, bladder, and sexual function:
following localized prostate cancer treated initially with the patient’s perspective. Urology 1996; 47:387.
androgen deprivation therapy or no therapy. J Natl 68. Widmark A, Fransson P, Tavelin B: Self-assessment
Cancer Inst 2002; 94:430. questionnaire for evaluating urinary and intestinal late
52. Litwin MS, Shpall AI, Dorey F, et al: Quality-of-life side effects after pelvic radiotherapy in patients with
outcomes in long-term survivors of advanced prostate prostate cancer compared with an age-matched control
cancer. Am J Clin Oncol 1998; 21:327. population. Cancer 1994; 74:2520.
53. Moinpour CM, Savage MJ, Troxel A, et al: Quality of life 69. Mantz CA, Nautiyal J, Awan A, et al: Potency
in advanced prostate cancer: results of a randomized preservation following conformal radiotherapy for
therapeutic trial [see comments]. J Natl Cancer Inst 0998; localized prostate cancer: impact of neoadjuvant
90:1537. androgen blockade, treatment technique, and patient-
54. Tyrrell CJ: Tolerability and quality of life aspects with the related factors [see comments]. Cancer J Scientific Am
anti-androgen Casodex (ICI 176,334) as monotherapy for 1999; 5:230.
prostate cancer. International Casodex Investigators. Eur 70. Lee WR, McQuellon RP, Harris-Henderson K, et al: A
Urol 1994; 26:15. preliminary analysis of health-related quality of life in the
55. Osoba D, Tannock IF, Ernst DS, et al: Health-related first year after permanent source interstitial brachytherapy
quality of life in men with metastatic prostate cancer (PIB) for clinically localized prostate cancer. Int J
treated with prednisone alone or mitoxantrone and Radiation Oncol Biol Phys 2000; 46:77.
prednisone [see comments]. J Clin Oncol 1999; 17:1654. 71. Lee WR, McQuellon RP, Case LD, et al: Early quality of
56. Litwin MS, Lubeck DP, Stoddard ML, et al: Quality of life assessment in men treated with permanent source
life before death for men with prostate cancer: results interstitial brachytherapy for clinically localized prostate
from the CaPSURE database. J Urol 2001; 165:871. cancer. J Urol 1999; 162:403.
57. Turner SL, Gruenewald S, Spry N, et al: Less pain does 72. Arterbery VE, Frazier A, Dalmia P, et al: Quality of life
equal better quality of life following strontium-89 therapy after permanent prostate implant. Semin Surg Oncol
for metastatic prostate cancer. Br J Cancer 2001; 84:297. 1997; 13:461.
112 Part I Principles of Urologic Oncology

73. Talcott JA, Clark JA, Stark PC, et al: Long-term impacts on the patient’s quality of life. Ann Surg Oncol
treatment related complications of brachytherapy for early 2000; 7:4.
prostate cancer: a survey of patients previously treated. 86. Litwin MS, Fine JT, Dorey F, et al: Health related quality
J Urol 2001; 166:494. of life outcomes in patients treated for metastatic kidney
74. Brandeis J, Litwin M, Burnison C, et al: Quality of life cancer: a pilot study. J Urol 1997; 157:1608.
outcomes after brachytherapy for early stage prostate 87. Kroger MJ, Menzel T, Gschwend JE, et al: Life quality of
cancer. J Urol 2000; 163:851. patients with metastatic renal cell carcinoma and chemo-
75. Wei JT, Dunn RL, Sandler HM, et al: Comprehensive immunotherapy–a pilot study. Anticancer Res 1999;
comparison of health-related quality of life after 19:1553.
contemporary therapies for localized prostate cancer. 88. Heinzer H, Mir TS, Huland E, et al: Subjective and
J Clin Oncol 2002; 20:557. objective prospective, long-term analysis of quality of life
76. Davis JW, Kuban DA, Lynch DF, et al: Quality of life during inhaled interleukin-2 immunotherapy. J Clin
after treatment for localized prostate cancer: differences Oncol 1999; 17:3612.
based on treatment modality. J Urol 2001; 166:962. 89. Osband ME, Lavin PT, Babayan RK, et al: Effect of
77. Bacon CG, Giovannucci E, Testa M, et al: The impact autolymphocyte therapy on survival and quality of life in
of cancer treatment on quality of life outcomes for patients with metastatic renal-cell carcinoma. Lancet
patients with localized prostate cancer. J Urol 2001; 1990; 335:994.
166:1804. 90. Shinohara N, Harabayashi T, Sato S, et al: Impact
78. Bjerre BD, Johansen C, Steven K: Health-related quality of nephron-sparing surgery on quality of life in patients
of life after cystectomy: bladder substitution compared with localized renal cell carcinoma. Eur Urol 2001;
with ileal conduit diversion. A questionnaire survey. Br J 39:114.
Urol 1995; 75:200. 91. Clark PE, Schover LR, Uzzo RG, et al: Quality of life and
79. Hunt Raleigh ED, Berry M, Montie JE: A comparison of psychological adaptation after surgical treatment for
adjustments to urinary diversions: a pilot study. JWOCN localized renal cell carcinoma: impact of the amount of
1995; 22:58. remaining renal tissue. Urology 2001; 57:252.
80. Hara I, Miyake H, Hara S, et al: Health-related quality of 92. Pace KT, Dyer SJ, Stewart RJ, et al: Health-related
life after radical cystectomy for bladder cancer: a quality of life after laparoscopic and open nephrectomy.
comparison of ileal conduit and orthotopic bladder Surg Endosc 2003; 17:143.
replacement. Br J Urol Int 2002; 89:10. 93. Joly F, Heron JF, Kalusinski L, et al: Quality of life in
81. Fujisawa M, Isotani S, Gotoh A, et al: Health-related quality long-term survivors of testicular cancer: a population-
of life with orthotopic neobladder versus ileal conduit based case-control study. J Clin Oncol 2002; 20:73.
according to the SF-36 survey. Urology 2000; 55:862. 94. Fossa SD, De Wit R, Roberts JT, et al: Quality of Life in
82. Boyd S, Feinberg SM, Skinner DG, et al: Quality of life Good Prognosis Patients With Metastatic Germ Cell
survey of urinary diversion patients: comparison of ileal Cancer: A Prospective Study of the European
conduits versus continent Kock ileal reservoirs. J Urol Organization for Research and Treatment of Cancer
1987; 138:1386. Genitourinary Group/Medical Research Council
83. Dutta SC, Chang SC, Coffey CS, et al: Health related Testicular Cancer Study Group (30941/TE20). J Clin
quality of life assessment after radical cystectomy: Oncol 2003; 21:1107.
comparison of ileal conduit with continent orthotopic 95. Arai Y, Kawakita M, Hida S, et al: Psychosocial aspects in
neobladder. J Urol 2002; 168:164. long-term survivors of testicular cancer. J Urol 1996;
84. Hardt J, Filipas D, Hohenfellner R, et al: Quality of life 155:574.
in patients with bladder carcinoma after cystectomy: 96. Weissbach L, Bussar-Maatz R, Flechtner H, et al:
first results of a prospective study. Quality Life Res RPLND or primary chemotherapy in clinical stage IIA/B
2000; 9:1. nonseminomatous germ cell tumors? Results of a
85. McGuire MS, Grimaldi G, Grotas J, et al: The type of prospective multicenter trial including quality of life
urinary diversion after radical cystectomy significantly assessment. Eur Urol 2000; 37:582.
C H A P T E R

7 Image-Guided Minimally
Invasive Therapy
Agnieszka Szot Barnes, M.D., M.S.,
and Clare M.C. Tempany, M.B., B.A.O., B.Ch.

The field of image-guided minimally invasive procedures and computer experts combine efforts to integrate imag-
has undergone a revolutionary change in the past decade. ing systems with therapy systems.
We have seen the development of advanced image- Image-guided minimally invasive therapy is experi-
guided therapies for treatment of many different diseases, encing rapid growth driven by the introduction of new
ranging from brain tumor resection and treatment to imaging modalities and significant improvement of exist-
magnetic resonance (MR)-guided prostate brachytherapy ing ones as well as improving computer performance.
and MR-monitored thermal therapies, such as cryother- The most important advancements of this field include
apy. In the field of urologic oncology, today there are integrating preprocedure and intraprocedure imaging,
many image-guided procedures for obtaining diagnoses further improving image quality, and testing the usability
and guiding and delivering treatment. These range from of the techniques in clinical settings.
simple biopsies to image-guided tumor ablations.
Minimally invasive therapy is used to treat the disease
HISTORY
by operating through natural body openings or small
incisions, thereby reducing the cosmetic or loco-regional IGT evolved over the years with major advances in image-
tissue damage and the potential complications of open guided neurosurgery spreading to other disciplines,
surgery. By reducing the need for invasive surgery, hospi- including urologic oncology. In many cases, brain tumors
talization is shortened, with fewer complications and may visually resemble healthy tissue to the naked eye or
faster recovery. These procedures have been allowed by the extent of tumor invasion may be obscured by overlying
the development of improved surgical techniques and, healthy tissue. Before image guidance the procedures were
perhaps more importantly, improved imaging tech- performed without proper visualization of the extent of
niques. Because direct visualization without surgical the tumor or its specific geometry.
intervention is not possible, the ability to combine mul- In the past decade or so, MR imaging (MRI) and com-
tiple imaging modalities to plan and execute the surgery puted tomography (CT) techniques have improved enor-
has permitted the full use of the new surgical techniques. mously. We have seen rapid MR scanners with high field
This combination of imaging and therapy is known as strengths become standard clinical tools in many radiol-
image-guided therapy (IGT). ogy departments around the world. New multidetector
The purpose of IGT is to integrate the anatomic and CT scanners allow rapid acquisition of high-resolution
physiologic information acquired before treatment with CT data sets that can now be reconstructed in coronal or
the therapy methods and allow the control and guidance sagittal planes. As the technology has advanced, the impact
of the treatment while it is being performed to improve of the image data has expanded. Now imaging alone diag-
the accuracy of treatment delivery. Not only can image- noses nearly all renal cell carcinomas. Imaging alone stages
guidance improve targeting of cancer tissue during ther- the extent of vascular invasion by a renal cell tumor and
apy but it can also spare adjacent tissues and organs from plans the surgical approach. Three-dimensional (3D)
being damaged during treatment. IGT is a multidiscipli- reconstructions allow the surgeon to determine the feasi-
nary field in which surgeons, radiologists, oncologists, bility of a partial nephrectomy.

113
114 Part I Principles of Urologic Oncology

The imaging of solid organs, both to identify pathology


and to accurately locate critical structures, has become the
province of x-ray CT and MRI. CT has been used prima-
rily for guiding biopsies, although the advent of “CT flu-
oroscopy” has stimulated use in guiding interventional
procedures, such as radiofrequency (RF) ablation.
Intraoperative ultrasound (US) has been used with
increasing success for many decades, particularly in the
imaging of solid organs, which can be directly con-
tacted by the probe, giving both excellent imagery and
explicit orientation of the image. US is particularly
useful in observing vascular structures, which are both
important landmarks and vital structures to be avoided
during the resection of solid organs. Laparoscopic US
shares the imaging advantages of intraoperative use,
but due to the small size of the imaging head and the
offset required for endoscopic insertion, it could be
more difficult to interpret the content and orientation Figure 7-1 Open 0.5 T MR system for performing image-
of the images. guided procedures.
The penetration of real-time CT and MRI into the
broad range of surgical procedures has been slow, due to
the complexity and cost of their implementation and dif- these scanners is the difficulty they pose to accessing
ficult access to the patient. In the case of MRI, additional the patient during the procedure, while their main
obstacles have been the incompatibility of surgical tools, advantage is higher field strength and therefore better
devices and operating room equipment with the magnetic image quality.
field environment and the challenge of interpreting the In the field of urology, IGT has advanced significantly
MR image, which may require extensive training and/or from lithotripsy—shockwave removal of kidney stones
expert consultation. under US or fluoroscopy guidance—to MR-guided pro-
The attractiveness of MRI for guiding simple proce- cedures. While fluoroscopy remains a very popular
dures, such as biopsies, was recognized as early as the method of image guidance in urology, it can expose
mid-1980s. Many of the initial obstacles of real-time both the patient and the physician to radiation. The
MRI guidance were overcome when an open MR scanner advances in CT imaging made it possible to perform
was introduced to guide neurosurgical procedures in CT fluoroscopy in real time. These advances, including
Brigham and Women’s Hospital in Boston in December 3D reconstruction, play a large role in the guidance of
1993. This new revolutionary method was envisioned by urologic procedures, including CT-guided tumor abla-
Dr. Ferenc Jolesz—radiologist and neurosurgeon—in tions and CT-guided prostate brachytherapy and
1987 when he began to put together a team of collabora- biopsy. Ultrasound guidance still remains very popular
tors to create the “operating room of the future.”1 The in clinical urology mainly because of its lower cost and
0.5 T intraoperative MR scanner was designed by GE portability, the possibility of real-time imaging, and the
Medical Systems (Signa SP) and installed in a designated safety for both patient and urologist. Currently, many
MR therapy (MRT) suite.2 The scanner has a vertical gap centers successfully use transrectal US (TRUS) to guide
that allows the physician to enter between the two mag- prostate biopsies and brachytherapy. MR has unique
nets and makes it easy to access the patient to perform assets as a guidance modality, allowing not only target
treatment (Figure 7-1). Images are generated using fast identification but also therapy monitoring. This is best
sequences resulting in near real-time imaging without illustrated by MR-guided focused ultrasound (MRgFUS).
disruption of the procedure. Initially, the scanner was Finally, advances in MR imaging include image recon-
used to guide percutaneous or transcranial biopsies but struction in multiple planes, a higher signal-to-noise
currently it is utilized in a variety of procedures from ratio that allows excellent differentiation between tis-
neurosurgery to prostate brachytherapy and biopsy. sues, and new contrast agents to make feasible MR-
Since then, many other open configuration magnets guided diagnostic techniques, such as MR angiography
have been introduced, including 0.2 T vertical-type mag- and MR spectroscopy. The creation of open interven-
nets (Picker, Siemens) and shorter-bore magnets (Philips, tional MR also made MR guidance possible for several
Picker). Conventional 1.5 T magnets are also used to urologic procedures, including prostate brachytherapy
guide various procedures. The main disadvantage of and biopsy.3
Chapter 7 Image-Guided Minimally Invasive Therapy 115

OVERVIEW OF CURRENT MR-GUIDED


injection of MR contrast agents (such as Gadolinium-
IMAGE-GUIDED THERAPY APPLICATIONS
DTPA); relative peak enhancement, time to peak, and
The field of MR imaging for guiding interventions and washout are of great importance in distinguishing and char-
therapy is attracting considerable research attention. MRI acterizing cancer.7–9 Added value comes from spec-
is superior to any other method in brain tumor localization troscopy, where metabolic differences can distinguish
and assessment and therefore is an excellent method for between cancer and healthy tissues. Normal prostate
surgical guidance. Tumor margins and extent can be well metabolism is characterized by high citrate and low
defined, which in turn provides the possibility of complete choline/creatinine levels, whereas in cancerous tissue these
tumor eradication with minimal damage to healthy brain ratios are reversed.10 Prostate multivoxel spectra convey-
tissue. MR-guidance for neurosurgery provides substantial ing metabolic information are superimposed on endorec-
help in performing brain tumor surgery.3 The use of com- tal and multiphase-array MR anatomic images, allowing
puterized navigation and 3D modeling further enhances for precise localization of the tumor. Prostate imaging is
precise tumor resection. now moving towards use of higher-field 3 T scanners,
These improvements in MR-guided neurosurgery which provide images with higher signal-to-noise ratio,
techniques, including 3D modeling, have provided a which in turn allow for better visualization of prostatic
framework for an MR-guided prostate intervention pro- substructures and increased MR-spectroscopy resolution.
gram guiding prostate cancer therapy with interstitial The development of an interventional MR therapy
brachytherapy—the permanent placement of radioactive (MRT) system has made it possible to perform prostate
sources (commonly I-125) directly into the prostate. brachytherapy under MR guidance. Even at lower field
Prostate MRI, especially with combined endorectal and strength than is routinely used for prostate cancer imag-
phase-array coils, provides images of even higher resolu- ing (0.5 T versus 1.5 T), MRI provides images of good
tion and is used in prostate cancer staging, as well as in quality for target visualization, as well as identification of
determination of extraprostatic disease with up to 82% the urethra and rectum (see Figure 7-2 for comparison of
accuracy.5,6 The T1- and T2-weighted images are helpful 1.5 T and 0.5 T images). Computer software has been
in differentiating between postbiopsy hemorrhage, which developed to provide dosimetry analysis, used for both
presents as a high T1 and a low T2 lesion, and prostate treatment planning and monitoring based on intraopera-
cancer, which presents as a low T1 and a low T2 lesion. tive MR images.11 Image-processing methods adapted
Contrast-enhanced images and prostate spectroscopy are from brain surgery are available to further facilitate pre-
of great importance in distinguishing between normal and cise radiation delivery to the prostate gland while sparing
cancerous tissues. Bladder and prostate cancers may expe- surrounding tissues. Currently, treatment delivery with a
rience higher perfusion than does normal tissue, which is robot assistance system is being developed and tested to
detected as signal enhancement following intravenous improve radioactive seed placement.

Figure 7-2 Prostate gland segmentation and registration. Segmentation identifies PZ


(solid arrow) and central gland (hollow arrow) in pre-therapy endorectal coil MR (1.5 T, left)
and MR-guided therapy images 0.5 T, right). Registration matches the segmented areas
in the different images.
116 Part I Principles of Urologic Oncology

After installation of the first interventional magnet at display and processing, especially when using MR pro-
Brigham and Women’s hospital in 1993, several other cen- cessing. As the image processing technology improved,
ters were created at teaching hospitals around the country, feedback from imaging to the therapy became possible
including Stanford University and the University of and the role of imaging became more prominent, using
Mississippi. Since then the number of centers has grown MRI for intraoperative guidance as well as diagnosis. As
and now includes many more sites in the U.S. and overseas. the processing improved, imaging became almost instan-
A great strength of MRI lies in its sensitivity to tem- taneous or “real-time,” allowing for tight integration of
perature changes.12–14 This sensitivity allows specialists imaging and therapy.
to monitor in real-time the delivery of several thermal There are several components critical to all image-
energy treatments, including RF and laser therapy for guided therapies; these are: planning, targeting, naviga-
brain tumors, and cryotherapy and high-intensity FUS tion, control, and monitoring. Pretreatment planning
for breast, prostate, liver, and uterine lesions. Currently, allows for the assessment of the approach that will pro-
MRI is a very useful guidance method for cryotherapy— vide the most effective eradication of the tumor and at
tumor ablation by use of freezing—because it allows for the same time the least damage to the surrounding vital
monitoring of the location and size of the ice ball in mul- organs. For example, when delivering radiation therapy,
tiple dimensions.4 Intraoperative MR images are used to pretreatment dosimetry planning determines the volume
depict the slow expansion of the ice ball, as well as tissue of the target and the placement of radiation sources.
damage caused by the freezing process. Targeting during IGT refers to precise pretreatment
MR-guided FUS is a very promising method for non- localization of all tumor targets that need to be treated
invasive cancer treatment. While other minimally inva- and is essential for precise navigation of equipment dur-
sive therapies require direct insertion of special probes to ing treatment delivery. Navigation refers to the guidance
reach the tumor, this method uses a high-intensity US of surgical equipment during procedures to target the
beam focused on the target lesion (as seen on the MR tumors precisely and spare healthy tissue. Current
image) without disruption of skin and other tissues. FUS research efforts in the field of navigation are directed
is based on the use of acoustic energy and its secondary towards automatization of the procedure and increasing
thermal effect, which cause thermal coagulation of the the use of surgical robots.19,20 Real-time controlling and
target tissue. As early as 1955, it was clinically shown that monitoring of the treatment delivery by means of intra-
FUS was capable of destroying mammalian tissues.15 operative imaging allows for necessary adjustments of the
Broad use of this treatment method has been hindered by therapy to reflect movement of the tissues and permits
a lack of appropriate image-guidance techniques for the alterations of he plan in response to initial therapy.
tumor-targeting, and most importantly for the real-time Many different image postprocessing techniques have
monitoring of temperature changes. The introduction of been developed to allow use of anatomic and functional
MR guidance provided an excellent method for monitor- information to improve tumor detection and treatment
ing treatment planning and delivery with direct temper- planning. These techniques include image segmentation,
ature mapping (using MR phase-contrast techniques), as fusion, and registration. The purpose of image segmenta-
well as posttreatment confirmation of necrotic tissue tion is to distinguish organs or structures of interest (e.g.,
changes.6 Using the MR images, the physician can iden- prostate or its peripheral zone [PZ]) from the surround-
tify the target lesions; temperature change during treat- ing organs and tissue in order to perform volumetric and
ment delivery is monitored using MR temperature maps. shape analyses, as well as treatment planning (see Figure
A special transducer moves from one spot to another, fol- 7-2). This is currently done by either manual outlining of
lowing a pretreatment plan, until the entire volume is the structure of interest or by semiautomated methods.21
treated. To date, this method has been successfully used The future of medical image segmentation is to automate
in the treatment of breast fibroadenomas, breast cancers, the process and replace manual segmentation.22,23
and uterine leiomyomas.16–18 Application of this treat- Registration is a technique used to match images taken using
ment to prostate cancer, liver lesions, and brain tumors is different modalities, the same modality at different time
currently under investigation. points, or different imaging sequences (see Figure 7-2).
The process involves mapping one image into the coordi-
nate system of another image. Fusion is the merging of
IMAGE-GUIDED THERAPY: ROLE OF IMAGE
the anatomic and functional information provided by dif-
PROCESSING
ferent imaging modalities into a single volume in order to
The key to IGT is the integration of a coordinated image provide better information about the underlying anatomy
process with the therapy process. Early problems with and tissue characteristics (Figure 7-3). Applications for
IGT included lack of integration of the imaging with fused images include not only IGT but also minimally
therapy instruments, as well as difficulties with image invasive diagnosis and treatment planning.
Chapter 7 Image-Guided Minimally Invasive Therapy 117

Figure 7-3 MR-CT image fusion of prostate gland. Registration allows fusion of MR image
(left) with CT image to yield fused image (right). Black arrows indicate radioactive seeds.

IMAGE-GUIDED MINIMALLY INVASIVE THERAPY


Minimally invasive image-guided procedures for early
IN UROLOGY
stage organ-confined prostate cancer include diagnosis
Many of the image-guided minimally invasive therapies using US-, CT- and MR-guided biopsy; and therapy
in urologic oncology are directed toward diagnosis and using cryotherapy, CT-guided brachytherapy (CTBT),
treatment of prostate cancer. 2D transrectal US-guided brachytherapy (USBT)—both
In prostate procedures, IGT and diagnosis can be with and without external beam radiation therapy, with
guided by different image modalities; transrectal TRUS and without neo-adjuvant hormonal therapy; MR-guided
(TRUS) is the most widely used method. TRUS provides brachytherapy (MRBT), with and without external beam
good delineation of the prostate margin, simplicity of radiation therapy; and FUS.
imaging, relatively low cost compared to other modali- In general, the group of patients that may benefit
ties, and availability. TRUS is a widely used technique for from IGT as monotherapy for prostate cancer is com-
guidance of both prostate biopsies and brachytherapy. prised of men with organ-confined disease. Lieberfarb
However, for prostate cancer diagnosis, the positive pre- et al.26 showed that in low-risk patients with clinical stage
dictive value of this method remains quite low (17% to ≤ T2a (according to the American Joint Commission on
57% for hypoechoic lesions, as summarized by Boges et Cancer Staging—AJCC system), PSA ≤10 ng/ml, and
al.24). Currently, research focuses on improving the accu- ≤ 50% positive biopsies, the likelihood of extracapsular
racy of TRUS in the detection and staging of prostate extension (ECE) with or without positive margins was
cancer, including features such as power and color 18%, and seminal vesicle involvement was 2%. These
Doppler, 3D imaging, and elastography. patients may be “ideal” candidates for IGTs. For an
CT guidance was used primarily for prostate biopsies overview of prostate therapy outcomes see Jani and
but has also been introduced in prostate cancer therapy Hellman (2003)27 and Peschel and Colberg (2003).28
guidance.25 CT provides visualization of prostate bound-
aries and with the placement of a Foley catheter in the
Cryotherapy
bladder allows for good visualization of the urethra that
helps avoid urethral damage during treatment delivery. Cryotherapy refers to the application of low tempera-
MR guidance of prostate procedures grew in impor- tures to necrotize the tumor. In addition to its use as a
tance after the development of the interventional MR primary treatment for prostate cancer it has also been
scanner described earlier. Compared to US and CT used as a salvage therapy after failure of radiation ther-
imaging, MR imaging provides superior visualization of apy.29–31 Cryosurgery was first proposed as a treatment
the prostate and its zonal anatomy, tumor location, and for prostate disease in 1966.30 In the following years,
surrounding vital organs like the rectum, neurovascular several open transperineal procedures were performed
bundles (NVBs), and urethra. under visual control. Because of many serious posttherapy
118 Part I Principles of Urologic Oncology

complications, including urethro-cutaneous and urethro- Complications of the treatment included incontinence,
rectal fistulas, cryosurgery was not commonly used until urethral sloughing, rectal fistula, and perirectal abscess.34–36
its revival with US guidance in 1993.33 Patients self-reporting erectile dysfunction (ED) follow-
ing cryosurgery were many compared to other minimally
Procedure Cryotherapy is usually performed with the invasive prostate cancer treatments, ranging from 53% to
patient placed in the lithotomy position and placed under 87%.34–37 A recent study showed pilot results on a new
general anesthesia. The specific technique and the num- “nerve sparing” cryosurgery with the preservation of
ber of freezing cycles vary slightly between centers, with potency in 7 of 9 treated men at a median follow-up of 36
2 cycles used most commonly. After positioning of the months (range from 6 to 72 months).38
TRUS probe, multiple suprapubic cryoprobes are placed
using US guidance. To prevent damage to the urethra, a
Brachytherapy
warming urethral catheter is placed. Thermal sensors are
placed around the periphery of the gland to allow good Interstitial brachytherapy refers to the permanent
temperature control in critical locations. At the end of placement of small radioactive sources directly into the
the procedure, the cryoprobes are thawed and removed. prostate. These are typically iodine (I-125) or palla-
A newer approach to the use of cryotherapy in image- dium sources contained within a titanium-jacket and
guided interventions is MR-guided cryotherapy, which measure about 4 mm in length. Similar to cryosurgery,
has the major advantage of allowing clear visualization of interstitial brachytherapy can be used as a primary
the “ice-ball” induced in the tissue, as it occurs. This treatment or as a salvage therapy after external beam
allows for direct thermal monitoring of the treatment radiation or initial implant failure.39–41 Interstitial
effect (Figure 7-4). brachytherapy for prostate cancer was introduced in the
1960s by Scardino and Carlton.42 The placement of
Outcomes Because of the fairly recent revival of radioactive seeds was performed using a freehand tech-
cryotherapy due to image guidance improvements, the nique that did not provide homogenous seed distribu-
long-term treatment results are still being investigated. tion and resulted in both underdosing of tumors and
Several groups reported their preliminary results follow- overdosing of vital structures (rectum, urethra, NVBs).
ing TRUS-guided cryotherapy. At 5 years, the progres- This resulted in many posttreatment complications,
sion-free rate defined as undetectable PSA (< 0.3 ng/ml) and the procedure was discontinued until its revival
ranged from 48% to 77%, depending on patients’ risk with US image guidance in 1983 by Holm and col-
factors.34,35 leagues.43 Further improvements in imaging techniques
and technology resulted in the first MR-guided implant
being performed at 1997 at Brigham and Women’s
Hospital in Boston.44

Ultrasound-Guided Brachytherapy
Procedure A patient is placed in the lithotomy position,
a Foley catheter is inserted, and general or spinal anes-
thesia is administered. The TRUS probe and probe stabi-
lizer are positioned and the probe stepper is attached to
the stabilizer. US images are obtained every 5 mm from
the apex to the base. Some centers use designated treat-
ment planning software for preplanning of the proce-
dure.45 The images are transferred to a laptop computer
connected to the US equipment. A 3D reconstruction of
the prostate, urethra, and rectum is produced, and the
dose of radiation to those structures is visualized. Dose-
volume histograms and the number of radioactive seeds
per needle are then calculated. The template for needle
Figure 7-4 Axial MR image showing a percutaneous guidance is placed against the patient’s perineum. After
cryotherapy probe in the lateral aspect of the left kidney insertion of each needle a sagittal mode of the US acqui-
during an MR-guided cryoablation of a small renal cell sition is also used to determine the depth of the needle
carcinoma. The “black” ice-ball is clearly seen (solid arrow); insertion. Some centers use fluoroscopy to visualize seed
note the close proximity of the left colon (hollow arrow). placement.45
Chapter 7 Image-Guided Minimally Invasive Therapy 119

Outcomes Although investigators used slightly differ- MR-Guided Brachytherapy


ent definitions of biochemical failure, the overall results Patient Selection The patient selection criteria for this
of similar studies are quite consistent. At present, there program in our institution are clinical stage T1cNXM0
are only a few studies presenting 10-years outcome data (according to AJCC), PSA less than 10 ng/ml, biopsy
for prostate USBT. Biochemical disease-free survival rates Gleason score not more than 3 + 4, low cancer volume,
after 10 years following treatment ranged from 70% to and endorectal MRI demonstrating organ-confined dis-
87%.46–48 At 5 years, relapse-free survival rates reached ease. Patients with prior transurethral resection of the
85% to 94% for the low-risk group, 77% to 82% for the prostate (TURP) are excluded. We do not exclude men
intermediate-risk group, and 62% to 65% for the high- with larger-volume prostates, as pubic arch interference
risk group.49–51 can be avoided in this approach. All patients undergo
Reported complications following USBT included endorectal coil MRI for prostate cancer staging prior to
urinary incontinence, urethral strictures, cystitis, uri- the treatment visit (Figures 7-5 and 7-6). An MR radiolo-
nary retention, prostatitis, proctitis, rectal ulceration, gist assesses prostate gland volume, tumor location and
and rectal fistulas. Transient irritation and obstruction volume, the presence or absence of extraglandular disease,
of the urinary tract 2 to 6 months after treatment were seminal vesicle invasion (SVI), and possible spread to
common and about 10% of patients showed symptoms pelvic lymph nodes or bones.
of acute urinary retention (AUR).52 Preservation of
potency ranged from 64% to 79% at 3 years to 39% at Procedure This multidisciplinary procedure uses
6 years. Pretreatment ED and a higher implant dose many different computer, imaging and technical skills
caused greater impotence.53–55 and therefore requires the cooperation of specialists from
various medical and nonmedical fields, including radia-
tion oncologists, medical physicists, radiologists, anes-
CT-Guided Brachytherapy thesiologists, urologists, nurses, radiology technologists,
Procedure The prostate gland immobilization before and computer scientists.
the procedure is similar to US-guided therapy. A Foley For the procedure the patient is placed in an open
catheter, with radio-opaque wire to fluoroscopically configuration 0.5 T Signa SP MR scanner in the litho-
localize the urethra, is placed, and general anesthesia is tomy position. The patient is positioned on the table
administered. Preoperative CT images collected using between two magnets with vertically oriented open space
5 mm slices are used to outline the prostate gland for for easier access to the patient during the treatment (see
treatment planning. Posteroanterior and lateral fluoro- Figure 7-1). A Foley catheter is inserted, skin prepared,
scopic images are used to determine needle position the template for needle guidance placed against the
before seeds are deposited. The seeds are placed under patient’s perineum and secured, and a rectal obturator is
fluoroscopic control. inserted (Figure 7-7). T2-weighted MRI images are
Recently, CT-guided transischiorectal stereotactic acquired in the axial, coronal, and sagittal planes. The
brachytherapy has been introduced and tested.56–58 This radiologist uses the T2-weighted images (see Figure 7-2,
approach can be used in patients with larger prostates. right) to identify the peripheral zone (PZ), urethra, and
Transischiorectal CT images acquired every 5 mm are anterior rectal wall on each axial MR slice. These are
used for pretreatment planning. The patient is placed in then outlined using the 3D Slicer surgical simulation
the prone position, a Foley catheter is inserted and software designed and operated by members of the
spinal or epidural anesthesia is administered. A 3D Surgical Planning Laboratory (SPL) at Brigham and
stereotactic template used to guide needle placement is Women’s Hospital in Boston (Figure 7-8). The 3D Slicer
attached to the CT table and tilted at the same angle as is free, open-source software for two- and three-dimen-
the gantry. Electronic grids are superimposed on every sional display, registration, and segmentation of medical
second CT image to determine needle depth. CT images (see www.slicer.org for more information on 3D
images are used for needle visualization, placement cor- Slicer). Pretreatment planning, as well as calculation of
rections are introduced if necessary, and radioactive the MRI-based peripheral zone as a clinical target vol-
seeds are deposited. ume (CTV), is then performed by the medical physicists
using designated planning software.11 The number of
Outcomes Biochemical disease-free survival rates I-125 seeds per catheter and the depth of catheter inser-
reached 99% for low, 96% for intermediate, and 90% tion are calculated. The physicians then insert each pre-
for the high-risk group at a median follow-up of loaded catheter into the prostate gland. After every
4.5 years (2 to 8 years).59 Treatment complications catheter insertion, axial gradient-echo MR images are
included urinary retention, incontinence, and rectal obtained in real-time and compared to the catheter’s
symptoms. expected location according to the plan. Dose volume
120 Part I Principles of Urologic Oncology

Figure 7-5 Endorectal coil MRI of prostate. Axial (left) and coronal (right) T2-weighted
images provide superior visualization of the prostate and its zonal anatomy. White solid
arrows indicate PZ, hollow arrows indicate central gland, and striped arrows indicate
endorectal coil.

Figure 7-6 Prostate cancer. Axial T2-weighted erMRI image Figure 7-7 Close up view of a patient in the open 0.5T Signa
shows low signal lesion located in PZ (white arrow). SP magnet, during MR-guided prostate brachytherapy. The
patient is in the lithotomy position and the perineal template
used for catheter guidance is seen in the center.

histograms (DVH) for the CTV, anterior rectal wall, and Outcomes Long-term biochemical outcomes were
urethra are recalculated, adjustment of the catheter place- compared for similar patients over similar time frames
ment is performed when necessary, and seeds are between MR-guided brachytherapy and radical prostate-
deposited. ctomy by D’Amico et al.60 At 5 years, PSA control was
Approximately 6 weeks after the procedure, MRI and 95% for brachytherapy and 93% for RP patients (median
CT imaging of the prostate is performed to identify the follow-ups were 3.95 and 4.2 years for brachytherapy and
location of radioactive seeds and calculate final DVHs. RP patients, respectively). The percentage of positive
Since seeds can be well visualized on CT images, and the prostate biopsies was found to be a significant predictor
underlying anatomy is better depicted on MR images, of the time to postbrachytherapy PSA failure.
MR-CT fused images are used to calculate dose distribu- Short-term toxicity following MR-guided brachyther-
tion to the surrounding tissues (Figure 7-9). apy was rare, and no patient reported gastrointestinal or
Chapter 7 Image-Guided Minimally Invasive Therapy 121

Figure 7-8 Image segmentation using 3D Slicer surgical navigation software. PZ (solid
arrow), rectal wall (hollow arrow), and urethra (striped arrow) are identified on T2-weighted
image acquired in a 0.5 T scanner for MRBT planning.

Figure 7-9 MR-CT fusion of post-MRBT images. Post-therapy MR image (left) and CT image
(middle) are fused resulting in MR-CT image (right) to allow better visualization of individual
seeds and facilitate dose distribution calculation. Black arrows indicate radioactive seeds.
122 Part I Principles of Urologic Oncology

sexual dysfunction during the first month after treat- treatment is planned. Therapy is performed using a 2.25
ment.61 Acute urinary retention (AUR) was observed in to 3-MHz transducer.
12% of men within 24 hours of removal of the Foley
catheter and was self-limiting within 1 to 3 weeks. MR- Outcomes Early results showed 56% to 100% therapy
determined prostate volume, transitional zone (TZ) vol- response rates when using Ablatherm probes; however,
ume, and total number of seeds were found to be the criteria of PSA failure after FUS are still under
significant predictors of AUR on univariate analysis. The debate.66–72 Blana et al.73 reported outcomes following
TZ volume was the only significant predictor of AUR on FUS for localized prostate cancer at a median follow-up
multivariate logistic regression analysis. The authors of 22.5 months (4 to 62 months). After a follow-up for 22
concluded that benign prostatic hyperplasia (BPH) that months, 87% of patients had a PSA level below 1ng/ml,
results in larger TZ volume is the most important pre- and 93.4% had negative control biopsies. Reported treat-
dictor of AUR. No urinary incontinence was seen at a ment adverse effects included urinary tract infection,
median follow-up of 14 months (from 9 months to 2 stress incontinence, rectal burn, rectourethral fistulas,
years).62 urethral stricture, and impotence.
MR-guided brachytherapy is a very new approach; MR-guided FUS for treatment of prostate disease is
thus, there is only one report to date summarizing currently under investigation; initial animal tests appear
long-term toxicity.63 Albert et al.63 found low inci- very promising. FUS treatment can be monitored by
dence of rectal bleeding (8%) and no urethral stric- thermal maps and contrast-enhanced MRI (Figure 7-10).
tures at a median follow-up of 2.8 years (0.5 to 5
years). While ED reached 82%, two-thirds of the
Diagnosis
patients reported good erectile function after sildenafil
US-Guided Prostate Biopsy
(Viagra). No radiation cystitis was estimated at 4 years
after MRBT. Currently, diagnosis of prostate cancer is aided using
Quality of life (QoL) outcomes collected using a pre- TRUS to guide the biopsy and is a widely used and
viously validated questionnaire64 are currently being accepted procedure.74–76 However, the sensitivity and
assessed, and early reports indicate that MR-guided positive predictive value of sextant biopsy remain quite
prostate brachytherapy has better symptomatic outcomes low, 60% and 25%, respectively.77–79 The first transper-
than the conventional US-guided approach (J. Talcott, ineal prostate biopsies under US control were performed
personal communication). in the mid-1980s, and a few years later TRUS become
Current research projects will continue to study the the primary modality for biopsy guidance.80 Initially, the
radiation dose distribution to vital organs and its sextant biopsy technique recommended the collection of
impact on the side effects. Image-segmentation tech- six samples from the base, mid-gland, and apex on both
niques are used to identify those important organs on sides. Subsequent literature, however, showed the advan-
endorectal coil MR images. Radiation dose to the tages of increasing the number of samples to 10, 11, or
organs can then be correlated with changes in patient- even 12 to detect cancer with up to a 96% success rate. It
reported QoL. was also recommended that the number of core biopsies
increase with the prostate volume, since bigger gland
sizes introduced high sampling error and therefore
Focused Ultrasound Surgery
required more sampling.81–85 However, the ideal number
US-Guided High-Intensity Focused Ultrasound
of cores is still not clear.
for Prostate Cancer
This approach uses a high-intensity US beam that is Procedure Prior to the insertion of the endorectal
focused on the target lesion, which then undergoes ther- probe, the patient undergoes a digital rectal exam
mal coagulation. In 1996, Galet et al.65 were the first to (DRE). The patient is positioned on the table in either
evaluate clinical application of FUS for treatment of litothomy or lateral decubitus position. The ultrasound
organ-confined prostate cancer. probe is inserted and stabilized and the prostate volume
is calculated using transverse and sagittal imaging. If the
Procedure For the treatment the patient is placed in procedure is performed in lithothomy position, the tem-
the lateral position and anesthetized. A suprapubic plate for needle guidance is placed against the patient’s
catheter is placed to assure urinary drainage, and an perineum. The positions of needles are identified by grid
imaging and treatment probe is inserted into the rectum. coordinates on the template and the depth by the probe
The probe is surrounded with a balloon filled with cool- stepper attached to the probe stabilizer. These coordi-
ing fluid to avoid overheating of the rectal wall. Target nates are used to guide biopsy under real-time US imag-
areas are identified using biplanar US imaging and the ing. Biopsy is performed using an 18-gauge biopsy gun.
Chapter 7 Image-Guided Minimally Invasive Therapy 123

MR-Guided Prostate Biopsy


In addition to being an excellent method for guiding
prostate cancer therapy, MR imaging also appears to
be useful for guiding diagnostic biopsy.86 Similar to its
use in therapy, metabolic information from spec-
troscopy and dynamic contrast MR data can be com-
bined with routine MR images to allow precise tumor
targeting.
Our group has adapted the interventional MR system
to perform MR-guided prostate biopsy.86,87 This
transperineal technique eschews endorectal devices and
provides an excellent diagnostic alternative for patients
who have undergone rectal surgeries and in whom
US-guided procedure is impossible to perform. An addi-
tional group of men who can be benefited from
MR-guided procedure are those with persistently rising
PSA values and have had prior negative US-guided
biopsies. Preliminary feasibility results of this method
for facilitating prostate cancer diagnosis are promising.73
One of the unique aspects of this approach is the inter-
active imaging provided by using the 3D Slicer, as
reported by Hata et al.86, which facilitates T2 imaging in
“near-real time.” D’Amico et al.88 reported results of the
procedure from two MRI-targeted lesions in a patient
who could not undergo US-guided procedure because of
previous rectal surgery. Several transurethral biopsies
yielded negative results in this patient. Following MR-
guided biopsy, cancer was confirmed in 15% and 25% of
the 2 cores.

Figure 7-10 MR-focused US surgery of uterine fibroid. A, Procedure Prior to the procedure, each patient under-
Coronal T2-weighted FSE image (4000/90) used for goes endorectal coil MRI using a 1.5 T imaging system.
treatment planning. The sonication locations and sizes The T1- and T2-weighted and contrast-enhanced images
(circles and grid) were determined by the planning software are collected, and multivoxel spectroscopy is performed.
from this prescription (and the tissue depth) and displayed on
Using this information, the radiologist identifies biopsy
top of the treatment plan. During the treatment, the
targets. Patient positioning for the procedure and initial
accumulated thermal dose (hollow arrow) was displayed on
top of the treatment planning images. A dose threshold of preparation is similar to MRBT except that an endorectal
240 equivalent minimum at 43 ˚ C is displayed. B, Sagittal T2- obturator may not be used in some cases with previous rec-
weighted image (2500/98) showing the treatment plan and tal surgery. Subsequently, T2-weighted images are col-
the area that achieved the threshold thermal dose. C–D, lected at 3.5 mm intervals in a 0.5 T interventional system.
Temperature sensitive phase-difference FSPGR images The information from preprocedure and intraprocedure
(39.9/19.7) acquired at peak temperature rise during two images are correlated and target lesions are identified.
sonications, one imaged perpendicular to the direction of the Computer software is used to calculate appropriate coordi-
US beam (Coronal, C), and one imaged parallel to the nates on the perineal template for the needle insertion, as
direction of the beam (sagittal, D). These images were used well as needle insertion depth. Additionally, 0.5 T T2-
to estimate the thermal dose (white line) for each sonication.
weighted images and intraprocedure fast gradient-echo
E–F, Result of the treatment. E, Sagittal contrast-enhanced
images are loaded into the 3D Slicer software and displayed
gradient-echo image (245/1.8) acquired 2 days after US
therapy. The nonenhancing area (white arrow) is clearly seen. in an alternating fashion to provide real-time image guid-
F, Gross pathologic cut specimen showing the central area ance during biopsy. All target locations along with sextant
of hemorrhagic necrosis. (From Tempany MC et al: Radiology biopsies of the PZ from the right and left apex, mid-gland,
2003; 226(3):902.) and base are sampled using MR-compatible 18 gauge
biopsy guns. Figure 7-11 shows an axial view of the needle
tip artifact in PZ after needle insertion and just before
124 Part I Principles of Urologic Oncology

B
Figure 7-11 Biopsy needle artifact. Axial (A) and coronal (B) view of prostate gland on pre-
(left) and intra-MR-guided biopsy images (middle and right). The black arrow indicates the
tip of the biopsy needle. (From D’Amico Av, Loeffter JS, Harris JR. Image guided diagnosis
and treatment of cancer. Totowa, NJ, Humana Press, 2003.)

biopsy. This procedure is currently done under anesthe-


sia as a day surgical procedure. It is well tolerated and
offers a second-line biopsy approach in selected patients.

SUMMARY
The areas covered in this chapter serve to illustrate the
significant advances that have occurred in image-guided
procedures and therapy for both diagnosis and treatment
of prostate cancer. These are only some of the many new
IGT applications available today. As the imaging tech-
niques continue to improve and as surgical approaches
become even less invasive or completely noninvasive (as
Figure 7-12 Biopsy needle antifact. Real-time 3D view of with FUS), the future looks very exciting for both urol-
prostate gland on intra-MR-guided biopsy images. ogy patients and their doctors.
Chapter 7 Image-Guided Minimally Invasive Therapy 125

REFERENCES 17. Huber PE, Jenne JW, Rastert R, et al: A new noninvasive
approach in breast cancer therapy using magnetic
1. Jolesz FA: 1996 RSNA Eugene P. Pendergrass New resonance imaging-guided focused ultrasound surgery.
Horizons Lecture. 1997. Cancer Res 2001; 61(23):8441–8447.
2. Silverman SG, Jolesz FA, Newman RW, et al: Design and 18. Tempany CM, Stewart EA, McDannold N, et al: MR
implementation of an interventional MR imaging suite. imaging-guided focused ultrasound surgery of uterine
Am J Roentgenol 1997; 168(6):1465–1471. leiomyomas: a feasibility study. Radiology 2003;
3. Chan DY, Solomon S, Kim FJ, Jarret TW: Image-guided 226(3):897–905.
therapy in urology. J Endourol 2001;15(February; 1): 19. Chinzei K, Hata N, Jolesz FA, Kikinis R: Surgical assist
105–110. robot for the active navigation in the intraoperative MRI:
4. Jolesz FA, Nabavi A, Kikinis R: Integration of hardware design issues, http://www.spl.harvard.edu:8000/
interventional MRI with computer-assisted surgery. pages/projects/robot-new/iros2000.pdf.
J Magn Reson Imaging 2001; 13(1):69–77. 20. MR Compatible Surgical Robot System,
5. Comet-Batlle J, Vilanova-Busquets JC, Saladie-Roig JM, http://www.aist.go.jp/MEL/soshiki/kiso/biomech/chin/M
Gelabert-Mas A, Barcelo-Vidal C. The value of MM/mrmani0-e.htm.
endorectal MRI in the early diagnosis of prostate cancer. 21. Pham DL, Xu C, Prince JL: Current methods in medical
Eur Urol 2003; 44(August; 2):201–207 (Discussion image segmentation. Annu Rev Biomed Eng 2000;
207-8). 2:315–337.
6. Hricak H, White S, Vigneron D, Kurhanewicz J, Kosco A, 22. Soler L, Delingette H, Malandain G, et al: Fully
Levin D, et al: Carcinoma of the prostate gland: MR automatic anatomical, pathological, and functional
imaging with pelvic phased-array coils versus integrated segmentation from CT scans for hepatic surgery. Comput
endorectal-pelvic phased-array coils. Radiology Aided Surg 2001; 6(3):131–142.
1994;193:703–709. 23. EPIDAURE Research Project, http://www-sop.inria.fr/
7. Barentsz JO, Engelbrecht M, Jager GJ, et al: Fast dynamic epidaure/research.php.
gadolinium-enhanced MR imaging of urinary bladder and 24. Bogers HA, Sedelaar JP, Beerlage HP, et al: Contrast-
prostate cancer. J Magn Reson Imaging 1999; enhanced three-dimensional power Doppler angiography
10(September; 3):295–304. of the human prostate: correlation with biopsy outcome.
8. Huisman HJ, Engelbrecht MR, Barentsz JO: Accurate Urology 1999; 54(July; 1):97–104.
estimation of pharmacokinetic contrast-enhanced dynamic 25. Wallner K, Roy J, Zelefsky M, Fuks Z, Harrison, L:
MRI parameters of the prostate. J Magn Reson Imaging Fluoroscopic visualization of the prostatic urethra to
2001; 3(April; 4):607–614. guide transperineal prostate implantation. Int J Radiat
9. Engelbrecht MR, Huisman HJ, Laheij RJ, et al: Oncol Biol Phys 1994; 29(4):863–867.
Discrimination of prostate cancer from normal peripheral 26. Lieberfarb ME, Schultz D, Whittington R, et al: Using
zone and central gland tissue by using dynamic contrast- PSA, biopsy Gleason score, clinical stage, and the
enhanced MR imaging. Radiology 2003; 229(October; 1): percentage of positive biopsies to identify optimal
248–254 (Epub 2003 August 27). candidates for prostate-only radiation therapy. Int J Radiat
10. Coakley FV, Qayyum A, Kurhanewicz J. Magnetic Oncol Biol Phys 2002; 53(4):898–903.
resonance imaging and spectroscopic imaging of prostate 27. Jani AB, Hellman S: Early prostate cancer: clinical
cancer. J Urol 2003; 170(December; 6 Pt 2):S69–S75 decision-making. Lancet 2003; 361(9362):1045–1053.
(Discussion S75-6). 28. Peschel RE, Colberg JW: Surgery, brachytherapy, and
11. Kooy HM, Cormack RA, Mathiowitz G, Tempany C, external-beam radiotherapy for early prostate cancer.
D’Amico AV: A software system for interventional Lancet Oncol 2003; 4(4):233–241.
magnetic resonance image-guided prostate brachytherapy. 29. Ellis DS: Cryosurgery as primary treatment for localized
Comput Aided Surg 2000; 5(6):401–413. prostate cancer: a community hospital experience.
12. Cline HE, Hynynen K, Hardy CJ, et al: MR temperature Urology 2002; 60(2 Suppl 1):34–39.
mapping of focused ultrasound surgery. Magn Reson Med 30. Ghafar MA, Johnson CW, De La Taille A, et al: Salvage
1994; 31(6):628–636. cryotherapy using an argon based system for locally
13. Jolesz FA, Hynynen K: Magnetic resonance image-guided recurrent prostate cancer after radiation therapy: the
focused ultrasound surgery. Cancer J 2002; 8(Suppl 1): Columbia experience. J Urol 2001; 166(4):1333–1337
S100–S112. (Discussion 1337-8).
14. Hynynen K, Darkazanli A, Damianou CA, Unger E, 31. da la Taille A, Katz AE: Cryosurgery: Is it an effective
Schenck JF: Tissue thermometry during ultrasound option for patients failing radiation? Curr Opin Urol
exposure. Eur Urol 1993; 23(Suppl 1):12–16. 2000; 10(5):409–413.
15. Fry WJ, Barnard JW, Fry FJ, Krunins RF, Brennan JF: 32. Gonder MJ, Soanes WA, Shulman S: Cryosurgical
Ultrasonic lesions in mammalian central nervous system, treatment of the prostate. Invest Urol 1966;
Science 1955; 122:517. 3(4):372–378.
16. Hynynen K, Pomeroy O, Smith DN, et al: MR imaging- 33. Onik GM, Cohen JK, Reyes GD: Transrectal ultrasound-
guided focused ultrasound surgery of fibroadenomas in the guided percutaneous radical cryosurgical ablation of the
breast: a feasibility study, Radiology 2001; 219(1):176–185. prostate. Cancer 1993; 72(4):1291–1299.
126 Part I Principles of Urologic Oncology

34. Donnelly BJ, Saliken JC, Ernst DS, et al: Prospective trial 49. Blasko JC, Grimm PD, Sylvester JE, et al: Palladium-103
of cryosurgical ablation of the prostate: five-year results. brachytherapy for prostate carcinoma. Int J Radiat Oncol
Urology 2002; 60(4):645–649. Biol Phys 2000; 46(4):839–850.
35. Long JP, Bahn D, Lee F, et al: Five-year retrospective, 50. Potters L, Cha C, Oshinsky G, et al: Risk profiles to
multi-institutional pooled analysis of cancer-related predict PSA relapse-free survival for patients undergoing
outcomes after cryosurgical ablation of the prostate. permanent prostate brachytherapy. Cancer J Sci Am 1999;
Urology 2001; 57(3):518–523. 5(5):301–306.
36. Badalament RA, Bahn DK, Kim H, et al: Patient-reported 51. Beyer DC, Brachman DG: Failure free survival following
complications after cryoablation therapy for prostate brachytherapy alone for prostate cancer: comparison with
cancer. Arch Ital Urol Androl 2000; 72(4):305–312. external beam radiotherapy. Radiother Oncol 2000;
37. Robinson JW, Moritz S, Fung T: Meta-analysis of rates of 57(3):263–267.
erectile function after treatment of localized prostate 52. Merrick GS, Butler WM, Tollenaar BG, Galbreath RW,
carcinoma. Int J Radiat Oncol Biol Phys 2002; Lief JH: The dosimetry of prostate brachytherapy-
54(4):1063–1068. induced urethral strictures. Int J Radiat Oncol Biol Phys
38. Onik G, Narayan P, Vaughan D, Dineen M, Brunelle R: 2002; 52(2):461–468.
Focal “nerve-sparing” cryosurgery for treatment of 53. Merrick GS, Butler WM, Galbreath RW, et al: Erectile
primary prostate cancer: a new approach to preserving function after permanent prostate brachytherapy. Int J
potency. Urology 2002; 60(1):109–114. Radiat Oncol Biol Phys 2002; 52(4):893–902.
39. Bice WS Jr, Freeman JE, Russell LF Jr, et al: Use of 54. Stock RG, Kao J, Stone NN: Penile erectile function after
image coregistration in salvage prostate brachytherapy. permanent radioactive seed implantation for treatment of
Tech Urol 2000; 6(2):151–156. prostate cancer. J Urol 2001; 165(2):436–439.
40. Beyer DC: Permanent brachytherapy as salvage treatment 55. Talcott JA, Clark JA, Stark PC, Mitchell SP: Long-term
for recurrent prostate cancer. Urology 1999; treatment related complications of brachytherapy for early
54(5):880–883. prostate cancer: a survey of patients previously treated.
41. D’Amico AV: Analysis of the clinical utility of the use of J Urol 2001; 166(2):494–499.
salvage brachytherapy in patients who have a rising PSA 56. Koutrouvelis P, Lailas N, Katz S, et al: High- and low-risk
after definitive external beam radiation therapy. Urology prostate cancer treated with 3D CT-guided
1999; 54(2):201–203. brachytherapy: 1- to 5-year follow-up. J Endourol 2000;
42. Scardino PT, Carlton CE: Combined interstitial and 14(4):357–366.
external irradiation for prostatic cancer. In Javadpour 57. Koutrouvelis PG, Three-dimensional stereotactic
N (ed): Principles and Management of Urologic Cancer, posterior ischiorectal space computerized tomography
pp 392–408. Baltimore, Williams and Williams, 1983. guided brachytherapy of prostate cancer: a preliminary
43. Holm HH, Juul N, Pedersen JF, Hansen H, Stroyer I: report. J Urol 1998; 159(1):142–145.
Transperineal 125 iodine seed implantation in prostatic 58. Molloy JA, Williams MB: Treatment planning
cancer guided by transrectal ultrasonography. J Urol 1983; considerations and quality assurance for CT-guided
130(2):283–286. transischiorectal implantation of the prostate. Med Phys
44. D’Amico AV, Cormack R, Tempany CM, et al: Real-time 1999; 26(9):1943–1951.
magnetic resonance image-guided interstitial 59. Koutrouvelis PG, Lailas N, Katz S, et al: Prostate cancer
brachytherapy in the treatment of select patients with with large glands treated with three-dimensional
clinically localized prostate cancer. Int J Radiat Oncol Biol computerized tomography guided pararectal brachytherapy:
Phys 1998; 42(3):507–515. up to 8 years of followup. J Urol 2003; 169(4):1331–1336.
45. Kaplan ID, Holupka EJ, Meskell P, et al: Intraoperative 60. D’Amico AV, Tempany CM, Schultz D, et al: Comparing
treatment planning for radioactive seed implant therapy PSA outcome after radical prostatectomy or magnetic
for prostate cancer. Urology 2000; 56(3):492–495. resonance imaging-guided partial prostatic irradiation in
46. Grimm PD, Blasko JC, Sylvester JE, Meier RM, select patients with clinically localized adenocarcinoma of
Cavanagh W: Ten-year biochemical (prostate-specific the prostate. Urology 2003;62(6):1063–1067.
antigen) control of prostate cancer with (125)I 61. D’Amico A, Cormack R, Kumar S, Tempany CM: Real-
brachytherapy. Int J Radiat Oncol Biol Phys 2001; time magnetic resonance imaging-guided brachytherapy
51(1):31–40. in the treatment of selected patients with clinically
47. Ragde H, Elgamal AA, Snow PB, et al: Ten-year disease localized prostate cancer. J Endourol 2000; 14(4):367–370.
free survival after transperineal sonography-guided 62. Thomas MD, Cormack R, Tempany CM, et al: Identifying
iodine-125 brachytherapy with or without 45-gray the predictors of acute urinary retention following
external beam irradiation in the treatment of patients with magnetic-resonance-guided prostate brachytherapy. Int
clinically localized, low to high Gleason grade prostate J Radiat Oncol Biol Phys 2000; 47(4):905–908.
carcinoma. Cancer 1998; 83(5):989–1001. 63. Albert M, Tempany CM, Schultz D, et al: Late
48. Blasko JC, Grimm PD, Sylsvester JE, Cavanagh W: The genitourinary and gastrointestinal toxicity after magnetic
role of external beam radiotherapy with I-125/Pd-103 resonance image-guided prostate brachytherapy with or
brachytherapy for prostate carcinoma. Radiother Oncol without neoadjuvant external beam radiation therapy.
2000; 57(3):273–278. Cancer 2003; 98(5):949–954.
Chapter 7 Image-Guided Minimally Invasive Therapy 127

64. Clark JA, Talcott JA: Symptom indexes to assess outcomes 77. Keetch DW, McMurtry JM, Smith DS, Andriole GL,
of treatment for early prostate cancer. Med Care 2001; Catalona WJ: Prostate specific antigen density versus
39(10):1118–1130. prostate specific antigen slope as predictors of prostate
65. Gelet A, Chapelon JY, Margonari J, et al: Prostatic tissue cancer in men with initially negative prostatic biopsies.
destruction by high-intensity focused ultrasound: J Urol 1996; 156(2 Pt 1):428–431.
experimentation on canine prostate. J Endourol 1993; 78. Terris MK: Sensitivity and specificity of sextant biopsies in
7(3):249–253. the detection of prostate cancer: preliminary report.
66. Gelet A, Chapelon JY, Bouvier R, et al: Treatment of Urology 1999; 54(3):486–489.
prostate cancer with transrectal focused ultrasound: early 79. Terris MK, McNeal JE, Freiha FS, Stamey TA: Efficacy
clinical experience. Eur Urol 1996; 29(2):174–183. of transrectal ultrasound-guided seminal vesicle biopsies
67. Gelet A, Chapelon JY, Bouvier R, et al: Transrectal high in the detection of seminal vesicle invasion by prostate
intensity focused ultrasound for the treatment of localized cancer. J Urol 1993; 149(5):1035–1039.
prostate cancer: factors influencing the outcome. Eur 80. Applewhite JC, Matlaga BR, McCullough DL, Hall MC.
Urol 2001; 40(2):124–129. Transrectal ultrasound and biopsy in the early diagnosis of
68. Gelet A, Chapelon JY, Bouvier R, et al: Transrectal high- prostate cancer. Cancer Control 2001; 8(March–April; 2):
intensity focused ultrasound: minimally invasive therapy 141–150.
of localized prostate cancer. J Endourol 2000; 81. Eskew LA, Bare RL, McCullough DL. Systematic 5
14(6):519–528. region prostate biopsy is superior to sextant method for
69. Chaussy CG, Thuroff S, High-intensive focused diagnosing carcinoma of the prostate. J Urol 1997;
ultrasound in localized prostate cancer. J Endourol 2000; 157(January; 1):199–202 (Discussion 202-3).
14(3):293–299. 82. Chang JJ, Shinohara K, Bhargava V, Presti JC Jr:
70. Gelet A, Chapelon JY, Bouvier R, Pangaud C, Lasne Y: Prospective evaluation of lateral biopsies of the peripheral
Local control of prostate cancer by transrectal high zone for prostate cancer detection. J Urol 1998;
intensity focused ultrasound therapy: preliminary results. 60(December; 6 Pt 1):2111–2114.
J Urol 1999; 161(1):156–162. 83. Chen ME, Troncoso P, Johnston DA, Tang K, Babaian
71. Beerlage HP, Thuroff S, Debruyne FM, Chaussy C, de la RJ: Optimization of prostate biopsy strategy using
Rosette JJ: Transrectal high-intensity focused ultrasound computer based analysis. J Urol 1997; 58(December; 6):
using the Ablatherm device in the treatment of localized 2168–2175.
prostate carcinoma. Urology 1999; 54(2):273–277. 84. Babaian RJ, Toi A, Kamoi K, et al: A comparative analysis
72. Uchida T, Sanghvi NT, Gardner TA, et al: Transrectal of sextant and an extended 11-core multisite directed
high-intensity focused ultrasound for treatment of biopsy strategy. J Urol 2000; 163(January; 1):152–157.
patients with stage T1b-2n0m0 localized prostate cancer: 85. Naughton CK, Miller DC, Mager DE, Ornstein DK,
a preliminary report. Urology 2002; 59(3):394–398 Catalona WJ. A prospective randomized trial comparing 6
(Discussion 398-9). versus 12 prostate biopsy cores: impact on cancer
73. Blana A, Walter B, Rogenhofer S, Wieland WF. High- detection. J Urol 2000; 164(August; 2):388–392.
intensity focused ultrasound for the treatment of localized 86. Hata N, Jinzaki M, Kacher D, et al: MR imaging-guided
prostate cancer: 5-year experience. Urology prostate biopsy with surgical navigation software: device
2004;63(2):297–300. validation and feasibility. Radiology 2001; 220(1):263–268.
74. Lee F, Gray JM, McLeary RD, et al: Prostatic evaluation 87. Cormack RA, D’Amico AV, Hata N, et al: Feasibility of
by transrectal sonography: criteria for diagnosis of early transperineal prostate biopsy under interventional MR
carcinoma. Radiology 1986; 158(1):91–95. guidance. Urology 2000; 56(4):663–664.
75. Lee F, Gray JM, McLeary RD, et al: Transrectal 88. D’Amico AV, Tempany CM, Cormack R, et al:
ultrasound in the diagnosis of prostate cancer: location, Transperineal magnetic resonance image guided prostate
echogenicity, histopathology, and staging. Prostate 1985; biopsy. J Urol 2000; 164(2):385–387.
7(2):117–129.
76. Rifkin MD, Kurtz AB, Goldberg BB: Prostate biopsy
utilizing transrectal ultrasound guidance: diagnosis of
nonpalpable cancers. J Ultrasound Med 1983;
2(4):165–167.
C H A P T E R

8 Adrenal Tumors
E. Darracott Vaughan, Jr, MD

The major adrenal tumors that will be discussed in this nance imaging (MRI) gives information concerning cell
chapter include adrenal cortical adenomas producing pri- type and aids in the differentiation of adenomas from
mary hyperaldosteronism and Cushing’s syndrome, adre- medullary tumors or metastatic carcinoma.4 Other
nal cortical carcinoma, the incidentally identified adrenal advantages of MRI scanning will be discussed later. The
mass, and pheochromocytoma. Actually, the most com- right adrenal lies above the kidney posterior and lateral
mon tumors involved in the adrenal gland are metastatic to the inferior vena cava (IVC) and its solitary venous
tumors to the adrenal, and the management of such drainage is via a short sturdy vein that enters the IVC in
lesions generally is dependent on the treatment of the a posterior fashion. Hence, the right adrenal gland is best
primary disease entity. It is fortunate that the diagnosis of approached through a posterior or modified posterior
adrenal disorders is extremely accurate using the combi- incision.5 The left adrenal is in more intimate contact with
nation of precise analytical methods for the measurement the kidney, overlying the upper pole of the kidney with its
of the abnormal secretion of adrenal hormones and anterior and medial surfaces behind the pancreas and
sophisticated radiographic techniques for the localization splenic artery. It is best exposed through a flank approach
and characterization of specific adrenal lesions.1,2 or a thoracoabdominal approach if the lesion is large.
The management of patients with adrenal tumors The adrenals have a delicate, rich blood supply esti-
requires a clear understanding of the normal physiology mated to be 6 to 7 ml/g/min without a dominant adrenal
of the adrenal medulla and cortex; a three-dimensional artery. The inferior phrenic artery is the main blood sup-
concept of the adrenal anatomy, as well as adjacent struc- ply with additional branches from the aorta and renal
tures; and the knowledge of the various pathologic enti- arteries. The small arteries penetrate the gland in a cir-
ties that may involve the adrenal. Moreover, the cumferential stellate fashion leaving both the anterior
operating surgeon must be well aware of the nuances and posterior surfaces avascular (Figure 8-1). During
involved in the diagnosis of the different adrenal entities, adrenalectomy, an important technical goal is to divide
be aware of potential intraoperative phenomena that are the superior and lateral blood supplies to the adrenal
unique to these patients, and be alert to specific postop- first, allowing the adrenal to remain attached to the kid-
erative complications that may occur.3 This chapter will ney, which can be used to draw the adrenal gland inferi-
review the preoperative, intraoperative, and postopera- orly and anteriorly during the resection. On the left side,
tive aspects of each of these specific entities and will out- the adrenal vein drains into the left renal vein; however,
line surgical approaches with operative hints to guide there is also a medially located phrenic drainage branch,
those interested in adrenal surgery. which, if not appropriately ligated, can cause trouble-
The adrenal glands are paired retroperitoneal organs some bleeding (Figure 8-2). The left adrenal vein is also
that lie within the perinephric fat, at the anterior, supe- a guide to the left renal artery, which often lies dorsal to
rior, and medial aspects of the kidneys. Their location in the vein. One potential complication of left adrenalec-
juxtaposition with other organs, as well as the periadrenal tomy is the inadvertent ligation of the apical renal arte-
fat, renders them ideal for sectional imaging by com- rial branch to the upper pole, which lies in close contact
puted tomography (CT). Thin-cut CT scanning allows to the inferior border of an adrenal tumor.
precise identification of lesions as small as 0.5 cm. The The basic physiology of the adrenal cortex and
CT scan remains the best imaging device for the identi- medulla, as well as the various pathologic entities, will be
fication of small adrenal lesions, whereas magnetic reso- discussed under specific disorders.

131
132 Part II Adrenal Gland

Figure 8-2 Venous drainage of left and right adrenal glands


with particular attention to the intercommunicating vein on
Figure 8-1 Arterial supply of left and right adrenal glands. the left.

CUSHING’S SYNDROME disorders mimic the clinical and even the biochemical
Cushing’s syndrome is the term utilized to describe the manifestations of Cushing’s syndrome. These patients
symptom complex caused by excessive circulating gluco- have been termed to have “pseudo”-Cushing’s syndrome;
corticoids. We must remember that the term is all- this may exist in patients with major depression or in
encompassing and includes: patients with pituitary patients with chronic alcoholism.6
hypersecretion of adrenocorticotrophic hormone There are a myriad of tests both to diagnose the pres-
(ACTH) (corticotropin); Cushing’s disease, which ence of Cushing’s syndrome and then to identify which
accounts for 75% to 80% of patients with endogenous subentity is present. Fortunately, due to recent develop-
Cushing’s; adrenal adenomas or carcinomas; ectopic ment of extremely accurate assays for urinary and plasma
secretion of ACTH, or corticotropin-releasing hormone cortisol, as well as plasma corticotropin, this task has
(CRH) syndrome.6 Before assuming that a patient has become much easier. The approach that has recently
one of these pathologic entities, there should be a thor- been reported by Orth6 is shown in Figure 8-3.
ough questioning of the patient about the use of steroid- The clinical diagnosis of Cushing’s syndrome is con-
containing preparations. At times patients are unaware firmed by the demonstration of cortisol hypersecretion.
that a substance they use, particularly creams or lotions, At the present time the determination of 24-hour urinary
contains steroids, and if the patient is on any type of excretion of cortisol in the urine is the most direct and
medication at all, it should be carefully reviewed for reliable index of cortical secretion. Orth recommends
steroid content. There are few diseases in which the clin- that urinary cortisol should be measured in two and
ical appearance of the patient can be as useful in suspect- preferably three consecutive 24-hour urine specimens,
ing the diagnosis. Old photographs are helpful in collected on an outpatient basis.
documenting recent changes in appearance that Once the diagnosis has been established, the next
occurred. The more common clinical manifestations of chore is to determine whether there is Cushing’s disease
Cushing’s syndrome found in different series of patients due to hypersecretion of plasma corticotropin (ACTH)
are shown in Table 8-1. The clinical findings do not dis- from the pituitary or primary adrenal disease. Herein is
tinguish patients with Cushing’s disease from those with the major change in our approach to patients with
adrenal adenoma; however, patients with adrenal carci- Cushing’s disease. In the past, high- and low-dose dex-
noma are more likely to show virilization in the female or amethasone suppression tests have been used to accom-
feminization in the male. Patients with ectopic ACTH plish this task. At present, the low-dose dexamethasone is
may present with manifestations of the primary tumor. It generally used to rule out pseudo-Cushing’s syndrome.
is also important to remember that some nonendocrine The differentiation of corticotropin-dependent Cushing’s
Chapter 8 Adrenal Tumors 133

Table 8-1 Clinical Manifestations of Cushing’s entiate between pituitary and adrenal Cushing’s syn-
Syndrome drome. Patients are given high-dose dexamethasone
All* Disease† Adenoma/
(2 mg every 6 hours for 2 days), and plasma cortisol and
(%) (%) Carcinoma‡ (%) urinary free cortisol levels are measured. In patients with
pituitary disease, there should be a 50% or greater sup-
Obesity 90 91 93 pression in cortisol. Patients with adrenal adenomas or
carcinomas fail to suppress cortisol secretion. The high-
Hypertension 80 63 93 dose dexamethasone suppression test may also be useful
Diabetes 80 32 79
to identify ectopic ACTH syndrome, where there is usu-
ally complete resistance to high-dose dexamethasone
Centripetal obesity 80 — — suppression.
Treatment is obviously dependent on the underlying
Weakness 80 25 82 lesion. Patients with adrenal adenomas or carcinomas are
Muscle atrophy 70 34 —
generally treated with surgical extirpation of the lesions.
Patients with Cushing’s disease have confirmation with
Hirsutism 70 59 79 pituitary CT or MRI and usually are treated with transsphe-
noidal pituitary tumor removal, and patients with ectopic
Menstrual abnormal/ 70 46 75 ACTH have treatment directed towards the primary tumor.
sexual dysfunction The surgical approach and preparation of patients with
Purple striae 70 46 36 adrenal Cushing’s disease will be discussed later.
If the patient is identified as having adrenal Cushing’s,
Moon facies 60 — — the next step is radiographic localization with CT scan-
ning.7 Adrenal adenomas are usually larger than 2 cm,
Osteoporosis 50 29 54 solitary, and associated with atrophy of the opposite
Early bruising 50 54 57 gland. The density is low because of the high concentra-
tion of lipid (Figure 8-4). Adrenal carcinomas are often
Acne/pigmentation 50 32 — indistinguishable from adenomas except for the larger
size, carcinomas usually being greater than 6 cm.8
Mental changes 50 47 57 Necrosis and calcification are also more common in asso-
Edema 50 15 — ciation with adrenal carcinomas but are not specific.
Clearly, large irregular adrenal lesions with invasion rep-
Headache 40 21 46 resent carcinoma; however, metastatic carcinoma to the
adrenal has the same appearance.
Poor healing 40 — — MRI is not usually necessary in patients with
From Scott HW Jr: In Scott HW (ed): Surgery of the Adrenal Cushing’s syndrome unless the lesion is large; the ration-
Glands. Philadelphia, JB Lippincott Co, 1990, with permission. ale for MRI is to obtain anatomic information concern-
*Hunt and Tyrell, 1978. ing surrounding structures or invasion of the IVC, a rare
†Wilson, 1984.
‡Scott, 1973.
but well-recognized entity.9
Adrenal cortical scanning with iodinated cholesterol
agents is no longer routinely utilized but can be helpful
syndrome versus corticotropin-independent Cushing’s in differentiating functional adrenal tissue from other
syndrome is determined by the concurrent late afternoon retroperitoneal lesions.10
or midnight measurement of collection of blood for the
simultaneous measurement of plasma corticotropin and
INCIDENTALLY DISCOVERED ADRENAL MASSES
cortisol. Thus, if the patient’s cortisol concentration is
above 50 μg/dl and the corticotropin concentration The increased utilization of abdominal ultrasound and
is below 5 pg/ml, then the cortisol secretion is ACTH CT scanning has led to a new classification of adrenal
independent and the patient has a primary adrenal prob- lesions termed the “incidentally identified unsuspected
lem. In contrast, if the plasma corticotropin concentra- adrenal mass” or “incidentaloma.”8 Our approach to the
tion is greater than 50 pg/ml, then the cortisol secretion incidentally identified adrenal mass is shown in Figure 8-5.
is ACTH dependent and the patient has Cushing’s syn- Several points do not warrant controversy. First, there is
drome or ectopic ACTH or CRH syndrome.5 In situa- an agreement that all patients with solid adrenal masses
tions where the two-site immunoradiometric assay test is should undergo biochemical assessment. If biochemical
not available, the high-dose dexamethasone suppression abnormalities are identified, the lesions should be treated
test has always been used as the standard test to differ- as described elsewhere in the chapter, usually by removal
134 Part II Adrenal Gland

Figure 8-3 Identifying Cushing’s syndrome and its causes.

of the offending lesion. However, the extent of biochem- feel that CT scanning may underestimate the size of an
ical assessment has been reviewed, and a selective adrenal, and we suggest that exploration be performed
approach has been outlined that markedly limits cost when lesions are more than 5 cm on CT or MRI.12
without sacrificing diagnostic accuracy.11 A very limited Furthermore, if lesions are purely cystic by CT or MRI,
evaluation is recommended, including tests only to rule cyst puncture is often not necessary and these lesions can
out pheochromocytoma, potassium levels in hypertensive be followed (Figure 8-6). The controversy arises in the
cases, and glucocorticoid evaluation only in the presence management strategy for the solid adrenal lesions smaller
of clinical stigmata of Cushing’s syndrome or virilization. than 5 cm in size. The current approach has been to use
The second point that is not controversial is that non- MRI imaging in this situation. Most adenomas appear
functioning solid lesions larger than 5 cm should be slightly hypointense or isointense relative to the liver or
removed. This is based on the finding that adrenal malig- spleen on T1-weighted images and slightly hyperintense
nancies are almost always larger than 6 cm. However, we or isointense relative to hepatic or splenic parenchyma
Chapter 8 Adrenal Tumors 135

Figure 8-4 CT scan of a patient with right adrenal adenoma.

Figure 8-5 Evaluation of incidentally found adrenal mass.

Figure 8-6 Multilocular benign renal cyst in an asymptomatic patient that was incidentally
identified. A, CT scan showing left adrenal cyst. B, Coronal MRI showing the lobular
suprarenal adrenal cyst. In this case, exploration was carried out because of multilocular
nature. The cyst was benign.
136 Part II Adrenal Gland

on T2-weighted images. There is little change in the what contrived, since many of these tumors will produce
intensity from T1- to T2-weighted studies. In contrast, multiple adrenal hormones and also because of the clear
the general notion is that adrenal cortical carcinoma is evidence that a tumor may secrete one hormone at
hypointense relative to liver or spleen on T1-weighted one point in its natural history and additional hormones
images and hyperintense to the liver or spleen on T2- at a later phase when there is increased tumor mass.
weighted images. Thus, if the mean signal intensity ratio The most commonly identified functional tumor is
between the lesion and the spleen is over 0.8, it is one causing Cushing’s syndrome. The most common
unlikely that the lesion is a benign adenoma. However, it characteristic to delineate Cushing’s syndrome due to
should be remembered that there are a number of enti- carcinoma rather than adenoma has been the presence of
ties other than adrenal carcinoma that can cause high virilization with elevated 17-ketosteroid levels. More
intensity, including neural tumors, metastatic tumors to recently, the measurement of DHEA has been useful in
the adrenal, adrenal hemorrhage, and other retroperi- identifying these patients.
toneal lesions.4,13,14 An additional study that has shown Other rare functional tumors include both testos-
accuracy is the fine-needle adrenal biopsy guided by terone- and estrogen-secreting adrenal cortical tumors.
ultrasound or CT. In a large series from Finland, signifi- Rarely, virilization can occur in the absence of elevated
cant cytologic material was obtained in 96.4% and the urinary 17-ketosteroids and raises the possibility of pure
accuracy to differentiate benign from malignant disease testosterone-secreting ovarian or adrenal lesions.17 Of
was 85.7%.15 However, the utilization of aspiration the two sites of origin, adrenal cortical tumors secreting
cytology requires an extremely experienced cytologist, testosterone are exceedingly rare. In contrast to other
and in fact there is often inability to distinguish an adre- tumors described in this section, these tumors are usually
nal adenoma from a carcinoma even on pathologic review small, less than 6 cm, and many behave in a benign fash-
of the entire specimen. ion. In contrast, most feminizing tumors occur in males
It is our general approach that if there is either any 25 to 50 years of age, and they are usually larger, often
radiographic evidence that argues against a characteristic palpable, and highly malignant.18 Characteristically, the
benign adenoma or any change in size of an adrenal patients present with gynecomastia; in addition they may
lesion with repeated studies, then we feel that adrenalec- exhibit testicular atrophy, impotence, or decreased libido.
tomy is indicated. This fairly aggressive approach is jus- We have also seen a presentation with infertility and
tified in view of the extremely poor prognosis of patients oligospermia. These tumors secrete androstenedione,
when adrenal carcinoma is diagnosed, even when the which is converted peripherally to estrogen. Other
lesion is localized. steroids may also be secreted, and the clinical picture may
be mixed with associated cushingoid features.
The management of adrenal cortical carcinoma is sur-
ADRENAL CARCINOMA
gical removal of the primary tumor. The most common
Adrenal carcinoma is a rare disease with a poor progno- sites of metastasis include lung, liver, and lymph nodes.19
sis. The incidence is estimated as 1 case per 1.7 million, Often these tumors extend directly into adjacent struc-
accounting for only 0.02% of cancers. A practical sub- tures, especially the kidney, and surgical removal may
classification for adrenal carcinomas is according to their require removal of the primary tumor and adjacent
ability to produce adrenal hormones. In a series by Luton organs, including the kidney, spleen, as well as local
et al.,16 79% of adrenal tumors were functional, a higher lymph nodes. Unfortunately, despite en bloc resection
percentage than previously reported due to more sensi- even in patients without evidence of metastatic disease,
tive assays. The varieties of functioning tumors are the 5-year survival rate is only approximately 50% with
shown in Table 8-2. However, this classification is some- complete resection and 25% overall.20 Because of the
poor prognosis there has been an intense search for effec-
tive adjunctive chemotherapy, but this search has been
Table 8-2 Classification of Adrenal Carcinoma frustrating and it is generally believed that conventional
Functional chemotherapy is not effective, probably because of
Cushing’s syndrome P-glycoprotein expression.21 The most success has
Virilization in females been reported with the adrenolytic 1,1-dichloro-
Increased DHEA, 17-ketosteroids 2-(o-chlorophenyl)-2-(p-chlorophenyl)-ethane(o,p¢-DDD)
Increased testosterone or Mitotane. This DDT derivative has been shown to
Feminizing syndrome in males induce tumor response in 35% in a review of 551 cases
Hyperaldosteronism
reported in the literature.22 However, despite these
Mixed combination of above
response rates, survival time has not been prolonged and
Nonfunctional
there is intense toxicity. Recently, it has been suggested
DHEA, dehydroepiandrosterone. that patients even without the presence of metastatic
Chapter 8 Adrenal Tumors 137

disease be given adjunctive o,p¢-DDD, and trials are of primary hyperaldosteronism is now identified by the
currently in progress to determine if this approach is combined findings of hypokalemia, suppressed plasma
efficacious. renin activity (PRA) despite sodium restriction, and a
In general, there is an extremely poor prognosis in high urinary and plasma aldosterone level after sodium
patients with adrenal cortical carcinoma and an obvious repletion in hypertensive patients. The current evalua-
need for the development of new treatment strategies. tion of patients suspected of having hyperaldosteronism
is shown in Figure 8-7. The primary physiologic control
of aldosterone secretion is angiotensin II (Figure 8-8).
HYPERALDOSTERONISM
Other control mechanisms are ACTH and potassium. A
The term hyperaldosteronism originally was coined by Dr. clear knowledge of the physiology of the renin–
Jerome Conn to describe the clinical syndrome charac- angiotensin–aldosterone system (RAAS) is mandatory in
terized by hypertension, hypokalemia, hypernatremia, order to understand the pathophysiology and evaluate
alkalosis, and periodic paralysis due to an aldosterone- patients with primary hyperaldosteronism.25,26 The criti-
secreting adenoma.23 We now realize that this metabolic cal sensor in the RAAS resides in the juxtaglomerular
syndrome can be caused by either a solitary adrenal ade- apparatus within the kidney. Thus, in response to a vari-
noma or by bilateral adrenal zona glomerulosa hyperpla- ety of stimuli, but primarily decreased renal perfusion, or
sia. One of the clinical chores is to delineate patients with a decreased intake of sodium, there is an increased renin
hyperplasia from those with adenoma.24 The syndrome release, formation of angiotensin II, and subsequent

Hypertension

>3.6 Serum K ⱕ3.6

ⱕ1.0 PRA >1.0


Replete
K and Na

1⬚ aldosteronism 1⬚ aldosteronism
is unlikely is unlikely
24-hour urine:
K > 40 mEq and
Aldosterone > 15 mcg
No Check urine:
Cortisol
DOC
Adrenal CT scan

Hyperplasia or normal Equivocal Unilateral adenoma

Postural stimulation test (+) (or)


Plasma 18 OHB > 100 ng% (or)
Elevated urinary 18 OH-F, 18 oxo-F

No
Yes

Adrenal sampling

Not
Lateralized Lateralized

Medication Adrenalectomy
Figure 8-7 Identifying primary hyperaldosteronism.
138 Part II Adrenal Gland

Effective

RENAL
circulating History Suspect
blood
potassium volume
physical exam
excretion BP x 3 > 140/90 pheochromocytoma
Renal sodium Renal perfusion
retention pressure Basic lab tests

Aldosterone Juxtaglomerular
secretion apparatus
Angiotensinogen Plasma norepinephrine
Angiotensin II Renin release Epinephrine
Diagnosis Elevated
Dopamine
Potassium pheochromocytoma
balance
Converting Angiotensin I Urinary catechols
enzyme

Normal
Figure 8-8 Control of aldosterone secretion by means of
Localize
interrelationships between the potassium and renin- − Evaluate for
angiotensin feedback loops. CTT − Venous sampling other causes
MIBG
+
aldosterone secretion. Therefore, the term secondary MRI
+
hyperaldosteronism is utilized when there is increased renin α- blockade, then remove
secretion and secondary aldosterone production.27,28 The
Figure 8-9 Identifying pheochromocytoma.
most common examples of secondary hyperaldostero-
nism would be renovascular hypertension and malignant
hypertension. In contrast, with an adrenal adenoma or
adrenal hyperplasia there is primary secretion of aldos- catheter placement. An appropriate way of analyzing
terone and subsequently the sodium retention that aldosterone levels is with comparative aldosterone/corti-
occurs leads to a suppression of plasma renin activity. sol ratios from each side. It is our general policy to have
Therefore, returning to Figure 8-7, the hallmark of positive lateralizing information, as well as a positive CT
the entity is hypokalemia. However, some patients real- scan, before recommending exploration and unilateral
ize that weakness occurs with increased sodium intake adrenalectomy. However, more recently, in patients who
and therefore restrict their sodium, and may have a have elevated plasma 18-hydroxy-B levels and elevated
more normal potassium than that first observed. urinary 18-hydroxy-F levels, at times we have not
Therefore, the entity should not be ruled out until the required sampling when a clear adenoma was demon-
patient has sodium loading with 10 g of sodium a day strated on CT scan. In contrast, we have demonstrated a
for several weeks and repeat potassium measurements. subset of patients with radiographic bilateral hyperplasia
A small subset of patients exhibits normokalemic who will lateralize adrenal vein sampling for aldosterone.
hyperaldosteronism, and if there is a high index of sus- In this setting, we have performed unilateral adrenalec-
picion for the disease, these patients should be studied tomy and a significant number of those patients have
further. If there is hypokalemia, a 24-hour urine should favorable biochemical and blood pressure responses,
be collected demonstrating that there is urinary loss of although most have required the continuation of some
potassium. The critical test is the measurement of antihypertensive medication.24
plasma renin activity at a time when the patient is Finally, in patients who have normal CT scans yet lat-
either on a low-sodium diet or is challenged with a eralize on sampling, if they show elevated 18-hydroxy
diuretic. If the patient has hyperaldosteronism, the products, we will operate; if not, we will follow those
plasma renin activity remains inappropriately low patients. The majority of patients with bilateral hyper-
despite sodium depletion. Because potassium is also a plasia will not lateralize with adrenal vein sampling for
stimulus of aldosterone, the patient should be potas- aldosterone. Those patients are treated with spironolac-
sium repleted before measuring 24-hour urine and tone at an appropriate dose to control blood pressure.
plasma aldosterone levels. Both of these values should Often, they will need other medications, such as calcium
be elevated in hyperaldosteronism. channel blockers.
At this point the question is whether the patient has a
unilateral adenoma or bilateral adrenal hyperplasia, and
PHEOCHROMOCYTOMA
the imaging study of choice is an adrenal CT scan with 3
to 5-mm cuts through both adrenal glands. The next step Pheochromocytoma is an uncommon entity, but one that
that is traditionally performed would be adrenal vein has potentially lethal sequelae for the patient if not diag-
sampling. The difficulty with adrenal vein sampling is nosed. Therefore, it is generally felt that all patients with
obtaining adequate collections from the short, stubby, sustained hypertension should have the appropriate
right adrenal vein, and when samples are collected, corti- studies performed to rule out pheochromocytoma
sol levels should also always be collected to ensure proper (Figure 8-9).29,30
Chapter 8 Adrenal Tumors 139

The clinical manifestations exhibited by patients with these tumors. The mechanism of the increased incidence of
pheochromocytoma are due to the physiologic effects of pheochromocytomas in association with neuroendocrine
the catecholamines, dopamine, epinephrine, and norep- dysplasias and medullary carcinoma of the thyroid may be
inephrine. However, other signs and symptom com- explained by the amine precursor uptake and decarboxyla-
plexes exhibited may be extremely variable, including tion (APUD) cell system of Pierce. The APUD cells
the asymptomatic patient in whom a lesion is picked up derived from the neural crest of the embryo share common
simply on CT scan. In all reported series, hypertension ultrastructural and cytochemical features and elaborate
is by far the most common sign (Table 8-3). As far as the amines by precursor uptake and decarboxylation.32,33
type of hypertension, the patients may have either sus- The laboratory diagnosis of pheochromocytoma is
tained hypertension, paroxysmal or dramatic attacks of now extremely accurate, utilizing the urinary plasma
hypertension, or sustained hypertension with superim- measurements of catecholamines and their by-products
posed paroxysms. Most series have shown this latter (see Figure 8-9). Extremely accurate assays exist for these
constellation of findings to be the most common in amines.34 At the present time it is felt that urinary cate-
patients with pheochromocytoma. In addition, the fre- cholamines remain the measurement of choice with the
quency of attacks among patients is quite variable, rang- measurement of total urinary catecholamines and
ing from a few times a year to multiple daily episodes. In metanephrines. Approximately 95% of patients will have
addition, the duration may be minutes or hours and the elevated levels of these substances. In the patient with a
nature of the attacks can vary dramatically. Most patients severe paroxysmal hypertension who presents in the
will exhibit a paroxysm or an episode once a week, and midst of hypertensive crisis, the plasma catecholamines
most of the attacks will last less than an hour. Usually, the are almost always elevated and can be utilized.
attacks occur in the absence of recognizable stimuli, but a Stimulation or suppression tests are generally not uti-
number of factors—particularly exercise, posture, trauma, lized at the present time. The one situation where they
or a variety of other situations—may precipitate an attack. may be useful is in the patient who appears to have essen-
One specific entity is noteworthy: catecholamine- tial hypertension but borderline elevated catecholamines,
induced cardiomyopathy. 31 Patients with cate- and in this setting a clonidine suppression test may be
cholamine-induced cardiomyopathy will present with useful. Following a single 0.3-mg oral dose of clonidine
decreased cardiac function and congestive heart failure, the patients with neurogenic hypertension at rest show a
and it is mandatory that their cardiac status be stabilized fall in norepinephrine, whereas patients with pheochro-
with the use of appropriate a- and b-adrenergic block- mocytomas do not.34
ing agents as well as a-methylparatyrosine (a tyrosine The radiographic test that is most useful in both iden-
hydroxylase inhibitor) (Figure 8-10) to cut down on cat- tifying and characterizing neuroendocrine adrenal
echolamine production before surgery is contemplated. tumors, and in identifying surrounding structures, is the
Generally, the cardiomyopathy is reversible, and the MRI scan. We have been impressed with the multiple
patients can be operated on within weeks or months uses of MRI scans in patients with pheochromocytoma.
after the initial diagnosis and treatment is instituted. Therefore, the test is as accurate as a CT scan in identi-
An appreciable number of pheochromocytomas have fying lesions and also has a characteristic bright light
been found in association with other disease entities and bulb appearance on the T2-weighted study (see Figure
hereditary syndromes. These entities include the associ- 8-10).3 In addition, sagittal and coronal imaging can pro-
ation of tumors of the glomus jugulare region, neurofi- vide excellent anatomic information concerning the rela-
bromatosis, Sturge-Weber syndrome, and the von tionship between the tumor and the surrounding
Hippel-Landau and familial multiendocrine adenopathy vasculature. Therefore, we feel that the MRI should be
(MEA) syndromes. Pheochromocytomas occur in MEA- the initial scanning procedure in patients with the bio-
2, a triad including pheochromocytoma, medullary carci- chemical findings of pheochromocytoma.
noma of the thyroid, and parathyroid adenomas (Sipple’s An alternative approach that also is useful at times,
syndrome). Pheochromocytomas may also be a part of particularly for residual or multiple pheochromocy-
MEA-3, which also includes medullary carcinoma of the tomas, is the metaiodobenzylguanidine (MIBG) scan that
thyroid, mucosal neuromas, thickened corneal nerves, images medullary tissue.35,36 This test may be more sen-
ganglioneuromatosis, and frequently marfanoid habitus. sitive than CT or MRI picking up small extra-adrenal
It is now believed that the relatives of patients with all of lesions and has major use in patients where multiple
these syndromes should be evaluated for the presence of lesions are suspected.
occult pheochromocytoma. In addition, there is a well-
known entity of familial pheochromocytoma in which
ADRENAL SURGERY
multiple members of the kindred will be found to have
multiple lesions and all members of such families should Adrenalectomy is the treatment of choice in most patients
be both screened and then followed for the appearance of who have undergone appropriate metabolic evaluation
140 Part II Adrenal Gland

Table 8-3 Symptoms Reported by 76 Patients (Almost all Adults) with


Pheochromocytoma Associated with Paroxysmal or Persistent Hypertension
Paroxysmal P e r s i s t e n t
Symptoms (37 Patients) (%) (39 Patients) (%)

Symptoms Presumably Due to Excessive Catecholamines or Hypertension


Headache (severe) 92 72
Excessive sweating (generalized) 65 69
Palpitations ± tachycardia 73 51
Anxiety or nervousness (± fear of impending death, panic) 60 28
Tremulousness 51 26
Pain in chest, abdomen (usually epigastric), lumbar regions,
lower abdomen, or groin 48 28
Nausea ± vomiting 43 26
Weakness, fatigue, prostration 38 15
Weight loss (severe) 14 15
Dyspnea 11 18
Warmth ± heat intolerance 13 15
Visual disturbances 3 21
Dizziness or faintness 11 3
Constipation 0 13
Paresthesia or pain in arms 11 0
Bradycardia (noted by patient) 8 3
Grand mal 5 3

Manifestations Due to Complications


Congestive heart failure ± cardiomyopathy
Myocardial infarction
Cerebrovascular accident
Ischemic enterocolitis ± megacolon
Azotemia
Dissecting aneurysm
Encephalopathy
Shock
Hemorrhagic necrosis in a pheochromocytoma

Manifestations Due to Coexisting Diseases or Syndromes


Cholelithiasis
Medullary thyroid carcinoma ± effects of secretions of serotonin,
calcitonin, prostaglandin, or ACTH-like substance
Hyperparathyroidism
Mucocutaneous neuromas with characteristic facies
Thickened corneal nerves (seen only with slit lamp)
Marfanoid habitus
Alimentary tract ganglioneuromatosis
Neurofibromatosis and its complications
Cushing’s syndrome (rare)
Von Hippel-Lindau disease (rare)
Virilism, Addison’s disease, acromegaly (extremely rare)

Symptoms Caused by Encroachment on Adjacent Structures or by Invasion and Pressure Effects


of Metastases

From Manger WM, Gifford RW Jr: Pheochromocytoma. In Laragh JH, Brenner BM (eds): Hypertension Pathophysiology Diagnosis and
Management. New York, Raven Press, 1990, with permission.
Chapter 8 Adrenal Tumors 141

Figure 8-10 MRI of pheochromocytoma.

and have been found to have a surgical lesion. Although pheochromocytoma is consultation with the anesthesiol-
most adrenal tumors are removed with a laparoscopic ogist, who can be well aware of the patient and can plan
approach, the principles of open adrenal surgery apply strategy for management.37
and warrant review. However, the surgeon must be aware Thus, the management of patients with an adrenal dis-
that there are unique aspects to the care in these patients, order is approached on a team basis, including experi-
including specific preoperative management as outlined enced endocrinologists, radiologists, anesthesiologists,
in Table 8-4. Accordingly, patients with hyperaldostero- and urologists or general surgeons.
nism who are generally healthy require spironolactone Numerous approaches can be made to the adrenal
100 to 400 mg/day to restore their potassium supply. gland (Table 8-5). The proper approach depends on the
Patients with Cushing’s syndrome have severe systemic underlying cause of adrenal pathology, the size of the adre-
effects from the hyperglucocorticoidism. They are often nal, the side of the lesion, the habitus of the patient, and
obese, have diabetic tendencies, are poor wound healers, the experience and preference of the surgeon. In most
easily sustain bony fractures, and are susceptible to infec- cases, there are a number of different options available,
tion. Thus, they are at high risk for complications. In and a careful review of all the variables is required before
selected patients with markedly elevated cortisol levels a choice is made. Thus, each case should be considered
the preoperative use of metabolic blockers, such as individually, although some approaches are preferable for
metyrapone, is required to reverse some of the clinical a given disease. For example, in patients with large adrenal
findings prior to adrenalectomy. Certainly, glucocorti- tumors, a thoracoabdominal approach is often utilized. In
coid replacement is required throughout the surgical contrast, a posterior or modified posterior approach is pre-
procedure and postoperatively until the function of the ferred for small localized lesions. Finally, in patients with
contralateral adrenal gland occurs. Finally, in patients multiple lesions, either extra-adrenal or bilateral will be
with a pheochromocytoma, adrenergic blockade gener- explored using a transabdominal chevron incision.
ally with Dibenzyline is required, and at times the block- Before describing the specific techniques, a number of
ade of catecholamine production with metyrosine is also unifying concepts warrant attention. First, adequate visu-
useful as previously discussed. The additional preopera- alization is imperative, as the adrenal glands lie high in
tive evaluation that is mandatory in patients with the retroperitoneum and quite posterior. Therefore, the
142 Part II Adrenal Gland

Table 8-4 Preoperative Management Table 8-5 Surgical Approaches in Adrenal Disorder
Treatment Approach

Primary hyperaldosteronism Spironolactone, 100–400 mg/ Primary hyperaldosteronism Posterior (left or right)
day, 2–3 weeks Modified posterior (right)
Follow K+ until normal Eleventh rib (left > right)
Blood pressure should fall Posterior transthoracic

Cushing’s syndrome Control of glucose Cushing’s adenoma Eleventh rib (left or right)
abnormalities Thoracoabdominal (large)
Documentation of Posterior (small)
osteoporosis
Cushing’s disease Bilateral posterior
Glucocorticoid replacement
Bilateral hyperplasia Bilateral eleventh rib
(before, during, and after
(alternating)
surgery)
Perioperative antibiotics Adrenal carcinoma Thoracoabdominal
Eleventh rib
Pheochromocytoma Adrenergic blockade
Transabdominal
Phenoxybenzamine
(Dibenzyline), Bilateral adrenal ablation Bilateral posterior
20–160 mg/day
Metyrosine (if needed) Pheochromocytoma Transabdominal (chevron)
Volume expansion Thoracoabdominal (large,
Crystalloid usually right)
β-Blockade if cardiac Eleventh rib
arrhythmias (only after
α-blockade established) Neuroblastoma Transabdominal
Anesthesia consultation Eleventh rib

From Vaughan ED Jr: Adrenal surgery. In Marshall FF (ed): Textbook From Vaughan ED Jr: Adrenal surgery. In Marshall FF (ed): Textbook
of Operative Urology. Philadelphia, WB Saunders Co, 1996, with of Operative Urology. Philadelphia, WB Saunders Co, 1996, with
permission. permission.

Posterior Approach
use of a headlight by both the surgeon and first assistant
is critical, and hemostasis should be rigorously maintained. The posterior approach can be used for either bilateral
The operator should bring the adrenal down by initially adrenal exploration or unilateral removal of small tumors
exposing the cranial attachments and dividing the rich (Figure 8-12).
blood supply between either right-angled clips or utiliz- The bilateral approach is rarely utilized today because
ing a forceps cautery or the harmonic scalpel. Thus, it is of our excellent localization techniques. It is now utilized
often simplest to begin the dissection laterally, identify- primarily for ablative total adrenalectomy. The options
ing the vascular supply and working around the cranial for incisions are shown in Figure 8-12; generally rib
edge of the gland. The posterior surface is generally resection is preferable to gain high exposure. After stan-
devoid of vasculature and after the gland is freed superi- dard subperiosteal rib resection, care should be taken
orly with gentle traction on the kidney, the gland can be with the diaphragmatic release, and the pleura should be
brought inferiorly for control of the adrenal vein. The avoided, and the diaphragm swept cranially.
only tumor handled in a different fashion would be a The fibrofatty tissues within Gerota’s fascia are swept
pheochromocytoma where intent should be made to away from the paraspinal musculature, exposing a subdi-
obtain control of the adrenal vein early so as to stabilize aphragmatic “open space” that is at the posterior apex of
the patient from a burst of catecholamine release during the resection. The liver within the peritoneum is dis-
manipulation. The adrenal gland is extremely friable and sected off the anterior surface of the adrenal and the cra-
fractures easily, which can cause troublesome bleeding. nial blood supply is divided. Medially on the right, the
Therefore, tension or traction should be maintained on IVC is visualized. The short, high adrenal vein entering
the kidney or surrounding structures and not on the the cava in a dorsolateral position is identified and can be
adrenal itself. The concept has been stated that the clipped or ligated. The adrenal can then be drawn cau-
“patient should be dissected from the tumor,” a view that dally by traction on the kidney. The adrenal arteries will
is particularly true for pheochromocytomas, in which the issue from behind the IVC and these must be carefully
glands should not be manipulated (Figure 8-11). clipped; otherwise, troublesome bleeding can occur.
Chapter 8 Adrenal Tumors 143

Figure 8-11 MRI of recurrent pheochromocytoma with an excellent demonstration of anterior


crossing right renal vein, feeding lumbar vein, and involvement of right renal artery.

Finally, the adrenal is removed from the superior


aspect of the kidney and care must be taken to avoid api-
cal branches of the renal artery. On the left, the approach
is similar with division of the splenorenal ligament given
initial lateral exposure.
The posterior approach can be modified for a
transthoracic adrenal exposure to the diaphragm38; how-
ever, this more extensive approach is rarely necessary for
small adrenal tumors.

Modified Posterior Approach


Although the posterior approach has the advantage of
rapid adrenal exposure and low morbidity, there are def-
inite disadvantages. This approach may impair respira-
tion, the abdominal contents are compressed posteriorly,
and the visual field is limited. In addition, if bleeding
occurs, it is difficult to extend the incision to gain a bet-
ter exposure. Therefore, we have developed a modified
posterior approach for right adrenalectomy utilizing the
Gil-Vernet position.39
The approach is based on the anatomic relationship
with the right adrenal, which lies deep posterior and high
in the retroperitoneum behind the liver (Figure 8-13A).
In addition, the short, stubby right adrenal vein enters
the IVC posteriorly at the apex of the adrenal. Hence, we
utilize an approach that is posterior, but the patient is in
a modified position, similar to that used for a Gil-Vernet
dorsal lumbotomy incision.40 The patient is first placed
in a formal lateral flank position and then allowed to fall
forward into the modified posterior position (see Figure
Figure 8-12 Posterior approach to the adrenals. 8-13B). Subsequently, the 11th or 12th rib is resected
144 Part II Adrenal Gland

Figure 8-13 Modified posterior approach to the right adrenal.

with care to avoid the pleura. The diaphragm then is dis- lateral, empty space can be found exposing the posterior
sected off the underlying peritoneum and liver in order abdominal musculature and often the IVC. Multiple
to gain mobility. Similarly, the inferior surface of the small arteries course behind the IVC and emerge over
peritoneum, closely associated with the liver, is sharply the paraspinal muscles, and these are clipped and divided.
dissected from Gerota’s fascia, which is gently retracted At this point the adrenal can usually be moved poste-
inferiorly. It is of note that the adrenal gland is not iden- riorly against the paraspinal muscles exposing the ante-
tified during the early portion of the dissection, and rior surface of the IVC below the adrenal gland.
because of the modified posterior approach, the surgeon The major advantage of this approach is that the adre-
can become disoriented if not thoroughly familiar with nal vein is easily identified because it emerges from the
anatomic relationships. segment of the IVC exposed and courses up to the adre-
The adrenal will become visible in the depth of the nal, which now rises toward the surgeon. In other flank
incision as the final hepatic attachments are divided. The or anterior positions the adrenal vein resides in its poste-
Chapter 8 Adrenal Tumors 145

rior relationship, requiring caval rotation and the chance kidney. The kidney is useful for retraction. The dissec-
of adrenal vein avulsion. After adrenal vein exposure, it is tion should continue from lateral to medial along the
doubly tied and divided or clipped with right-angle clip- posterior abdominal and diaphragmatic musculature,
pers and divided (see Figure 8-13D). with precise ligation or clipping of the small but multiple
The remaining removal of the adrenal is as we previ- adrenal arteries. While the operator clips these arteries
ously described for the posterior approach. with one hand, the opposite hand is employed to retract
On the left side we do not use this modified approach both adrenal and kidney inferiorly. With release of the
and use a standard flank approach with a fairly small superior vasculature, the adrenal becomes easily visual-
incision. ized. On the left medially, the phrenic branch of the
We have used the modified posterior approach for all venous drainage must be carefully clipped or ligated
patients with right adrenal aldosterone-secreting tumors (Figure 8-15). This vessel is not noted in most atlases but
and for other patients with benign adenomas of less than can cause troublesome bleeding if divided. The medial
6 cm. We do not recommend the approach for patients dissection along the crus of the diaphragm and aorta will
with large lesions or malignant adrenal neoplasms. The lead to the renal vein; finally, the adrenal vein is con-
approach has been used for patients with relatively small trolled, doubly tied, and divided. The adrenal is then
pheochromocytomas. removed from the kidney with care to avoid the apical
branches of the renal artery (see Figure 8-15).
On the right side, the dissection is similar. However,
Flank Approach
after release of the adrenal from the superior vasculature,
The standard extrapleural, extraperitoneal 11th rib resec- it is helpful to expose the IVC and divide the medial arte-
tion is excellent for either left or right adrenalectomy. rial supply. This maneuver allows mobilization of the cava
After completion of the incision, the lumbocostal arch is for better exposure of the high posterior adrenal vein,
utilized as a landmark showing the point of attachment of which is doubly tied or clipped and divided (Figure 8-16).
the posterior diaphragm to the posterior abdominal mus-
culature. Gerota’s fascia, containing the adrenal and kid-
ney, can be swept medially and inferiorly, giving exposure
to the splenorenal ligament on the left, which should be
divided to avoid splenic injury (Figure 8-14). Working
anteriorly on the left, the spleen and pancreas within the
peritoneum can be lifted cranially, exposing the anterior
surface of the adrenal gland.
On the right side, a similar maneuver is used to lift the
liver within the peritoneum off the anterior surface of the
adrenal. Quite often the adrenal gland cannot be identi-
fied precisely until these maneuvers are performed. One
should not attempt to dissect into the body of the adre-
nal or to dissect the inferior surface of the adrenal off the

Figure 8-14 Release of splenorenal ligament early in Figure 8-15 Further exposure of left adrenal including
exposure of left adrenal. phrenic vein.
146 Part II Adrenal Gland

Figure 8-16 Exposure of right adrenal with and without nephrectomy.

Patients with large adrenal carcinomas may require an en the adrenal vein is divided because a marked drop in
bloc resection of the adrenal and kidney following the blood pressure often occurs, even when the patient is
principles of radical nephrectomy (see Figure 8-16). adequately hydrated.
A major deviation from this technique is used for the After removal of the adrenal, inspection should be
patient with pheochromocytoma, in whom the initial dis- made for any bleeding and for pleural tears of the
section should be aimed toward early control and divi- diaphragm. The kidney should also be inspected. The
sion of the main adrenal vein on either side. Obviously, in incision is closed without drains with interrupted 0 poly-
this setting, the anesthesiologist should be notified when dioxanone sutures.
Chapter 8 Adrenal Tumors 147

Thoracoabdominal Approach
The thoracoabdominal 9th or 10th rib approach is uti-
lized for large adenomas; for some large adrenal carcino-
mas, and for well-localized pheochromocytomas. The
incision and exposure are standard, with a radial incision
through the diaphragm and a generous intraperitoneal
extension. The techniques described for adrenalectomy
with the 11th rib approach are used.

Transabdominal Approach
The transabdominal approach is commonly selected for
patients with pheochromocytomas, for children, and for
some patients with adrenal carcinomas. The concept is to
have the ability for complete abdominal exploration to
identify either multiple pheochromocytomas or adrenal
metastases.
I use the transverse or chevron incision, which I
believe gives better exposure of both adrenal glands than
a midline incision. The rectus muscles and lateral
abdominal muscles are divided, exposing the peritoneum.
Upon entering the peritoneal cavity, the surgeon should
gently palpate the para-aortic areas and the adrenal areas.
Close attention is given to blood pressure changes in an
attempt to identify any unsuspected lesions if the patient Figure 8-17 Exposure of right adrenal and left adrenal
utilizing a transabdominal approach.
has a pheochromocytoma. This maneuver is less impor-
tant today because of the excellent localization tech-
niques previously discussed. In fact, with precise
preoperative localization of the offending tumor, the
chevron incision does not need to be completely sym-
metric and may be limited on the contralateral side.
If the patient has a lesion on the right adrenal, the
hepatic flexure of the colon is reflected inferiorly. The
incision is made in the posterior peritoneum lateral to the
kidney and carried superiorly, allowing the liver to be
reflected cranially (Figure 8-17). Incision in the peri-
toneum is carried downward, exposing the anterior sur-
face of the IVC to the entrance of the right renal vein.
Once the cava is cleared, one or two accessory hepatic
veins are often encountered, which should be secured
(Figure 8-18B). These veins are easily avulsed from the
cava and may cause troublesome bleeding. Ligation of
these veins gives 1 to 2 cm of additional caval exposure of
the short posterior right adrenal vein. Small accessory
adrenal veins may also be encountered. The cava is then
rolled medially, exposing the adrenal vein, which should
be doubly tied or clipped and divided (Figure 8-18C).
After control of the adrenal vein, it is simplest to pro-
ceed with the superior dissection, lifting the liver off the
adrenal and securing the multiple small adrenal arteries
arising from the inferior phrenic artery, which is rarely
seen. The adrenal can be drawn inferiorly with retraction
on the kidney, and the adrenal arteries traversing to the
adrenal from under the cava can be secured with right-
angled clips. The final step is removing the adrenal from Figure 8-18 Further transabdominal exposure of the right
the kidney. adrenal with ligation of an accessory right hepatic vein.
148 Part II Adrenal Gland

The left adrenal vein is not as difficult to approach lesions. Preoperative evaluation should identify these
because it lies lower, partially anterior to the upper pole lesions, but, regardless, a careful abdominal exploration
of the kidney, and the adrenal vein empties into the left should be carried out.
renal vein. Accordingly, on the left side, the colon is In patients with suspected malignant pheochromocy-
reflected medially, exposing the anterior surface of tomas, en bloc dissections may be necessary to obtain
Gerota’s capsule; the initial dissection should involve adequate margins, a concept that also applies in patients
identification of the renal vein (see Figure 8-17B). In with adrenal carcinomas. Evaluation with MRI to obtain
essence, the dissection is the same as for a radical transverse, coronal, and sagittal images is extremely use-
nephrectomy for renal carcinoma. Once the renal vein is ful to define clearly the adrenal relationships to the IVC
exposed, the adrenal vein is identified, doubly ligated, and renal vessels as well as to localize the adrenal vein.
and divided. After this maneuver the pancreas and splenic In patients with pheochromocytomas, postoperative
vasculature are lifted off the anterior surface of the adre- management includes maintenance of arterial and venous
nal gland. Because of additional drainage from the adre- lines in an intensive care setting until they are stable. Often,
nal into the phrenic system, I generally continue the 24 to 48 hours are required for the full effect of phenoxy-
medial dissection early to control the phrenic vein. I then benzamine, the α-blocking agent commonly given, to wear
work cephalad and lateral to release the splenorenal liga- off and for normal α-receptor activity to be restored.
ment and the superior attachments of the adrenal. The
remainder of the dissection is carried out as previously
PARTIAL ADRENALECTOMY
described.
After removal of the tumor, regardless of size, careful The standard treatment for patients with the adrenal
inspection is made to ensure hemostasis and the absence lesions described has been total adrenalectomy.
of injury to adjacent organs. Careful abdominal explo- However, there recently has been reported an excellent
ration is carried out, after which the wound is closed with paper showing the utility of partial adrenalectomy in
the suture material of choice. No drains are used. patients with primary hyperaldosteronism.41 I have not
Patients with multiple endocrine adenopathy of family used partial adrenalectomy in a patient for normal con-
histories of pheochromocytoma, as well as pediatric tralateral adrenal, but certainly have used the technique
patients, should be considered at high risk for multiple in patients with bilateral disease (Figure 8-19). Thus, in

Figure 8-19 MRI showing bilateral adrenal pheochromocytoma in a patient with bilateral
glomus jugulare tumors. A, Small right adenoma that was enucleated and which was
partially resected. B, Large left bright pheochromocytoma that was totally removed.
Chapter 8 Adrenal Tumors 149

one patient with a pheochromocytoma on one side and A variety of laparoscopic approaches to the adrenal
a nonfunctioning adenoma on the other, the adenoma exist.50,51 The lateral transperitoneal, anterior transperi-
was simply enucleated from the adrenal. In a second toneal, lateral retroperitoneal, and posterior retroperi-
patient with bilateral pheochromocytomas, the larger toneal techniques have been described similar to the
lesion was totally excised with partial adrenalectomy rationale for an open approach. The laparoscopic
was utilized to remove the contralateral tumor. Care has approach depends on the patient’s habitus, the underly-
to be taken to obtain thorough hemostasis when per- ing pathology and the skill and experience of the operat-
forming a partial adrenalectomy because of the vascular ing surgeon.1 The results of these approaches mirror the
nature of the adrenal. Partial adrenalectomy or adrenal results of open adrenalectomy with less morbidity and
sparing surgery is most useful in patients at risk for hospitalization time for the patient.52
multiple adrenal tumors, such as von Hippel-Landau The lateral transperitoneal approach is the technique
kindreds.42,43,44 most often reported in the literature. Most laparoscopic
surgeons have extensive experience identifying, dissect-
ing, and mobilizing the adjacent organs required in order
CRYOSURGERY
to obtain adrenal exposure and removal. For this
Cryoablation is currently used as a surgical alternative for approach the patient is placed in a full lateral position
the treatment of prostatic, lung, brain, pharyngeal, and (Figure 8-20A and B). Bilateral adrenalectomy requires
liver tumors. We have demonstrated in a canine model45 repositioning and redraping.
that adrenal cryoablation is effective in destroying adre- In contrast, the retroperitoneal approaches avoid dis-
nal tissue and is safe. We have successfully used the tech- section and mobilization of intra-abdominal viscera
nique in one patient with primary hyperaldosteronism. (Figure 8-21A and B). The major limitation is the small
Adrenal laparoscopic cryoablation may shorten operative working space that compromises instrument placement
time and be as effective as total adrenalectomy in patients and crossing of instruments can occur. The approach is
with small lesions. best for patients with small adrenal tumors. Balloon infla-
tion is used to dissect the retroperitoneal space. During
this procedure, close blood pressure monitoring is neces-
ABLATION
sary in patients with pheochromocytoma since the
Successful adrenal ablation using transcatheter arterial expanding balloon may compress the tumor with catec-
infusion of ethanol has been described in 33 cases of pri- hol release.
mary hyperaldosteronism; the approach was successful in Regardless of the approach utilized the principles of
27 cases (82%). Five patients required surgical adrenalec- adrenal surgery previously described are the same.
tomy. This technique may be useful in the patient who is
at high risk with use of general anesthesia.46 More
SUMMARY
recently, direct percutaneous tumor injection with
ethanol has given excellent results in 41 patients with We are fortunate that our ability to diagnose the specific
pheochromocytoma with reversal of hormonal abnor- adrenal entities that mandate a surgical approach is
malities.47 extremely accurate. The combination of analytic
methodology to measure the appropriate adrenocortical
and medullary hormonal production and the radiologic
LAPAROSCOPIC ADRENALECTOMY
techniques for localization are superb. The management
Laparoscopic adrenalectomy, first reported in 1991,48 is of these adrenal disorders usually employing a laparo-
now the surgical approach of choice for adrenal removal scopic approach following localization is highly success-
in the majority of patients.49 The exceptions are patients ful, resulting in a reversal of both metabolic
with large irregular adrenal carcinoma where adjacent abnormalities and the hypertension that often accompa-
organs may be involved, large pheochromocytomas, nies these diseases. Indeed, this is a true success story
patients with large adrenal hemorrhage, and in some with the evolution of these different techniques over the
cases of metastatic disease. past 50 years.
150 Part II Adrenal Gland

Figure 8-20 A, Trocar placement for left transperitoneal adrenalectomy. The distribution is a
mirror image of that used for the left side. Dissection of the left adrenal gland: the spleen (3),
pancreas (4), left lobe of the liver (2), renal vein (5), and kidney are shown. The left adrenal
vein (1) has been isolated. A clip is applied to the adrenal vein before dividing it (inset, right).
The inset (left) shows the patient’s position on the operating table. B, Trocar placement for a
right transperitoneal adrenalectomy: supra-umbilical trocar for camera (if only three trocars
are used) or splenic retractor (if four trocars are used). Trocars at anterior axillary line and
midaxillary line for instruments for dissection. Fourth trocar halfway between midline and
anterior is shown. A clip is applied to the adrenal vein before dividing it (inset, right). The
inset (left) shows the patient’s position on the operating table.

Figure 8-21 A, The retroperitoneal approach for the left adrenal gland (1): the adrenal vein
(2) is seen anterior to the renal artery (4); the renal artery (4) and vein (6) are identified early
in the dissection. The kidney (7) and ureter (8) are also depicted. The inset (left) shows the
patient’s position on the operating table.
Continued
Chapter 8 Adrenal Tumors 151

Figure 8-21 cont’d B, The retroperitoneal approach for the right adrenal gland (1): the
adrenal vein (2) is seen at its takeoff from the vena cava (5); the renal artery (4) and vein (6)
are identified early in the dissection. The kidney (7) and ureter (8) are also depicted. The
inset (left) shows the patient’s position on the operating table.

REFERENCES incidentally discovered adrenal and juxtaadrenal masses.


Ann Nucl Med 1993; 7(3):157–166.
1. Vaughan ED Jr, Blumenfeld JD, Del Pizzo J, Schichman SJ, 11. Ross NS, Aron DC: Hormonal evaluation of the patient
Sosa RE: The adrenals. In Walsh PC, Retik AB, Vaughan with an incidentally discovered adrenal mass. N Engl J
ED Jr, Wein AJ (eds): Campbell’s Urology, 8th edition. Med 1990; 323:1401.
Philadelphia, WB Saunders Co, 2002. 12. Cerfolio RJ, Vaughan ED Jr, Brenan TC, Hiruela ER:
2. Belldegrun A, Ritchie A, Figlin R, Oliver R, Vaughan ED Accuracy of computed tomography in predicting adrenal
Jr (eds): Renal and Adrenal Tumors. Oxford, Oxford tumor size. Surg Gynecol Obstet 1993; 176:307.
University Press, 2003. 13. Mayo-Smith WW, Lee MJ, McNicholas MM et al:
3. Taneja SS, Smith RB, Ehrlich RM (eds): Complications of Characterization of adrenal masses (<5 cm) by use of
Urologic Surgery Prevention and Management. chemical shift MR imaging: observer performance vs.
Philadelphia, WB Saunders Co, 2001. quantitative measures. AJR 1995; 165:91–95.
4. Lee MJ, Mayo-Smith WW, Hann PE, et al: State-of-the- 14. Lubat E, Weinreb JC: Magnetic resonance imaging of the
art MR imaging of the adrenal gland. Radiographics 1994; kidneys and adrenals. Top Magn Reson Imaging 1990;
14:1015–1029. 2:17–36.
5. Vaughan ED Jr: Adrenal surgery. In Marshall FF (ed): 15. Tikkakoski T, Taavitsainen M, Paivansalo M, et al:
Textbook of Operative Urology. Philadelphia, WB Accuracy of adrenal biopsy guided by ultrasound and CT.
Saunders Co, 1996. Acta Raiol 1991; 32:371–374.
6. Orth DN: Cushing’s syndrome. N Engl J Med 1995; 16. Luton J-P, Cerdas S, Billaud L, et al: Clinical features of
32:791. adrenocortical carcinoma, prognostic factors and the
7. Teeger S, Papanicolaou N, Vaughan ED Jr: effect of mitotane therapy. N Engl J Med 1990; 322:1195.
Imaging of adrenal masses. In Belldegrun A, Ritchie A, 17. Imperato-McGinley J, Young IS, Huang T, et al:
Figlin R, Oliver R, Vaughan ED Jr (eds): Renal and Testosterone secreting adrenal cortical adenomas. Int J
Adrenal Tumors. Oxford, Oxford University Press, Gynaecol Obstet 1981; 19:421.
2003. 18. Gabrilove JL, Sharma DC, Waitz HH, Dorfman R:
8. Murai M, Marumo K: Selection of patients with Feminizing adrenal cortical tumors in the male: a review
incidentally discovered adrenal masses for operation. In of 52 cases including a case report. Medicine 1965; 44:37.
Belldegrun A, Ritchie A, Figlin R, Oliver R, Vaughan ED 19. Richie JP, Gittes RF: Carcinoma of the adrenal cortex.
Jr (eds): Renal and Adrenal Tumors. Oxford, Oxford Cancer 1980; 45:1957.
University Press, 2003. 20. Schulick RD, Brennan MF: Adrenocortical carcinoma. In
9. Ng L, Libertino JM: Adrenal cortical carcinoma: Belldegrun A, Ritchie A, Figlin R, Oliver R, Vaughan ED
diagnostic evaluation and treatment. J Urol 2003; 169: Jr (eds): Renal and Adrenal Tumors. Oxford, Oxford
1—11. University Press, 2003.
10. Nakajo M, Nakabeppu Y, Yonekura R, et al: The role of 21. Haak HR, van Seters AP, Moolenaar AJ, Fleuren GJ:
adrenocortical scintigraphy in the evaluation of unilateral Expression of P-glycoprotein in relation to clinical
152 Part II Adrenal Gland

manifestation, treatment and prognosis of adrenocortical 37. Malhotra V (ed): Anesthesia for Renal and Genitourinary
cancer. Eur J Cancer 1993; 29A:1036–1038. Surgery. New York, McGraw-Hill, 1995.
22. Wooten MD, King DK: Adrenal cortical carcinoma. 38. Novick AC, Straffon RA, Kaylor W: Posterior
Epidemiology and treatment with mitotane and a review transthoracic approach for adrenal surgery. J Urol 1989;
of the literature. Cancer 1993; 72:3145–3155. 141:254.
23. Conn JW: Primary hyperaldosteronism. A new clinical 39. Vaughan ED Jr, Phillips H: Modified posterior approach
syndrome. J Lab Clin Med 1955; 45:3. for right adrenalectomy. Surg Gynecol Obstet 1987;
24. Blumenfeld JD, Vaughan ED Jr.: Diagnosis and treatment 165:453–455.
of primary hyperaldosteronism. In Belldegrun A, Ritchie A, 40. Gil-Vernet J: New surgical concepts in removing renal
Figlin R, Oliver R, Vaughan ED Jr (eds): Renal and calculi. Urol Int 1965; 20:255–262.
Adrenal Tumors. Oxford, Oxford University Press, 2003. 41. Nakada T, Kubota Y, Sasagawa I, et al: Therapeutic
25. Laragh JH, Angers M, Kelly WG, et al: The effect of outcome of primary aldosteronism: adrenalectomy versus
epinephrine, norepinephrine, angiotensin II, and others enucleation of aldosterone-producing adenoma. J Urol
on the secretory rate of aldosterone in man. JAMA 1960; 1995; 153:1775–1780.
2:174–234. 42. Walther MM, Keiser HR, Choyke PL, et al: Management
26. Laragh JH, Sealey JE: The renin-angiotensin-aldosterone of hereditary pheochromocytoma in von Hippel-Lindau
system and the renal regulation of sodium, potassium, and kindreds with partial adrenalectomy. J Urol 1999;
blood pressure homeostasis. In Windhager EE (ed): 161:395–398.
Handbook of Physiology. New York, Oxford University 43. Walther MM, Herring J, Choye PL, Linehan WM:
Press, 1992. Laparoscopic partial adrenalectomy in patients with
27. Vaughan ED Jr, Sosa RE: Renovascular hypertension and hereditary forms of pheochromocytoma. J Urol 2000;
ischemic nephropathy. In Gillenwater JY, Grayhack JT, 164:14–17.
Howards SS, Mitchell ME (eds): Adult and Pediatric 44. Pavlovich CP, Walther MM: Partial adrenalectomy:
Urology, 4th edition, Chap 23, pp 973–998. Lippincott, indications and technique. In Belldegrun A, Ritchie A,
Williams & Wilkins, 2002. Figlin R, Oliver R, Vaughan ED Jr (eds): Renal and
28. Mann SJ, Atlas SA: Hypertensive emergencies. In Laragh Adrenal Tumors. Oxford, Oxford University Press,
JH, Brenner BM (eds): Hypertension, Pathophysiology, 2003.
Diagnosis and Management. New York, Raven Press, 1995. 45. Schulsinger DA, Sosa RE, Perlmutter AP, Vaughan ED
29. Manger WM, Gifford RW Jr: Pheochromocytoma: Jr.: Acute and chronic interstitial cryotherapy of the
a clinical review. In Laragh JH, Brenner BM (eds): adrenal. In Belldegrun A, Ritchie A, Figlin R, Oliver R,
Hypertension, Pathophysiology, Diagnosis and Vaughan ED Jr (eds): Renal and Adrenal Tumors. Oxford,
Management. New York, Raven Press, 1995. Oxford University Press, 2003.
30. Walther MM, Eisenhoffer G, Pacak K, Linehan WM: 46. Ueno K, Nakajo M, Miayazono, N, et al: Transcatheter
Pheochromocytoma. In Belldegrun A, Ritchie A, Figlin R, adrenal arterial embolization of cortisol-producing
Oliver R, Vaughan ED Jr (eds): Renal and Adrenal tumors: two cases of Cushing’s Syndrome. J Vascular
Tumors. Oxford, Oxford University Press, 2003. Interventional Radiol 2000; 11:141.
31. Imperato-McGinley J, Gautier T, Ehlers K, et al: 47. Wang P, Zuo C, Qian Z, et al: CT-Guided percutaneous
Reversibility of catecholamine-induced dilated ethanol injection in the treatment of hyperfunctioning
cardiomyopathy in a child with a pheochromocytoma. pheochromocytoma. J Urol 2003; 170:1132–1134.
N Engl J Med 1987; 316:793–797. 48. Gagner M, Lacroix A, Bolte E: Laparoscopic
32. Bolande RP: The neurocrestopathias: a unifying concept adrenalectomy and Cushing’s syndrome and
of disease arising in neurocrest maldevelopment. Hum pheochromocytoma. N Engl J Med 1992; 327, 1003–1006.
Pathol 1974; 5:409. 49. Del Pizzo JJ, Shichman SJ, Sosa RE: Laparoscopic
33. Pearse AG, Polak JM: Cytochemical evidence for the adrenalectomy: the New York-Prebyterian Hospital
neural crest origin of mammalian ultimobranchial C cells. experience. J Endosc 2002; 16:591.
Histochemie 1971; 27:96. 50. Meraney AM, Gill IS: Laparoscopic versus open
34. Liu Z, Siragy HM, Carey RM: Diagnostic tests of adrenal adrenalectomy. In Belldegrun A, Ritchie A, Figlin R,
cortical and medullary function. In Belldegrun A, Ritchie Oliver R, Vaughan ED Jr (eds): Renal and Adrenal
A, Figlin R, Oliver R, Vaughan ED Jr (eds): Renal and Tumors. Oxford, Oxford University Press, 2003.
Adrenal Tumors. Oxford, Oxford University Press, 2003. 51. Shichman S, Sosa RE, Vaughan ED Jr.: Lateral
35. Shapiro B, Copp JE, Sisson JC, et al: Iodine-131 transperitoneal laparoscopic adrenalectomy. In Belldegrun
metaiodobenzylguanidine for the locating of suspected A, Ritchie A, Figlin R, Oliver R, Vaughan ED Jr (eds):
pheochromocytoma: experience in 400 cases. J Nucl Med Renal and Adrenal Tumors. Oxford, Oxford University
1985; 26:576. Press, 2003.
36. Campeau RJ, Garcia OM, Correa OA, Rege AB: 52. Baba S, Iwamura M: Laparoscopic adrenalectomy
Pheochromocytoma: diagnosis by scintigraphy using by the posterior lumbar approach. In Belldegrun A,
iodine-131 metaiodobenzylguanidine. South Med J 1991; Ritchie A, Figlin R, stetal (eds): Renal and Adrenal Tumors.
84:1221–1230. Oxford, Oxford University Press, 2003.
C H A P T E R

9 Open and Laparoscopic Surgery


of Adrenal Tumors
David S. Wang, MD, and Howard N. Winfield, MD

The adrenal glands are known to harbor a variety of typical examples of adrenal lesions diagnosed on CT and
benign and malignant tumors. Due to the size and loca- MRI. The differential diagnosis of the incidental adrenal
tion of the adrenals, lesions of the adrenal gland are mass is extensive and includes the benign nonfunctioning
rarely detected because of local symptomatic growth. adenoma, hormonally active cortical tumor, myelolipoma,
Instead, most adrenal tumors are diagnosed either inci- pheochromocytoma, adrenocortical carcinoma, and
dentally or because of hormonal activity. In general, metastatic lesion.
lesions that should be surgically removed include lesions Tumors diagnosed incidentally on CT scan or MRI are
that are hormonally active, tumors suspicious for adreno- managed according to size and hormone functional status.
cortical carcinoma, or nonfunctioning adrenal lesions Patients with hormonally active adrenal tumors, such as
5 cm or greater in size. aldosteronoma, Cushing’s syndrome, or pheochromocy-
Traditionally, surgical procedures involving the adre- toma, should generally undergo surgical removal.
nal gland were performed through an open incision with Hormonal evaluation of these patients is critical because
a variety of approaches. In 1992, the laparoscopic preoperative and postoperative considerations regarding
approach to adrenalectomy was introduced,1 offering a hypertensive control, electrolyte imbalances, and fluid
less invasive alternative to open adrenalectomy. As expe- shifts are paramount to ensure good surgical outcomes and
rience with laparoscopic adrenalectomy is increasing, the minimize complications. A summary of standard labora-
indications for laparoscopic adrenalectomy have tory tests in the evaluation of an adrenal lesion is listed in
expanded while the absolute contraindications have Table 9-1. Most hormonally active tumors should be
diminished. Indeed, laparoscopic adrenalectomy has removed, particularly in the case of pheochromocytoma
become the standard of care and the technique of choice and cortisol-secreting tumors.2,3 Occasionally, medical
for most benign adrenal lesions. management of aldosteronomas may be satisfactory to cir-
This chapter reviews the basic diagnosis and hormonal cumvent the need for surgical management, particularly in
evaluation, indications, preoperative considerations, and patients who are poor surgical candidates.4 However, side
techniques of open and laparoscopic adrenalectomy for effects of pharmacotherapy may become intolerable.
benign and malignant tumors of the adrenal gland. The Hormonally inactive tumors have traditionally been
full evaluation of the patient with an adrenal lesion is dis- managed according to size. Tumors less than 3 cm are
cussed in the previous chapter. almost always benign adenomas and generally require no
further treatment unless clinical signs of hormonal activ-
ity develop. Tumors greater than 6 cm are worrisome for
DIAGNOSIS
adrenocortical carcinomas, and thus surgical excision is
Adrenal lesions historically were diagnosed secondary to recommended given the aggressive nature of adrenal can-
clinical manifestations of endocrinopathies. However, cer.5 Nonfunctional lesions between 3 and 5 cm generally
widespread use of abdominal ultrasound, computed require close follow-up with serial imaging studies every
tomography (CT) scanning, and magnetic resonance 6 months. These lesions should be removed if tumors
imaging (MRI) has led to the not-infrequent finding of demonstrate interval change in appearance or develop
the incidental adrenal mass. Figures 9-1 and 9-2 show endocrine activity.

153
154 Part II Adrenal Gland

Table 9-1 Routine Laboratory Tests Useful in the


Evaluation of Adrenal Lesions
Frequency
of Usage

Cushing’s Syndrome

24-hour urine cortisol Common

Plasma ACTH and plasma cortisol Common

Low-dose dexamethasone suppression test Occasional

High-dose dexamethasone suppression Occasional


test

Metapyrone stimulation test Rare

Figure 9-1 CT scan of the abdomen demonstrating left Petrosal sinus ACTH measurement Extremely rare
adrenal lesion (arrow).
Hyperaldosteronism

Unprovoked hypokalemia Common

Plasma aldosterone level Common

Urinary aldosterone level Common

Aldosterone-to-renin ratio Common

Postural stimulation test Rare

Adrenal vein sampling of aldosterone Extremely rare

Pheochromocytoma

Plasma catecholamines Common

Urine catecholamines Common

Clonidine suppression test Occasional


Figure 9-2 MRI of the abdomen demonstrating left adrenal
lesion (arrow). Adrenal vein sampling of catecholamines Extremely rare

As mentioned previously, lesions of the adrenal gland


greater than 6 cm are worrisome for adrenal cancer. In
ALDOSTERONOMAS
one meta analysis, 105 of 114 adrenocortical carcinomas
measured 6 cm or greater in diameter.6 Because CT scan Primary hyperaldosteronism (Conn’s syndrome) is a rare
can underestimate the size of lesions by as much as 1 cm,7 etiology of hypertension (less than 1%). Other clinical
it is suggested that all lesions on CT scans that are 5 cm manifestations of Conn’s syndrome arise from increased
or greater be removed. In cases when there is concern for total body sodium content and a deficit in total body
adrenal carcinoma with local extension into adjacent potassium. Symptoms include urgency, frequency, noc-
organs, such as the kidney, colon, or spleen, then open turia, muscle weakness, paresthesias, or visual distur-
radical adrenalectomy with possible en bloc resection of bances.4,11 CT scan or MRI can detect adrenal adenomas
adjacent organs is the preferred approach.8,9 More as small as 1 cm. Laboratory manifestations include
recently, improvements in radiologic imaging tech- hypokalemia, elevated plasma and urinary aldosterone
niques, such as unenhanced and delayed enhanced CT level, elevated serum aldosterone-to-renin ratio, and sup-
with densitometry, chemical-shift MRI, and NP-59 pressed plasma renin activity.4,11 Once an important part
scintigraphy, have further assisted in differentiating of the evaluation, adrenal vein sampling is rarely used to
benign from malignant neoplasms.10 confirm and localize the lesion.
Chapter 9 Open and Laparoscopic Surgery of Adrenal Tumors 155

Once the diagnosis is confirmed, medical control of cytomas were treated through an open approach, with
hypertension and correction of hypokalemia should be early control of the adrenal vein. However, with increas-
instituted at least several weeks prior to adrenalectomy. ing experience worldwide with laparoscopic adrenalec-
The most effective medication for management of tomy, pheochromocytoma is no longer considered a
hyperaldosteronism is spironolactone, a competitive contraindication to laparoscopic surgery. In fact, laparo-
antagonist of the aldosterone receptor.4 Side effects of scopic adrenalectomy for pheochromocytomas has now
aldosterone include hyperkalemia, sexual dysfunction, been performed successfully at many centers of laparo-
gynecomastia, gastrointestinal disturbances, and meta- scopic excellence and reported in several series.14–16
bolic acidosis.12 Alternative medications include potas- Regardless of the surgical approach chosen, preopera-
sium-sparing diuretics, calcium channel blockers, and tive medical preparation is essential, and includes optimal
converting-enzyme inhibitors. 11 Hypertension is control of blood pressure with alpha blockade or calcium
improved or cured in more than 90% of patients fol- channel antagonists.2 Beta-blockers may be used to con-
lowing adrenalectomy.13 trol reflex tachycardia after initiation of alpha blockade.
In addition, aggressive fluid expansion is necessary to
increase circulating plasma volume and prevent postop-
CUSHING’S SYNDROME
erative hypotension. Close monitoring intraoperatively
Cushing’s syndrome is used to describe the symptom includes careful attention to blood pressure, central
complex that results from excess circulating glucocorti- venous pressure, and urinary output. An arterial line and
coids, regardless of etiology.3 Nonadrenal causes of central venous line are routinely used, and occasionally a
hypercortisolism include pituitary adenomas, ectopic Swan-Ganz catheter is employed. Severe hypertension
corticotrophin production, and exogenous steroid use. can be controlled with sodium nitroprusside or phento-
The urologist is most often confronted with an adrenal lamine, and hypotension can be controlled with fluid
lesion as the etiology of Cushing’s syndrome. resuscitation and norepinephrine.
Cushing’s syndrome manifests with a variety of well-
recognized clinical features, including hypertension,
ADRENOCORTICAL CARCINOMA
truncal obesity, moon facies, easy bruising, and mood dis-
orders. Diagnosis is confirmed by laboratory testing.3 Adrenocortical carcinoma is a rare malignancy with an
Hypercortisolism is best diagnosed by 24-hour urinary estimated incidence of 0.5 to 2.0 cases per million annu-
cortisol measurement. The low-dose dexamethasone ally.17,18 Although adrenal cancer is known to occur at all
suppression test can be used to further diagnose ages from early infancy to the 8th decade of life, there
Cushing’s syndrome if urinary cortisol measurement is appears to be a bimodal distribution during the first
equivocal. Abdominal CT scan and MRI can identify decade of life in children and in the fourth or fifth decade
adrenal adenomas or bilateral adrenal hyperplasia. of life in adults.19,20 The prognosis is uniformly poor,
although pediatric patients generally have a better prog-
nosis because they more commonly present earlier with a
PHEOCHROMOCYTOMA
hormonal syndrome.
Pheochromocytomas can be challenging tumors to treat The majority of adrenocortical carcinomas are func-
because of the unique manifestations of chronic and tional, with 62% of adrenal cancers being functional in
acute catecholamine excess. Successful surgical manage- one study.17 The most common syndrome associated with
ment of pheochromocytoma requires close collaboration adrenocortical carcinoma is Cushing’s syndrome. Typical
among the surgeon, endocrinologist, and anesthesiolo- clinical symptoms of adrenocortical carcinoma include
gist. It is essential to have an anesthesiologist familiar abdominal pain, weight loss, fever, weakness, anorexia,
with pheochromocytoma that is able to adequately man- nausea, and myalgia. Unfortunately, many of these symp-
age blood pressure intraoperatively and who is familiar toms are often associated with advanced disease.
with which anesthetic agents to avoid. In general, cate- The primary treatment modality for suspected adreno-
cholamine excess results in hypertension, tachycardia, and cortical carcinomas is complete surgical excision of the
a host of clinical manifestations. Laboratory diagnosis is tumor. Complete surgical excision appears to offer the
made by elevated levels of catecholamines in the blood and patient the best chance of cure. In one series, the 5-year
urine. Radiographic diagnosis is achieved with either CT survival of adrenocortical carcinoma after surgery was
scan or MRI. MRI imaging classically demonstrates a 55% following complete removal with negative margins
bright image on a T2-weighted study. Additionally, versus 5% for incomplete removal.21 When surrounding
metaiodobenzylguanidine (MIBG) nuclear medicine scan- organs, such as kidney, colon, or spleen, are infiltrated by
ning can help confirm and localize pheochromocytomas. adrenocortical carcinoma, then en bloc excision of sur-
The treatment of choice for most pheochromocy- rounding organs offers the patient the best chance of long-
tomas is surgical excision. In the past, all pheochromo- term survival. It is unclear whether the debulking of
156 Part II Adrenal Gland

tumors that cannot be excised completely offers a survival phrenic artery, aorta, and renal artery. A complex arcade
advantage. Radiation therapy and chemotherapy are of small arteries enters the adrenal gland from the medial
largely ineffective for metastatic disease.18 and superior border of the gland, and thus the anterior,
The suspected adrenocortical carcinoma should be sur- posterior, and inferolateral surfaces of the adrenal gland
gically approached in an open fashion rather than laparo- are relatively avascular.
scopic fashion. Because complete surgical excision of the The right and left adrenal glands have key anatomic
adrenocortical carcinoma portends a much better progno- differences in location and vasculature. The main right
sis, the ability to fully remove the lesion should not be adrenal vein exits the gland from the superomedial sur-
compromised solely for decreasing patient morbidity. The face and enters the inferior vena cava (IVC) directly. The
additional patient morbidity endured following an open longer left main adrenal vein exits the inferomedial
surgical procedure is insignificant if a more thorough exci- aspect of the gland and drains into the left renal vein at
sion is possible. The threshold to convert from a laparo- an oblique angle. The right adrenal gland is more inti-
scopic procedure to an open one should be low when there mately related to the IVC than the left gland is related to
is a clinical suspicion of a malignant adrenal neoplasm. the aorta. The capsule surrounding the adrenal gland is
Laparoscopic adrenalectomy for suspected malignant neo- very fragile, and direct grasping of the adrenal gland can
plasm should be performed only by those with great expe- lead to parenchymal fracture resulting in persistent and
rience in laparoscopic surgery, so that the surgeon can feel troublesome bleeding. The lymphatic drainage of the
that an equivalent or even more thorough excision can be adrenal gland includes all lateral aortic lymph node tissue
achieved by the laparoscopic approach. between the diaphragm and ipsilateral renal artery.

PREOPERATIVE PATIENT EVALUATION Open Surgical Approaches to the Adrenal Gland


AND PREPARATION
There are a variety of approaches to the adrenal gland,
Careful preoperative control and management of hor- including the flank, posterior, modified posterior, trans-
monally active tumors is critical prior to performing abdominal, and thoracoabdominal approaches. The
adrenal surgery, whether laparoscopic or open. choice of surgical approach is influenced by the tumor
Inadequate preoperative control of hormonally active pathology, size of the lesion, patient’s body habitus, and
lesions can lead to catastrophic intraoperative conse- surgeon familiarity and preference.
quences. Close collaboration with an endocrinologist and
anesthesiologist experienced with adrenal disorders is
Lateral Flank Approach
helpful. The urologist should have an understanding of
the physiology of adrenal disorders in order to appropri- The standard extrapleural, extraperitoneal lateral flank
ately manage patients in the peri- and postoperative approach offers excellent exposure to the adrenal gland.
period with regard to fluid management, electrolyte In addition, the lateral flank approach is familiar to most
abnormalities, and blood pressure control. Hormonally urologists. Generally, an incision above the 11th rib is
functional tumors must be adequately evaluated and utilized with or without resection of a rib. This approach
appropriate preoperative interventions initiated in con- is ideal for either left or right adrenalectomy.
cert with an endocrinologist. After adequate general anesthesia, a Foley catheter is
Preoperatively, all patients should receive a mechani- inserted to decompress the bladder and an orogastric
cal bowel preparation. Clear liquids should be started the tube to decompress the stomach. Lower extremity pneu-
day before surgery. A broad-spectrum antibiotic should matic compression stockings are placed. The patient is
be administered on call to the operating room. then placed in the full lateral position with the flank over
the flexion point of the operating table. The operating
table is flexed 30 to 45 degrees and the kidney rest is
OPEN SURGICAL TECHNIQUES
raised. An axillary roll is placed under the contralateral
Surgical Anatomy
arm. The patient is then carefully secured, and all pres-
A thorough knowledge of the anatomy of the adrenal sure points are adequately padded.
gland and its relationship to adjacent organs is essential A supracostal incision above the 11th rib is made extend-
to avoid intraoperative complications. Familiarity with ing from the lateral border of the rectus abdominus muscle
the vascular supply of the adrenal gland is important in anteriorly to the sacrospinalis muscle posteriorly. The exter-
minimizing the chances of intraoperative hemorrhage. nal oblique, internal oblique, latissimus dorsi, and serratus
The adrenal gland, like the kidney, is enveloped by posterior inferior muscles are incised. The lumbodorsal fas-
Gerota’s fascia; it is however located in a distinct fascial cia and transversus abdominus muscles are then incised to
compartment that is separate from the kidney. The arte- expose the peritoneum and preperitoneal fat. Care is taken
rial supply to the adrenal gland arises from the inferior to sweep away and avoid the peritoneum and the pleura.
Chapter 9 Open and Laparoscopic Surgery of Adrenal Tumors 157

Once the flank is entered, the peritoneum and colon kidney optimizes exposure to the superior aspect of the
are dissected away from Gerota’s fascia and the kidney is adrenal gland. The lateral attachments of the left adrenal
partially mobilized. It is important to avoid dissecting the gland are generally relatively avascular and are therefore
adrenal gland off of the surface of the kidney gland until easily mobilized. Posteriorly, the adrenal arteries that
mobilization of the adrenal gland is nearly complete. The arise from the aorta must be carefully ligated. Generally,
kidney is extremely useful for downward retraction of the the medial blood supply is taken last. Great care must be
adrenal gland. The capsule of the adrenal gland is quite taken to avoid injury to the renal artery and vein. Once
friable and should not be directly grasped in order to the left renal vein is identified, the left adrenal vein can
avoid fracturing the gland with resultant troublesome be safely divided. The adrenal gland is then dissected free
bleeding. In patients with pheochromocytoma, the adre- from the kidney and inspection should be made for
nal vein should be controlled early in the procedure to bleeding.
limit catecholamine surges. The anesthesiologist should Dissection on the right side (Figure 9-3) differs only
be notified when the adrenal vein is divided because slightly from the left side. Again, the kidney is useful for
occasionally a marked decrease in blood pressure can downward retraction of the adrenal gland. The superior
occur despite adequate fluid hydration. and lateral dissection of the adrenal gland should be per-
On the left side, the superior dissection of the adrenal formed initially, taking care to ligate the multiple small
gland is performed initially, taking care to clip or ligate adrenal vessels supplying the gland. The medial dissec-
the blood supply to the adrenal gland arising from the tion of the adrenal gland is next performed, carefully
inferior phrenic vessels. The adrenal gland is dissected dividing small arterial vessels. The IVC must be exposed
away from the diaphragm. Downward retraction on the to allow for exposure of the adrenal vein. The right adrenal

Figure 9-3 Flank approach to right adrenal.


158 Part II Adrenal Gland

vein is short and broad and great care must be under-


taken to prevent inadvertent avulsion. The right adrenal
vein is ligated. Lastly, the right adrenal gland is dissected
off of the superior surface of the kidney.
In the case of adrenocortical carcinoma, en bloc radi-
cal nephrectomy is often necessary. The kidney should be
removed, adhering to the general principles of oncologic
surgery, without detaching it from the adrenal gland. If
the tumor appears to be invading the colon, then subto-
tal colectomy may be required. Because the overall sur-
vival following complete surgical excision is improved,
then every effort should be made to remove all of tumor
if possible.
After the adrenal gland is removed, inspection for
hemostasis and injury to surrounding structures is per-
formed. The flank incision is then closed in the standard
fashion in layers.

Thoracoabdominal Approach
The thoracoabdominal approach (Figure 9-4), usually
through an incision above the 9th or 10th rib, is most
commonly used for very large tumors or suspected adre-
nal carcinomas. The incision is the same as the standard
approach for a radical nephrectomy, described elsewhere
in this text. The thoracoabdominal approach offers supe-
rior operative exposure and is particularly useful for large
adrenocortical carcinomas.
Figure 9-4 Thoracoabdominal approach to left adrenal.

Transabdominal Approach
The transabdominal approach (Figure 9-5) is useful when ful in pheochromocytomas, when tumors may be bilateral
exposure to both adrenal glands is necessary, particularly or may occur at sites distant from the adrenal gland.
in pediatric patients. This approach is occasionally chosen However, improved imaging techniques have allowed for
for patients with large adrenal tumors or pheochromocy- more precise localization of pheochromocytomas, thus
tomas. The transabdominal approach allows for excellent limiting the indications for this approach.
exposure to both adrenal glands and allows for full and The anterior abdominal approach is performed usually
complete abdominal exploration. This is particularly help- through a chevron incision, although a midline, subcostal,

Figure 9-5 Transabdominal approach to the adrenal.


Chapter 9 Open and Laparoscopic Surgery of Adrenal Tumors 159

or transverse incision may also be employed. Generally,


the type of incision for the anterior transabdominal
approach is dictated by the size of the lesion, presence of
bilateral lesions, patient body habitus, and surgeon pref-
erence.
The patient is placed in the supine position with the
kidney bar slightly elevated. A subcostal chevron incision
is made in the upper abdomen, and all of the muscles of
the anterior abdominal wall are divided. The peritoneum
is then entered and the abdominal organs inspected for
metastatic disease. The hepatic flexure must be reflected
for right-sided tumors and the splenic flexure for left-
sided tumors.
On the right side, the duodenum is reflected (Kocher
maneuver) to expose the IVC (Figure 9-5A). The kidney
is used to provide traction to the adrenal gland, and
direct grasping of the adrenal gland is minimized. The
superior attachments of the adrenal gland are divided
carefully, followed by the lateral adrenal gland attach-
ments. The medial surface of the adrenal gland is freed
by dividing arterial branches. The short right adrenal
vein is divided and the remaining attachments of the
adrenal gland are divided to release the specimen. On the
left side, after identification of the adrenal gland and kid-
ney, the superior attachments of the gland are divided
first. The spleen and pancreas often must be dissected
bluntly away from the adrenal gland (Figure 9-5B). The
left adrenal vein is divided during the medial dissection of
the gland and the adrenal gland is then freed from the
superior surface of the kidney.

Posterior Approach Figure 9-6 Posterior approach to the adrenal.

The posterior approach (Figure 9-6) is rarely used


today because of improvements in diagnostic imaging. the kidney is exposed. Inferior retraction on the kidney
In the past, the posterior approach was used for bilateral allows for visualization of the adrenal gland on the
adrenal exploration or for removal of small adrenal superior surface of the kidney. Similar to the flank
lesions. The posterior approach is performed through a approach, the superior vessels supplying the adrenal
subcostal incision and provides a limited surgical field. gland are divided, the adrenal vein is ligated, and the
It does provide for direct access to the adrenal gland adrenal gland is circumferentially mobilized. Again,
and is ideal for thin patients. The limited surgical field care must be taken to ensure that the renal hilar vessels
makes this unsuitable for excision of large adrenal are not injured during the dissection.
masses and lesions that are suspicious for adrenocortical
carcinoma. With the advent of laparoscopic surgery,
Modified Posterior Approach for Right
there are no specific indications for the posterior
Adrenalectomy
approach. Most lesions that are small enough to be per-
formed through a posterior approach should be A modified posterior approach (Figure 9-7) for accessing
removed laparoscopically. the right adrenal gland has been described.22 The patient
The patient is placed in the prone position with the is placed in a modified prone position, and the 11th or
table slightly flexed. The incision is placed at the level 12th rib is resected. The primary advantage of this
of the 11th or 12th rib. The incision is carried through approach is that the adrenal vein is identified more easily
the lumbodorsal fascia and the latissimus dorsi muscles. because it is seen emerging from the IVC toward the
The pleura and peritoneum are swept away from the operating surgeon. Again, however, this approach is
11th and 12th ribs until Gerota’s fascia is identified. rarely used because most such tumors are removed by the
Gerota’s fascia is incised, and the posterior surface of laparoscopic technique.
160 Part II Adrenal Gland

Figure 9-7 Modified posterior approach to the right adrenal.

LAPAROSCOPIC SURGERY
lectomy in the past, it is clear that the procedure can be
OF THE ADRENAL GLAND
performed safely as long as the same precautions are
Indications for Laparoscopic Adrenalectomy
taken as those for open surgery.14 The current indica-
The indications for laparoscopic adrenalectomy have tions for performing a laparoscopic adrenalectomy are
expanded as more surgeons have become proficient with listed in Table 9-2.
the technique and the advantages of this approach have There are few absolute contraindications to laparo-
become apparent. Laparoscopic adrenalectomy has, in scopic adrenalectomy. It is generally felt that a known or
many centers, become the surgical procedure of choice suspected primary adrenal carcinoma, particularly with
for the management of functional tumors less than 6 cm extension into surrounding organs, should be removed
in size. Although the presence of pheochromocytoma by an open technique. Given the aggressive nature of the
was a relative contraindication for laparoscopic adrena- disease, the open approach allows for en bloc resection
Chapter 9 Open and Laparoscopic Surgery of Adrenal Tumors 161

Table 9-2 Indications for Laparosocpic determining the approach. Most surgeons are familiar
Adrenalectomy with the anterior transperitoneal approach, but many
who have overcome the learning curve of the anterior
Aldosterone secreting adrenal gland, adenoma, or
unilateral hyperplasia transperitoneal approach are becoming skilled with the
retroperitoneal approach.25–30 Although each approach
Cushing’s syndrome secondary to adrenocortical adenoma has purported advantages and disadvantages, there is no
clear-cut evidence that one is superior.31,32
Non-functional adrenal mass ≤ 8 cm with negative
metastatic workup
Preoperative Preparation
Non-functional adrenal mass ≤ 8 cm with progressive
growth on CT or MRI The preoperative evaluation and preparation of patients
prior to undergoing laparoscopic adrenalectomy is iden-
Adrenal pheochromocytoma (benign) ≤ 8 cm tical to that prior to open adrenalectomy. All patients
should undergo a mechanical bowel preparation on the
Solitary adrenal gland metastasis
day prior to surgery and a broad-spectrum antibiotic
should be administered on the day of surgery. As with all
laparoscopic procedures, the stomach should be decom-
and potential removal of surrounding organs.8 Other pressed with an orogastric tube and the bladder with a
contraindications to laparoscopic adrenalectomy include Foley catheter.
uncorrectable coagulopathy and cardiopulmonary disease
precluding general anesthesia. Patients who will not tol-
Anterior Transperitoneal Approach for Right
erate an open operation are generally poor candidates for
Adrenalectomy
laparoscopic adrenalectomy.
Relative contraindications to laparoscopic adrenalec- After general anesthetic by agents other than nitrous
tomy include previous abdominal surgery or significant oxide, the patient is positioned with the right side ele-
morbidity. Lesions greater than 8 cm in size, even if not vated 45 to 70 degrees upward and the table slightly
suspected to be primary adrenal carcinomas, should be flexed at the level of the umbilicus. The patient should be
approached cautiously because of the increased risk of positioned on a beanbag with extensive padding over
hemorrhage and injury to surrounding viscera. With pressure points. Figure 9-8 shows the general modified
increasing experience in performing laparoscopic flank position used for laparoscopic adrenalectomy. Next,
adrenalectomy, relative contraindications become less the patient should be secured with tape to allow the table
of a factor. A variety of approaches to laparoscopic to be tilted side to side so as to facilitate exposure.
adrenalectomy, including transperitoneal and retroperi- For a right laparoscopic adrenalectomy, four subcostal
toneal, have further decreased some relative contraindi- ports are used and placed two to three fingerbreadths
cations. below the costal margin, as depicted in Figure 9-9. Initial
Occasionally, the urologist will encounter a patient
with a suspected solitary metastatic lesion to the adre-
nal gland. If the lesion is less than 6 cm in size and not
obviously adherent to surrounding viscera, then a
laparoscopic approach is reasonable.23 The surgeon
should be skilled in laparoscopic adrenalectomy before
attempting to remove a solitary metastatic lesion given
the more difficult surgical planes that are often present.

Laparoscopic Surgical Approaches


Perhaps no other urologic laparoscopic procedure has as
many different surgical approaches as does adrenalec-
tomy. Commonly used approaches to the adrenal gland
include the transperitoneal approaches, and the posterior
or lateral retroperitoneal approaches. Recently, a
transthoracic approach has been described for patients
who have undergone extensive previous transperitoneal
and retroperitoneal surgery.24 Surgeon preference and Figure 9-8 Patient positioning for right transperitoneal
experience appear to be the most important factors in adrenalectomy.
162 Part II Adrenal Gland

Figure 9-9 Port placement for right transperitoneal adrenalectomy.

entry into the peritoneal cavity is made using the Veress peritoneum and extension through the triangular liga-
needle just below the costal margin in the midclavicular ments of the liver allow for upward and medial retraction
line. Three or four additional ports are placed under of the liver (Figure 9-10). A fixed retractor may be placed
direct vision; the most medial port is important for under the liver through an additional trocar port to allow
upward and medial retraction of the right lobe of the the liver to remain out of the field of dissection for the
liver. Exposure of the right adrenal gland is dependent on remainder of the case.
adequate mobilization of the liver. The IVC is eventually identified once there is ade-
Mobilization of the liver is the first step in exposing quate liver mobilization. Continued and careful dissec-
the right adrenal gland. Unlike laparoscopic nephrec- tion along the lateral surface of the IVC will reveal the
tomy, full mobilization of the ascending colon and right adrenal vein. The adrenal vein should then be
hepatic flexure is unnecessary. Incision of the posterior divided between standard clips. Dissection is further
Chapter 9 Open and Laparoscopic Surgery of Adrenal Tumors 163

Figure 9-10 T-shaped incision through the posterior


peritoneum for left and right adrenalectomy. On right, incision Figure 9-11 Dissection of left adrenal gland allowing
from second part of the duodenum to triangular ligaments at visualization of left adrenal vein.
liver edge and then lateral to hepatic flexure. On left, incision
developed across phrenocolic and splenocolic ligaments
and at inferior border of spleen (IVC, inferior vena cava; image as for a right adrenalectomy. The important sur-
R, right; L, left). rounding structures to identify when performing a
transperitoneal left laparoscopic adrenalectomy include
continued towards the diaphragm, and the inferior the spleen, tail of the pancreas, splenic flexure of the
phrenic vessels should next be identified and divided. colon, and left kidney. Full mobilization of the splenic
The inferior pedicle of the adrenal gland is then flexure of the colon is necessary to provide adequate
released, separating the adrenal gland from the upper exposure to the left adrenal gland.
pole of the kidney. Gerota’s fascia is next incised at the The first step after diagnostic laparoscopy is to incise
junction of the upper pole of the kidney and the adrenal the posterior peritoneum along the line of Toldt and
gland. There is often an arterial branch to the adrenal mobilize the splenic flexure of the colon to allow the
gland arising from the renal pedicle. Once the kidney is colon to fall medially. The splenocolic and lienorenal lig-
completely mobilized away from the adrenal gland, all aments are then mobilized to allow the spleen to be safely
that remains holding the adrenal gland in place is the rel- separated from the field of dissection (Figure 9-10). This
atively avascular lateral attachments, which are divided. creates an adequate plane between the spleen and the
Use of the harmonic scalpel can facilitate mobilization of upper pole of the left kidney. If necessary, the tail of the
the adrenal gland once the main vascular pedicles have pancreas can be separated away from Gerota’s fascia to
been ligated. allow the pancreas to fall away with the spleen to provide
Once the adrenal gland is completely separated, it more exposure.
should be placed in a specimen retrieval bag and removed Next, Gerota’s fascia is incised between the upper pole
en bloc. Assuming adequate hemostasis, the laparoscopic of the left kidney and the adrenal gland. The left adrenal
ports are removed under direct vision and fascia closed gland should not be grasped directly to avoid adrenal
with the Carter–Thomason fascial closure device. gland fractures, which are associated with troublesome
A drain is usually not necessary. The orogastric tube is bleeding. Dissection continues through the perirenal
removed at the conclusion of the procedure. fibrofatty tissue. The inferior border of the adrenal gland
is defined and the dissection continued medially. Medial
dissection will eventually lead to the takeoff of the left
Anterior Transperitoneal Approach
adrenal vein emanating directly from the left renal vein
for Left Adrenalectomy
(Figure 9-11). The left adrenal vein is then isolated,
The patient is positioned with left side elevated 45 to 70 clipped, and divided. In the case of a pheochromocytoma,
degrees with the table slightly flexed at the level of the early exposure and ligation of the left adrenal vein is ideal
umbilicus. Three or four trocars are placed in a mirror to reduce the risk of a hypertensive crisis. We have found
164 Part II Adrenal Gland

it easiest to initially identify the left renal vein and deter- lumbodorsal fascia is incised, access into the retroperi-
mine the take-off of the left adrenal vein. Clipping of the toneal space is confirmed by inserting one finger in and
adrenal vein at this juncture minimizes catecholamine palpating the 12th rib, the iliac crest, and the psoas mus-
surges. cle. The trocar-mounted balloon is then inserted and
Again, the lateral attachments of the left adrenal gland inflated in order to mobilize Gerota’s fascia anteriorly
should be saved until the remainder of the gland is mobi- away from the posterior abdominal wall. Carbon dioxide
lized. The superior aspect of the adrenal gland is then insufflation is then attached to the trocar to generate
mobilized, taking care to divide the phrenic vessels sup- pneumoretroperitoneum at a pressure of 15 mm Hg.
plying the gland. Once the superior and inferior borders Once the primary port is inserted, the working ports are
of the gland are dissected adequately, attention is placed. The second port is placed posterior to the pri-
directed towards the head of the gland, which is adjacent mary port, below the angle formed by the 12th rib and
to the aorta. The left adrenal vein is divided if not previ- the paraspinous muscles. The third port is placed approx-
ously done. The left adrenal artery arising from the aorta imately 3 to 4 cm medial to the primary port in the ante-
is next divided. The adrenal gland has a highly variable rior axillary line. The working ports are either 5 or 10 mm
vasculature, especially in larger lesions with increased in size. The optional fourth port is placed in the anterior
blood supply. The use of a harmonic scalpel or hook axillary line 5 to 7 cm inferior to the third port and may
cautery electrode can facilitate adrenal gland mobiliza- be used to assist with retraction throughout the duration
tion and adequately ligate small blood vessels supplying of the case.
the gland. Once pneumoretroperitoneum is established, identi-
Lastly, the lateral attachments of the adrenal gland are fication of key landmarks helps to establish the orienta-
divided to fully free the gland from all surrounding tis- tion. The psoas muscle is usually easily seen and
sues. The specimen is then placed into a retrieval bag and establishes longitudinal orientation. By retracting the
removed intact. Closure is similar to that for the right kidney upwards and anteriorly, subsequent medial dis-
adrenalectomy. section will eventually reveal the great vessels running
parallel to the psoas muscle. The renal artery can be
identified by identifying pulsations, although full
Retroperitoneal Technique
mobilization of the renal hilar vessels is generally not
The retroperitoneal approach to the adrenal gland is par- necessary during adrenalectomy. On occasion intraop-
ticularly useful in the patient with a prior history of erative ultrasonography may be helpful in identifying a
extensive abdominal surgery. Unlike the transperitoneal small adrenal lesion in the midst of abundant per-
approach, the patient is positioned in the full flank posi- inephric fat.
tion. Three or four ports are typically used. Patient posi-
tioning and port placement are as shown in Figure 9-12.
Left Adrenalectomy
Access into the retroperitoneum requires the creation of
a working space using a dilating balloon (U.S. Surgical, Dissection on the left side should be conducted along the
Norwalk, CT). psoas muscle to the upper pole of the kidney.
A Hasson technique is used to place the first port. Identification of the renal hilum allows for rapid identifi-
A small 2-cm incision is made just below the tip of the cation of the left adrenal vein. The adrenal vein can be
12th rib, and S-type retractors are used to split the mus- found along the inferomedial border of the adrenal
cles until the lumbodorsal fascia is identified. Once the gland. If difficulty is encountered identifying the left
adrenal vein, the adrenal vein can be found by first locat-
ing the left renal vessels and noting the junction of the
left adrenal vein with the left renal vein. Unlike a
transperitoneal laparoscopic adrenalectomy, the dissec-
tion and identification of the left adrenal vein must be
done from a posterior approach. Once the left adrenal
vein is divided, the remainder of the adrenal gland can be
detached, again trying to minimize direct grasping of the
friable adrenal tissue. Generally, the lateral and inferior
surface of the gland is dissected away from the kidney.
The superior margin is dissected free by dividing the
inferior phrenic vessels. The use of the harmonic scalpel
Figure 9-12 Patient positioning and trocar placement for is useful for complete mobilization of the adrenal gland.
retroperitoneal adrenalectomy (A = camera port; B and C = Once detached, the adrenal gland is placed in a speci-
working ports). men retrieval bag and then removed through the primary
Chapter 9 Open and Laparoscopic Surgery of Adrenal Tumors 165

port, the largest incision. The remaining ports are then and the retroperitoneum entered. The adrenal gland is
closed in the standard fashion. then identified and dissected free. Once the adrenal
gland is removed, the diaphragm is repaired. A chest tube
is kept in place at the conclusion of the procedure.
Right Adrenalectomy
The right adrenalectomy proceeds using the same prin-
Postoperative Care
ciples of retroperitoneal laparoscopy. The dissection pro-
ceeds along the psoas muscle in a superior direction. The The advantages of the laparoscopic approach to the adre-
kidney may be held anteriorly by an optional retractor. nal gland are immediately apparent in the postoperative
The right adrenal gland is situated more medial to the period. The orogastric tube is removed immediately at
kidney than the left adrenal gland, and as such the upper the end of the operation and the Foley catheter removed
pole of the kidney may interfere with exposure of the as soon as the patient is ambulatory. Postoperative pain is
adrenal gland. The IVC is identified medial to the psoas. controlled with parenteral narcotics in the first 24 hours,
Along the posterolateral aspect of the IVC, the main ketorolac or oral narcotics thereafter. Supplemental cor-
adrenal vein is identified. Caution must be exercised ticosteroids and appropriate antihypertensive medica-
when manipulating the main adrenal vein to prevent tions are administered as needed depending on the type
inadvertent avulsion. The adrenal vein is then clipped of tumor removed. Postoperative care can be coordinated
and divided. in concert with an endocrinologist if necessary.
Once the adrenal vein is divided, then inferior and Discharge is usually within 24 to 48 hours from surgery
medial borders of the adrenal gland are dissected free and full recovery requires 10 to 14 days.
from the right renal vein and the IVC. Small adrenal ves-
sels can then be effectively ligated using the harmonic
Complications of Laparoscopic Adrenalectomy
scalpel. The inferior phrenic vessels are then divided to
release the adrenal gland and the specimen removed in The most significant intraoperative complication is hem-
the usual fashion. orrhage. The adrenal gland is highly vascular which can
result in troublesome bleeding if not adequately con-
trolled.35 The use of a harmonic scalpel during dissection
Posterior Retroperitoneal Approach
of the adrenal gland can limit the amount of hemorrhage.
The posterior retroperitoneal approach to the adrenal In addition, the adrenal gland itself is very easily frac-
gland has been described.33 Purported advantages of this tured, often resulting in bleeding.
approach include less risk of injury to organs confined to Other intraoperative complications from laparoscopic
the peritoneal or retroperitoneal cavity and is better adrenalectomy are similar to those for any laparo-
suited for smaller tumors. With this technique, the oper- scopic procedure and can include injuries to the colon,
ation is performed with the patient in the prone position. small bowel, liver, gallbladder, pancreas, spleen, and
A 3 to 4-port technique is used following standard bal- diaphragm.35 In general, major complications occur less
loon dilatation. Gerota’s fascia is incised along the medial often as surgeon experience increases. Conversion to an
crus of the diaphragm. The medial surface of the adrenal open case should be done if hemorrhage is uncontrol-
gland, including the adrenal vessels, is exposed first lable or intraoperative injury cannot be repaired through
before mobilization of the entire gland. Once the main a laparoscopic approach.
vascular supply of the adrenal gland is divided, the
remainder of the gland is dissected free. Although results
Results of Laparoscopic Adrenalectomy
with this technique are promising,34 experience with this
approach is limited. Worldwide experience with laparoscopic adrenal surgery
has increased since its original introduction in 1992.
Several centers have now reported large series in the lit-
Transthoracic Technique
erature documenting the decreased blood loss, shortened
Recently, the technique of thorascopic transdiaphrag- hospital stay, and faster return to normal activity.
matic adrenalectomy has been described.24 This tech- Selected recent series in the literature are summarized in
nique has potential for use when both the transperitoneal Table 9-3.
and retroperitoneal spaces have been violated by prior Gagner et al.46 reported on 100 consecutive laparo-
surgery. Following double lumen endotracheal intuba- scopic adrenalectomy procedures performed through the
tion, the patient is placed in the prone position. A 4-port transperitoneal approach. The mean operative time was
transthoracic technique is used. 123 minutes with an estimated blood loss of 70 cc. In his
In order to gain exposure to the adrenal gland, the series, the open conversion rate was 3%. Average length
diaphragm is incised under ultrasonographic guidance of hospital stay was 3 or less days, and morbidity was
Table 9-3 Selected Laparoscopic Adrenalectomy Series
166

Number OR Time EBL Hospital Conversion


Reference of Cases Age Approach (minutes) (cc) Stay (days) Rate Complications

MacGillivray36 (2002) 60 — Transperitoneal 183 63 2 0/60

Valeri37 (2002) 91 — Transperitoneal 92–148 — 3.5 2/91 2 postoperative hemorrhage, 1 port site bleed
1 UTI, 1 death from myocardial infarction

Kebebew et al.38 (2001) 176 — Transperitoneal 168 — 1.7 0/176 5.1%


Part II Adrenal Gland

Lezoche et al.31 (2001) 216 45.9 149 transperitoneal 100 — — 4/216 1 death, 1 hemoperitoneum, 1
67 retroperitoneal wound infection

Salamon et al.30 (2001) 115 49.3 115 retroperitoneal 118 77 4 1/118 3.5% intraoperative
12.1% postoperative

Guazzoni et al.39 (2001) 161 39.4 Transperitoneal 160 — 2.8 4/161 5.5%

Suzuki et al.32 (2001) 118 51.7 78 transperitoneal 171 96.3 — 6/118 2 paralytic ileus
40 retroperitoneal 4 shoulder tip pain

Soulie et al.40 (2000) 52 46.9 52 retroperitoneal 135 80 5 1/52 5.7% intraoperative


11.5% postoperative

Mancini et al.41 (1999) 172 — Transperitoneal 132 — 5.8 12/172 8.7%, 2 deaths

Schichman et al.42 (1999) 50 54 Transperitoneal 219 142 3 0/50 10%

Winfield et al.43 (1998) 21 52.2 Transperitoneal 219 183 2.7 0/21 1 subcutaneous bleed
2 pneumothorax
1 pulmonary edema

Yoshimura et al.44 (1998) 28 42 11 transperitoneal 375 370 2.7 0/28 4 blood transfusion
17 retroperitoneal 4 subcutaneous emphysema
2 postoperative bleeding

Chee et al.45 (1998) 14 46.2 8 transperitoneal 135 Min 3 0/14 1 pneumonia


6 retroperitoneal

Gagner et al.46 (1997) 100 46 Transperitoneal 123 70 3 3/100 12%, 3 DVT, 2 pulmonary embolus

Gasman et al.29 (1997) 23 49.6 23 retroperitoneal 97 70 3.3 0/23 1 postop hematoma

Terachi et al.47 (1997) 100 — Transperitoneal 240 77 — 3/100 —

Rutherford et al.48 (1996) 67 54 Transperitoneal 124 — 5.1 0/67 3 DVT, 2 pulmonary emboli 1 port
site hernia, 1 postoperative bleed

Average 47.1 153.5 98.6 36/1567 (2.3%)


Chapter 9 Open and Laparoscopic Surgery of Adrenal Tumors 167

encountered in 12% of patients. The lesions removed were alive with a mean follow-up of 8.3 months. These
included pheochromocytomas, aldosteronomas, Cushing’s data suggest that the laparoscopic approach to some
lesions, and others. malignant neoplasms either originating from or metas-
In the largest published series identified in the litera- tasizing to the adrenal gland is reasonable, but the con-
ture by Lezoche et al.,31 a total of 216 laparoscopic version to an open procedure should be performed if
adrenalectomies were performed through the anterior local invasion is present.
transperitoneal, lateral transperitoneal, and the posterior
retroperitoneal approaches. The study was a combined
PARTIAL ADRENALECTOMY
experience of surgeons in Italy and The Netherlands.
The average operating time of all approaches was 100 Adrenal sparing surgery for benign tumors, particularly
minutes with a conversion rate of only 1.9%. Average aldosterone producing lesions, has been reported using
hospital stay for all approaches was 3 to 4 days. both the open and laparoscopic techniques. However, the
Comparison studies have been made between laparo- indications and techniques are unclear. Nakada et al.57
scopic and open adrenalectomy to determine if there are performed open partial adrenalectomy on 26 patients
significant benefits in the laparoscopic approach.43,44,48–54 with primary hyperaldosteronism and noted no cases of
In general, the operative times for laparoscopic surgery recurrent disease at 5-year follow-up. Partial adrenalec-
are longer than for the open technique, particularly early tomy has also been performed for patients with bilateral
in the learning curve. However, the operative times pheochromocytomas and the rare patient with the soli-
decrease as surgeon experience increases. In addition, the tary adrenal gland.
laparoscopic approach offers less blood loss, significantly Laparoscopic partial adrenalectomy has also been
less postoperative narcotic use, overall shorter hospital described. Keschke et al.58 recently reported on 13
stay, and a faster return to normal activity. The cost of a patients with primary hyperaldosteronism and a radi-
laparoscopic adrenalectomy was shown to be comparable ographically proven adrenal adenoma who underwent
to that of an open adrenalectomy in one study.55 laparoscopic partial adrenalectomy. Exclusion criteria
included tumors greater than 3 cm in size or centrally
located tumors. Laparoscopic partial adrenalectomy has
Laparoscopic Adrenalectomy for Malignant Tumors
been reported for pheochromocytoma59 and may be use-
Increased surgeon experience and comfort with laparo- ful in patients with von Hippel-Lindau disease.
scopic adrenalectomies has led to performing laparoscopic Laparoscopic instruments useful in performing partial
adrenalectomies for larger and potentially malignant adrenalectomy include harmonic scissors, bipolar coagu-
tumors. Henry et al.56 performed laparoscopic adrenalec- lating forceps, and intraoperative ultrasound.
tomies on 19 patients with potentially malignant tumors, Although the indications for open and laparoscopic
all of which were greater than 6 cm in size. Median oper- partial adrenalectomy are unclear at this time, there are
ating time was 150 minutes, and conversion was neces- patients for whom adrenal sparing surgery should be
sary in two patients because of intraoperative evidence of considered. Possible indications for partial adrenalec-
invasive carcinoma. Six of the 19 patients had an adreno- tomy include patients with solitary adrenal glands,
cortical carcinoma on pathologic diagnosis. One of these aldosteronomas less than 3 cm in size, bilateral pheo-
patients presented with a liver metastasis 6 months after chromocytomas, and patients at risk for multiple adrenal
surgery and died shortly after. The other 5 patients are tumors due to a hereditary syndrome.
alive with a follow-up ranging from 8 to 83 months. The
authors concluded that laparoscopic adrenalectomy for
carcinoma can be performed on select patients in experi- REFERENCES
enced hands; however, conversion to open radical
adrenalectomy should be performed if there is evidence 1. Gagner M, Lacroix A, Bolte E: Laparoscopic
of local tissue invasion observed during surgery. adrenalectomy in Cushing’s syndrome and
Laparoscopic adrenalectomy has also been safely pheochromocytoma. North Engl J Med 1992; 327:1033.
performed in patients with solitary adrenal metastases. 2. Walther MM, Keiser HR, Linehan WM:
In a recent series by Heniford et al.,23 laparoscopic Pheochromocytoma: evaluation, diagnosis, and treatment.
World J Urol 1999; 17:35–39.
adrenalectomy was performed in 11 patients, 10 of
3. Goldfarb DA: Contemporary evaluation and management
whom had the adrenalectomy performed for metastatic of Cushing’s syndrome. World J Urol 1999; 17:22–25.
disease. Average operative time was 181 minutes, and 4. Blumenfeld JD, Vaughan ED Jr: Diagnosis and treatment
blood loss was minimal at 138 cc. One patient required of primary aldosteronism. World J Urol 1999; 17:15–21.
conversion to an open approach due to local invasion of 5. Murai M, Baba S, Nakashima J, et al: Management of
the tumor into the lateral wall of the vena cava, which incidentally discovered adrenal masses. World J Urol
was removed with the specimen. Ten of the 11 patients 1999; 17:9–14.
168 Part II Adrenal Gland

6. Belldegrun A, Hussain S, Seltzer S, et al: Incidentally 26. Suzuki K: Laparoscopic adrenalectomy: retroperitoneal
discovered mass of the adrenal gland. Surg Gynecol approach. Urol Clin North Am 2001; 28:85–95.
Obstet 1986; 163:203–208. 27. Soulie M, Mouly P, Caron P, et al: Retroperitoneal
7. Cerfolio RJ, Vaughan ED Jr, Brennan TG Jr, et al: laparoscopic adrenalectomy: clinical experience in 52
Accuracy of computed tomography in predicting procedures. Urology 2000; 56:921–925.
adrenal tumor size. Surg Gynecol Obstet 1993; 28. Baba S, Ito K, Yanaihara H, et al: Retroperitoneoscopic
176:307–309. adrenalectomy by a lumbodorsal approach: clinical
8. Schulick RD, Brenna MF: Adrenocortical carcinoma. experience with solo surgery. World J Urol 1999;
World J Urol 1999; 17:26–34. 17:54–58.
9. Vaughan ED Jr: Surgical options for open adrenalectomy. 29. Gasman D, Droupy S, Koutani A, et al: Laparoscopic
World J Urol 1999; 17:40–47. adrenalectomy: the retroperitoneal approach. J Urol 1998;
10. Teeger S, Papanicolaou N, Vaughan ED Jr: Current 159:1816–1820.
concepts in imaging of adrenal masses. World J Urol 30. Salomon L, Soulie M, Mouly P, et al: Experience with
1999; 17:3–8. retroperitoneal laparoscopic adrenalectomy in 115
11. Ferriss JB, Beevers DG, Brown JJ, et al: Clinical, procedures. J Urol 2001; 166:38–41.
biochemical and pathological features of low-renin 31. Lezoche E, Guerrieri M, Feliciotti F, et al: Anterior,
(“primary”) hyperaldosteronism. Am Heart J 1978; lateral, and posterior retroperitoneal approaches in
95:375–388. endoscopic adrenalectomy. Surg Endosc 2002;
12. deGasparo M, Whitebread SE, Preiswerk G, et al: 16:96–99.
Antialdosterones: incidence and prevention of sexual side 32. Suzuki K, Kageyama S, Hirano Y, et al: Comparison of
effects. J Steroid Biochem 1989; 32(1B):223–227. 3 surgical approaches to laparoscopic adrenalectomy: a
13. Blumenfeld JD, Sealey JE, Schlussel Y, et al: Diagnosis nonrandomized, background matched analysis. J Urol
and treatment of primary aldosteronism. Ann Intern Med 2001; 166:437–443.
1994; 121:877–885. 33. Nakagawa K, Murai M, Deguchi N, et al: Laparoscopic
14. Edwin B, Kazaryan AM, Mala T, et al: Laparoscopic and adrenalectomy: clinical results in 25 patients. J Endourol
open surgery for pheochromocytoma. BMC Surg 2001; 1995; 9:265–267.
1:2. 34. Baba S, Miyajima A, Uchida A, et al: A posterior lumbar
15. Salomon L, Rabii R, Soulie M, et al: Experience with approach for retroperitoneoscopic adrenalectomy:
retroperitoneal laparoscopic adrenalectomy for assessment of surgical efficacy. Urology 1997;
pheochromocytoma. J Urol 2001; 165:1871–1874. 50:19–24.
16. Gotoh M, Ono Y, Hattori R, et al: Laparoscopic 35. Henry JF, Defechereux T, Raffaelli M, et al:
adrenalectomy for pheochromocytoma: morbidity Complications of laparoscopic adrenalectomy: results of
compared with adrenalectomy for tumors of other 169 consecutive procedures. World J Surg 2000;
pathology. J Endourol 2002; 16:245–249. 24:1342–1346.
17. Ng L, Libertino JM: Adrenocortical carcinoma: diagnosis, 36. MacGillivray DC, Whalen GF, Malchoff CD, et al:
evaluation, and treatment. J Urol 2003; 169:5–11. Laparoscopic resection of large adrenal tumors. Ann Surg
18. Wajchenberg BL, Albergaria Pereira MA, Medonca BB, Oncol 2002; 9:480–485.
et al: Adrenocortical carcinoma: clinical and laboratory 37. Valeri A, Borrelli A, Presenti L, et al: The influence of
observations. Cancer 2000; 88:711–736. new technologies on laparoscopic adrenalectomy. Surg
19. Stratakis CA, Chrousos GP: Adrenal cancer. Endocrinol Endosc 2002; 16:1274–1279.
Metab Clin North Am 2000; 29:15–25. 38. Kebebew E, Siperstein AE, Duh QY: Laparoscopic
20. Liou LS, Kay R: Adrenocortical carcinoma in children: adrenalectomy: the optimal surgical approach. J
review and recent innovations. Urol Clin North Am 2000; Laparoendosc Adv Surg Tech A 2001; 11:409–413.
27:403–421. 39. Guazzoni G, Cestari A, Montorsi F, et al: Eight-year
21. Schulick RD, Brennan MF: Long-term surival after experience with transperitoneal laparoscopic adrenal
complete resection and repeat resection in patients with surgery. J Urol 2001; 166:820–824.
adrenocortical carcinoma. Ann Surg Oncol 1999; 40. Soulie M, Mouly P, Caron P, et al: Retroperitoneal
6:719–726. laparoscopic adrenalectomy: clinical experience in 52
22. Vaughan ED Jr, Phillips H: Modified posterior approach procedures. Urology 2000; 56:921–925.
for right adrenalectomy. Surg Gynecol Obstet 1987; 41. Mancini F, Mutter D, Peix JL, et al: Experience with
165:453–455. adrenalectomy in 1997. Apropos of 247 cases: a multicenter
23. Heniford BT, Arca MJ, Walsh RM, et al: Laparoscopic prospective study of the French-speaking Association of
adrenalectomy for cancer. Semin Surg Oncol 1999; Endocrine Surgery. Chirurgie 1999; 124:368–374.
16:293–306. 42. Shichman SJ, Herndon CD, Sosa RE, et al: Lateral
24. Gill IS, Meraney AM, Thomas JC, et al: Thoracoscopic transperitoneal laparoscopic adrenalectomy. World J Urol
transdiaphragmatic adrenalectomy: the initial experience. 1999; 17:48–53.
J Urol 2001; 165:1875–1881. 43. Winfield HN, Hamilton BD, Bravo EL, et al:
25. Bonjer HJ, Sorm V, Berends FJ, et al: Endoscopic Laparoscopic adrenalectomy: the preferred choice? A
retroperitoneal adrenalectomy: lessons learned from 111 comparison to open adrenalectomy. J Urol 1998;
consecutive cases. Ann Surg 2000; 232:796–803. 160:325–329.
Chapter 9 Open and Laparoscopic Surgery of Adrenal Tumors 169

44. Yoshimura K, Yoshioka T, Miyake O, et al: Comparison of 53. MacGillivray DC, Schichman SJ, Ferrer FA, et al: A
clinical outcomes of laparoscopic and conventional open comparison of open vs laparoscopic adrenalectomy. Surg
adrenalectomy. J Endourol 1998; 12:555–559. Endosc 1996; 10:987–990.
45. Chee C, Ravinthiran T, Cheng C: Laparoscopic 54. Miccoli P, Raffaelli M, Berti P, et al: Adrenal surgery
adrenalectomy: experience with transabdominal and before and after the introduction of laparoscopic
retroperitoneal approaches. Urology 1998; 51:29–32. adrenalectomy. Br J Surg 2002; 89:779–782.
46. Gagner M, Pomp A, Heniford BT, et al: Laparoscopic 55. Ortega J, Sala C, Garcia S, et al: Cost-effectiveness of
adrenalectomy: lessons learned from 100 consecutive laparoscopic vs open adrenalectomy: .small savings in an
procedures. Ann Surg 1997; 226:238–246. expensive process. J Laparoendosc Adv Surg Tech A 2002;
47. Terachi T, Matsuda T, Terai A, et al: Transperitoneal 12:1–5.
laparoscopic adrenalectomy: experience in 100 patients. 56. Henry JF, Sebag F, Iacobone M, et al: Results
J Endourol 997; 11:361–365. of laparoscopic adrenalectomy for large and
48. Rutherford JC, Stowasser M, Tunny TJ, et al: potentially malignant tumors. World J Surg 2002;
Laparoscopic adrenalectomy. World J Surg 1996; 26:1043–1047.
20:758–760. 57. Nakada T, Kubota Y, Sasagawa I, et al: Therapeutic
49. Schell SR, Talamimi MA, Udelsman R: Laparoscopic outcome of primary aldosteronism: adrenalectomy versus
adrenalectomy for nonmalignant disease: improved safety, enucleation of aldosterone-producing adenoma. J Urol
morbidity, and cost-effectiveness. Surg Endosc 1999; 1995; 153:1775–1780.
13:30–34. 58. Jeschke K, Janetschek G, Peschel R, et al: Laparoscopic
50. Vargas HI, Kavoussi LR, Bartlett DL, et al: Laparoscopic partial adrenalectomy in patients with aldosterone-
adrenalectomy: a new standard of care. Urology 1997; producing adenomas: indications, technique, and results.
49:673–678. Urology 2003; 61:69–72.
51. Bolli M, Oertli D, Staub J, et al: Laparoscopic 59. Kaouk JH, Matin S, Bravo EL, et al: Laparoscopic
adrenalectomy: the new standard? Swiss Med Wkly 2002; bilateral partial adrenalectomy for pheochromocytoma.
132:12–16. Urology 2002; 60:1100–1103.
52. Hazzan D, Shiloni E, Golijanin D, et al: Laparoscopic vs
open adrenalectomy for benign adrenal neoplasm. Surg
Endosc 2001; 15:1356–1358.
C H A P T E R

10 Diagnosis and Staging of Renal Cell


Carcinoma
Frederick A. Klein, MD, Judson R. Gash, MD,
and W. Bedford Waters, MD

Renal cell carcinoma (RCC) accounted for 3% of all patients diagnosed with RCC survive 5 years after diagno-
adult malignancies in 2003. Approximately 31,900 new sis, comparable to the expected survival experience for the
cases were diagnosed in 2003 and 11,900 deaths occurred general population. The survival patterns are similar for
in the United States.1 Although the overall incidence of men and women. The overall 5-year relative survival rates,
noncutaneous cancers fell by an average of 1.1% annually as well as survival rates by tumor stage at diagnosis have
in the United States between 1992 and 1998, the inci- improved substantially over time for whites but not for
dence of RCC has risen by an average of 3.7% annually blacks. The 5-year relative survival rates for black versus
over the same period.1 This increased incidence has been white men were (75% versus 89%) for localized disease. In
attributed to the increased use of ultrasonography (US), contrast, black women had survival rates similar to white
computed tomography (CT) scan, and magnetic reso- women. The race and gender differentials in survival pat-
nance imaging (MRI) for the evaluation of a variety of terns argue against early detection of preclinical tumors as
abdominal, back, or gastrointestinal complaints.2 RCC the sole explanation for the increasing trends of RCC.5 A
has been called the “internists tumor” in the past because recent report analyzing population-based statistics from
of the multitude of constitutional symptoms that patients the SEER registry demonstrates an increase in detection of
presented with.3,4 Now it is called the “radiologist all stages of RCC but no change in the rate of advanced or
tumor” because 25% to 40% are detected incidentally metastatic disease. The rates of localized, regional, and dis-
using these radiologic imaging modalities. tant RCC were 45%, 23%, and 32%, respectively, during
1973–1985 and 54%, 21%, and 25%, respectively, from
1986 to 1998. Plotting incidence rate versus diagnosis by
INCIDENCE
year and by stage revealed a trend toward increased inci-
Incidence rates of RCC and mortality rates of kidney can- dence in all three stages. The three groups had significantly
cer have increased steadily over time in the United different rates of incidence increase.10 Even with the
States.2,5 Between 1975 and 1995, incidence increased increased use of abdominal imaging, there was no signifi-
annually by 2.3% among white men, 3.1% among white cant difference in renal cancer stage at presentation in
women, 3.9% among black men, and 4.3% among black 1986–1998, compared to 1973–1985. These results imply
women. Since the mid-1980s, the incidence rates among that the increased use of abdominal imaging may be detect-
blacks have exceeded those among whites of both sexes. ing some indolent kidney cancers and further suggests that
Rapid increases in the incidence of kidney cancer also have other factors may be contributing to an overall increase in
been observed in other countries.5–9 Overall, approximately the incidence of renal cancer.
one-half of the RCCs reported to the U.S. Surveillance,
Epidemiology, and End Results (SEER) program from
ETIOLOGY/RISK FACTORS
1975 to 1995 were diagnosed while localized, with regional
and distant tumors comprising approximately 20% and The etiology of RCC is not known, although some risk
25%, respectively. In general, approximately 60% of factors are known. Cigarette smoking has at least a 30%

173
174 Part III Kidney and Ureter

to 50% increase in men and women. The risk has been palpable mass is rarely found.22 When this classic triad is
shown to decline after smoking cessation. An association found, there is usually metastatic disease present. Before
with other tobacco products, including cigars, pipes, and the use of US, CT, and MRI most patients presented
chewing tobacco, has not been consistently observed.5 with one of these signs or symptoms. Patients often pre-
Obesity, particularly in women, has a positive association. sented with other signs and symptoms of advanced dis-
Obese persons have experienced elevated risks of 20% to ease: weight loss, fever of unknown origin, and night
more than 3-fold. The risks tend to rise with increasing sweats. Physical findings included palpable mass, nonre-
levels of body mass index. The mechanism by which obe- ducing varicocele, bilateral lower extremity edema, pal-
sity predisposes to RCC is unclear. Obesity may influ- pable cervical/supraclavicular lymphadenopathy, abdominal
ence risk by increasing the levels of endogenous bruit, penile/vaginal nodules, and caput medusa.
estrogens5,11 and may increase the bioavailability of free Paraneoplastic syndromes are found in approximately
insulin-like growth factors5,12 that may be involved in 10% to 40% of patients with RCC (Table 10-1) as well as
renal carcinogenesis.5,13 Hypertension and the long-term bizarre metastatic patterns with the disease.5 The presence
use of diuretics have been examined. The cumulative evi- of a paraneoplastic syndrome does not imply metastatic dis-
dence suggests that of the two variables, hypertension ease. For example, the presence of hepatic dysfunction (i.e.,
itself, rather than diuretics, may have a role in the etiol- Stauffer’s syndrome) is manifested by an elevated alkaline
ogy of RCC. The mechanism by which high blood pres- phosphatase, α2-globulin, a prolonged partial thrombo-
sure may affect risk is unclear.5 plastin, or hypoalbuminemia. Twenty percent to thirty per-
Patients on long-term hemodialysis may develop cys- cent have elevated serum bilirubin or transaminases.3,16
tic changes of the kidney with associated carcinoma. Other common findings include thrombocytopenia and
Ishikawa14 reported that 43% of 96 patients who were on neutropenia, and typical symptoms include fever and
dialysis for less than 3 years developed cystic disease and weight loss.3,16 Hepatic metastases must be excluded.
79.3% of those who had been on dialysis for more than Biopsy, when indicated, often demonstrates nonspecific
3 years also developed cystic disease. Four patients had hepatitis associated with a prominent lymphocytic infil-
adenocarcinoma of the kidney; all 4 were in their 3rd and trate.16,23 Elevated serum levels of IL-6 have been found in
4th decade and had been on dialysis for more than 5 patients with Stauffer’s syndrome, and it is believed that
years. In the United States, one series reported 17/31 this and other cytokines may play a pathogenic role.16,24
(55%) developed cystic disease on hemodialysis and 3/12
(25%) on peritoneal dialysis. The incidence of develop-
ing cystic disease was 30% for patients on dialysis for less
than 2 years, 33% for 2–4 years, and 67% for those dia- Table 10-1 Paraneoplastic Syndromes Associated
lyzed longer than 4 years. The calculated incidence with Renal Cell Cancer
reported for hemodialysis was 0.10% per patient per year Syndrome Incidence (%)
and 0.08% per patient per year on continuous ambula-
tory peritoneal dislysis (CAPD). Solid renal tumors were Anemia 20–40
found in 3/43 (6.9%) who had nephrectomy. Adult cystic
Cachexia, fatigue, weight loss 33
disease of the kidney is felt to be a consequence of renal
failure or a humoral factor rather than caused by Fever 30
hemodialysis itself.15 Other studies have shown the rela-
tive risk of RCC in patients with end-stage renal failure Hypertension 24
has been estimated to be 5- to 100-fold higher than in the
Hypercalcemia 10–15
general population.16–21 Current recommendations are to
screen only patients with a long life expectancy and no Hepatic-dysfunction (Stauffer’s syndrome) 3–6
major comorbidities with periodic ultrasound or CT
beginning during the 3rd year on hemodialysis.16 Amyloidosis 3–5
Twenty-seven percent of patients with RCC will have a
Erythrocytosis 3-4
diagnosis of at least one other malignancy in their lifetime,
the most common malignancies are breast cancer, prostate Enteropathy 3
cancer, bladder cancer, and non-Hodgkin’s lymphoma.
Neuromyopathy 3

CLINICAL PRESENTATION From Chow W, Devesa SS, Fraumeni JF Jr: Epidemiology of renal
cell carcinoma. In Vogelzang NJ, Scardino PT, Shipley WU,
In those patients presenting with signs or symptoms, Coffey DS (eds): Comprehensive Textbook of Genitourinary
60% have hematuria, 50% have pain, and 30% have a Oncology, 2nd edition, pp 101–110. Philadelphia, Lippincott
mass. The classic triad of gross hematuria, flank pain, and Williams & Wilkins, 2000.
Chapter 10 Diagnosis and Staging of Renal Cell Carcinoma 175

Hepatic function normalizes after nephrectomy in 60% to is associated with good survival.35,38,39 Ki-67 identifies an
70% of cases. Persistence or recurrence of hepatic dysfunc- antigen of 395 kDa whose expression is detectable during
tion is almost always indicative of the presence of viable the G1 phase, increases during S phase and rapidly
tumor and thus portends a poor prognostic finding.16 decreases after mitosis. There is an agreement that Ki-67
These paraneoplastic syndromes may be due to spe- is an excellent maker of proliferation in histologic mate-
cific hormone production by the tumor cells or an rial analyzed immunohistochemically. According to the
immune response to the tumor. Hormones produced by result of several studies, the Ki-67 index is correlated
RCCs include parathyroid-like hormone, gonadotropins, with the histologic grade and stage,35,36,37 and might be a
placentolactogen, adrenocorticotrophic hormone-like more powerful prognostic factor than the PCNA index.
substance, renin, erythropoietin, glucagons, human Rini and Vogelzang39 concluded that because Ki-67 is
chorionic gonadotropin, and insulin.25,26 present in cells during all cell cycle phases, it provides a
In general, treatment of paraneoplastic syndromes more accurate determination of the proliferation rate
associated with RCC has required nephrectomy and/or than the PCNA index but more multivariate studies are
systemic immunotherapy, and, except for hypercalcemia, needed.29,35,39,40
medical therapies have not proven helpful.16 Cell adhesion molecules and angiogenesis factors have
also been evaluated. Cadherins are a large family of trans-
SERUM MARKERS membrane proteins responsible for mediating cell-to-cell
adhesion, and when expression decreases, their inherent
Serum ferritin,27 erythropoietin,28 calcium,25 and renin3 ability to modulate and preserve epithelial integrity
levels have been shown historically to be significantly diminishes. Lack of E-cadherin expression correlates
elevated and higher than controls in patients with with aggressiveness in several tumors, but in RCC, only
RCC.29 Hematocrit and platelet levels can also prove 20% express this glycoprotein.41 E-cadherin is localized
credible in regards to prognosis and recurrence. Symbas to Bowman’s capsule and other tubular segments rather
et al.30 report a mean difference in survival of 59 months than the proximal tubular epithelium, calling into ques-
with pathologic stage IV RCC and a normal platelet tion whether it plays an integral role in RCC carcino-
count versus those with thrombocytosis (>400,000) when genesis.29 Shimazui42 has shown that cadherin-6 is the
controlling for pathologic stage, nuclear grade, and cell major one in the proximal renal tubules and the tumors
type.29,30,31 Anemia and serum iron have also been useful themselves. His group found that aberrant expression of
as tumor markers for initial evaluation and follow-up.29,32 cahderin-6 connoted poor survival.29,42,43 Paul et al.44
Normochromic and normocytic anemia and anemia of have reproduced these data wherein the majority of
chronic diseases are the most common hematologic RCCs with histology-associated poor prognosis (i.e.,
abnormalities associated with RCC.29 high-grade clear cell cancers and sarcomatoid renal
tumors) showed aberrant expression. The tumor with a
MOLECULAR MAKERS historically good prognosis (low-grade clear cell carcino-
mas and papillary cancers) exhibited normal cadherin-6
The search for better markers has increasingly moved expression.29,44
toward the molecular level. The prognosis for patients Studies investigating the role of angiogenesis, using
with locally confined RCC is known to be variable and microvessel density (MVD) as a marker, as a correlate
emphasis on morphologic character, proteins, antigens with the development of metastases have not been
and other prognostic markers is being sought to aid in rewarding. There was no correlation to clinical stage,
the diagnosis and prognosis of RCC.29 Molecular mark- pathologic stage, or tumor grade.29,45 RCC is a vascular
ers of proliferation like Ki-67, silver staining nucleolar tumor, and its direct relationship to angiogenesis has yet
organizer regions (AgNOR), and proliferating cell to be completely determined.29
nuclear antigen (PCNA) are present in cycling cells and The p53 protein has also been studied in RCC. The
therefore have potential utility in estimating the biologic p53 protein binds DNA and is believed to regulate tran-
aggressiveness of a given tumor. AgNOR reflect tran- scription, acting as a “checkpoint” to induce cell cycle
scription activity of ribosomal RNA and cellular mitotic arrest.29,46 This tumor suppressor gene, when mutated,
activity. For some authors the AgNOR score is an inde- inactivates the normal function of DNA damage surveil-
pendent prognostic factor associated with survival,33–35 lance. Aneuploid cells originate, carcinogenesis occurs,
whereas for others it is associated with histologic grade and tumor progression can ensue.47 Mutant p53 proteins
but is not an independent factor.36 PCNA is a protein have a prolonged half-life and with accumulation are
synthesized during the late G1 and S phases of the cell detectable with immunohistochemical analysis.48
cycle. For some authors a low PCNA index (less than However, controversy exists with regards to the
10%) is an independent positive predictor of disease-free frequency of the mutation in RCC, ranging from 4% to
survival but not of overall survival,37 whereas for others it 40% of RCC specimens tested, and its resultant
176 Part III Kidney and Ureter

prognostic power. The clinical significance of p53 and and has an equal male to female ratio. Tumors associated
other apoptotic markers has yet to be determined.29,49,50 with VHL are predominantly of the clear cell type and are
Carbonic anhydrase IX (CAIX) protein, a member of associated with germline mutations of the tumor suppres-
the carbonic anhydrase family, is thought to play a role in sor gene, VHL gene, located on chromosome 3p. After
the regulation of cell proliferation in response to hypoxic extensive work, the VHL gene has been mapped to the
conditions and may be involved in oncogenesis and 3p25–26 region and VHL inactivation by point mutation
tumor progression.51–54. Constitutive expression of and allelic loss has been reported to occur in both sporadic
CAIX as a result of von Hippel-Lindau (VHL) protein and VHL-associated RCC. Specific sites or types of muta-
mutations has been described for RCC.55 Recent studies tions within the VHL gene appear to correlate with spe-
now indicate that expression of CAIX is regulated by the cific phenotypic expression of the gene: VHL type I (VHL
hypoxia-inducible factor (HIF) 1 transcriptional complex without pheochromocytomas) and VHL type II (VHL
that mediates expression of a number of genes in with pheochromocytomas). VHL disease is characterized
response to hypoxic conditions.56 It has been postulated by renal cysts, RCC (clear cell histology), retinal heman-
that cell surface carbonic anhydrase regulate acid-base giomas, hemangioblastomas of the cerebellum and spinal
balance to optimize conditions in the tumor invasive- cord, pancreatic carcinomas and cysts, epididymal cysts
ness.54 Acidification of the extracellular matrix is known and cystadenomas, and pheochromocytomas.60–68
to induce expression of angiogenic factors57 and may The VHL gene product forms a complex that degrades
inhibit cellular immunity,58 which additionally promotes two α subunits of HIF, an intracellular protein that plays
tumor aggressiveness. In addition, there is some evidence an important role in regulating cellular responses to
for the association of CAIX with loss of contact inhibi- hypoxia, starvation, and other stresses.16,69 The HIFα sub-
tion and anchorage dependence of cancer cells.59 Bui units are transcription factors that regulate the expression
et al.51 have investigated CAIX as kidney cancer marker of a number of proteins, including vascular endothelial
as an independent predictor of progression and survival. growth factor (VEGF), the primary proangiogenic growth
Immunohistochemical analysis using a CAIX mono- factor in RCC, contributing to the pronounced neovascu-
clonal assay was performed on tissue microassays con- larity associated with RCC,16,63,70 glucose transporter
structed from paraffin-embedded specimens from 321 (GLUT-1), and transforming growth factor (TGF)-α.
patients treated by nephrectomy for clear cell RCC. Hereditary papillary renal carcinoma (HPRC) is a
CAIX staining was correlated with response to treatment, hereditary cancer syndrome, which generally develops in
clinical factors, pathologic features, and survival. CAIX older patients (50s and 60s). The affected individuals are
staining was present in 94% of clear cell RCCs. Survival at risk to develop bilateral, multifocal papillary RCC.
tree analysis determined that a cutoff of 85% CAIX Linkage analysis of the families led to the discovery of the
staining provided the most accurate prediction of sur- HPRC gene on chromosome 7.71–73 This syndrome,
vival. Low CAIX (≥85%) staining was an independent which has an autosomal dominant inheritance pattern, is
poor prognostic factor for survival for patients with caused by missense mutations in the tyrosine kinase
metastatic RCC, with a hazard ratio of 3.10 ( p < 0.001). domain of the MET proto-oncogene at 7q31.73 Patients
CAIX significantly substratified patients with metastatic with germline or somatic mutations in the MET proto-
disease when analyzed by T stage, Fuhrman grade, nodal oncogene develop a specific subtype of papillary renal
involvement, and performance status ( p < 0.001, p = 0.001, carcinoma—papillary renal carcinoma type 1. In vitro
p = 0.009, p = 0.005, respectively). For patients with and in vivo studies suggest that MET functions as a dom-
nonmetastatic RCC and at high risk for progression, inantly acting oncogene in HPRC and in sporadic papil-
low CAIX predicted a worse outcome similar to patients lary renal carcinoma.74
with metastatic disease ( p = 0.058). CAIX status may Patients with hereditary hair follicle tumors (fibrofol-
potentially aid in the selection of patients who might liculomas) on their face and neck have a high risk for
benefit from IL-2 or CAIX-targeted therapies. developing kidney cancer (20% to 30%), lung cysts
Furthermore, patients with high-risk localized RCC and (90%), and pneumothorax (20%). Inherited fibrofollicu-
low CAIX may be potential candidates for adjuvant loma is called Birt-Hogg-Dubé syndrome (BHD). The
immunotherapy.51 BHD gene has been localized to the short arm of chro-
mosome 17. BHD patients are at risk for the develop-
ment of chromophobe RCC, oncocytic renal carcinoma,
MOLECULAR GENETICS
and oncocytoma.75–78
To date, four major dominantly inherited forms of RCC Familial renal oncocytoma is an entity currently
have been described. RCC occurs in association with VHL undergoing evaluation and definition, in which multiple
disease. About 45% of patients with VHL disease have bilateral renal oncocytomas develop in affected family
RCC, which is metastatic in half of the cases at diagnosis, members. The genetic defect involved in the pathogene-
often bilateral and multifocal, occurs in younger patients, sis of familial renal oncocytoma has not yet been identi-
Chapter 10 Diagnosis and Staging of Renal Cell Carcinoma 177

fied. However, clues to the location of this gene have bivalved. Microscopically they can include clear cell, gran-
come from cytogenetic studies: one characterized by the ular cell, or mixed types.16 The loss of loci on the short
loss of chromosomes 1 and Y,79 and the other by translo- arm of chromosome 3 has been the predominant mutation
cations involving the breakpoint region 11q13.68,80–82 linked to clear cell carcinoma, the most common subtype
The untreated natural history of familial renal oncocy- of RCC.66,88 Allelic loss of 3p is now thought to be an early
toma is currently being studied.83 event in the pathogenesis of clear cell carcinoma, whereas
Hereditary leiomyomatosis renal carcinoma (HLRC) others now consider additional allelic losses on chromo-
is another type of hereditary renal carcinoma. Affected some 17 to be associated with advanced disease.89
HLRC individuals are at risk for the development of Papillary RCC represents 10% to 15% of all RCCs.
cutaneous leiomyomas, uterine leiomyomas (fibroids), They can be inherited, multifocal, bilateral, and gener-
and renal carcinoma. The HLRC renal tumors are of ally portend a more favorable prognosis. The cytogenetic
varying histologic type, but the predominate histologic features are associated with loss of Y chromosome along
phenotype is type 2 papillary renal carcinoma.84 with trisomies of chromosomes 3q, 7, 12, 16, 17, and
20.85 Papillary RCC is more likely to be hypovascular,
perhaps owing to lack of VHL mutations, which regulate
PATHOLOGY
VEGF, the primary proangiogenic molecule in RCC.16
RCCs originate from renal tubular cells. Renal cancer The other genetic features of papillary RCC have been
has an equal frequency on the right and left and is dis- discussed earlier.
tributed equally throughout the kidney. Fifteen percent Chromophobe cell carcinoma accounts for approxi-
of tumors extend to the renal vein, and 8% extend to the mately 5% of all RCCs. It appears to be derived from the
vena cava. The histologic classification of RCC has cortical portion of the collecting duct. Histologically,
undergone a major revision since 1990. Traditionally, they are composed of two distinct cell types, those with
renal cancers have been classified as clear cell, granular, granular eosinophilic cytoplasm and those with pale
sarcomatoid, and papillary types. Malignant renal epithe- cytoplasm. Ultrastructural analysis shows numerous
lial neoplasms are now subdivided under the new classifi- cytoplasmic microvesicles, and characteristic cytoplasm
cation system (Table 10-2) based on prominent staining with Hale’s colloidal iron is typical.89,90 The clin-
morphologic features.85 Major changes include the addi- ical behavior of patients with chromophobe carcinoma
tion of a new histologic subtype, the chromophobe cell shows that the majority of tumors appear to be localized
carcinoma; the reclassification of granular cell tumors to the kidney at the time of diagnosis. Although some
into other categories; and the recognition that sarcoma- may be large, clinical behavior seems to be similar to
toid lesions represent poorly differentiated elements respective stages of clear cell carcinoma.90,91 However,
derived from other histologic subtypes, rather than a dis- Renshaw and colleagues92 reported a more adverse prog-
tinct tumor type.16,85–87 Common or conventional RCC nosis associated with chromophobe RCC. In their series,
accounts for approximately 70% to 80% of all RCCs. high-grade disease was common, and 7 of 25 patients
They are highly vascular and are typically yellow when eventually developed metastatic disease.16,92
Collecting duct, or Bellini’s duct, carcinomas repre-
sent a rare group of renal neoplasms. They arise from the
distal portion of the nephron, commonly strike in a
Table 10-2 Classification of Renal Cell Carcinoma
young population, and carry a usually poor prognosis.
Incidence Affected They are usually high grade, present at an advanced
Subtype (%) Chromosomes stage, and do not respond to conventional therapy. There
Conventional 70–80 3p, 17
is a frequent loss of heterozygosity at chromosome
regions on 1q, 6p, 8p, 13q, and 21q.93 High density map-
Papillary 10–15 3q, 7, 12, 16, 17, 20, Y ping of arm 1q in 13 collecting tumors shows an area of
minimal deletion around the area located at 1q32.1–32.2
Chromophobe 4–5 1, 2, 6, 10, 13, 17, 21 in 69% of examined specimens.94
Collecting duct <1 1q, 6p, 8p, 13q, 21q
Renal medullary carcinoma is a relatively new histo-
logic subtype of RCC that occurs almost exclusively in
Medullary cell <1 Not defined association with the sickle cell trait. It is typically diag-
nosed in young African Americans often in the third
Oncocytoma 3–7 1, Y decade of life.16,95 Many cases are both locally advanced
and metastatic at the time of diagnosis. Most patients do
From Störkel S, Ebie JN, Adlakha K, et al: Classification of renal cell
carcinoma: Workgroup no. 1, Union Internationale Contre le Cancer
not respond to therapy and succumb to their disease
(UICC) and the American Joint Committee on Cancer (AJCC). within several months.16,95 Histologically, this tumor
Cancer 1997; 80:987–989. shares many features with collecting duct carcinoma.
178 Part III Kidney and Ureter

The site of origin (renal papillae) and association with rounded bulge to the renal contour or displacing the col-
sickle cell trait suggest that a relatively hypoxic environ- lecting system (Figure 10-1). Calcifications, although
ment may contribute to tumorigenesis.16 observed in only about 10% of RCCs on the IVP, suggest
RCC, unclassified, is a category reserved for malig- neoplasm, especially when they are chunky and centrally
nant epithelial tumors that do not conform to any of the located within the mass.98 Unlike cysts, which should not
other subtypes. Of the total group of RCCs, they com- enhance and therefore appear lucent, a mass with density
prise approximately 4% to 5%.85,89 Specifically, compos- approaching that of surrounding renal parenchyma sug-
ites of recognized subtypes, tumors with mucin gests enhancement and is worrisome for neoplasm.
production, and tumors with extensive sarcomatoid Uncommonly, RCC may present as diffuse enlargement
changes without recognizable epithelial elements are of the kidney when the tumor is large or infiltrating.
examples of this classification.89 Diffuse replacement of the renal parenchyma or venous
invasion may result in lack of normal renal function and
nonvisualization on the IVP. Despite even the most
Imaging Evaluation
suggestive findings, virtually all parenchymal masses
RCC may be radiologically discovered in one of two identified on the IVP are nonspecific and thus require
ways. It may be diagnosed in patients with symptoms, further evaluation. Since more than 80% of suspected
such as hematuria or pain or, alternatively, it may be renal masses on excretory urography are simple cysts or
found incidentally in patients undergoing imaging stud- pseudomasses, ultrasound should be performed in most
ies for other reasons. With the increasing utilization of cases as the diagnosis of a cyst can usually be made with
cross-sectional modalities, such as CT and US, the ultrasound and the workup terminated.99 However, if
majority of renal cell cancers are now discovered the mass has an appearance suggestive of a solid lesion,
serendipitously.96 Once identified, renal masses usually such as calcification or increased density, then a dedi-
require further characterization. Although imaging does cated renal CT is an appropriate next test for further
not allow a histologic diagnosis, radiologic studies are evaluation.
critical to exclude benign entities, such as cysts, abscesses,
and pseudomasses. Also, an attempt should be made to
Ultrasonography
eliminate certain neoplastic conditions, such as angiomy-
olipomas, metastases, and lymphoma. Finally, imaging is The ease of use, safety and wide availability of ultrasound
indispensable in the staging of RCC. Many radiologic make this an excellent and widely utilized modality for
techniques are available for the detection, characteriza- evaluation of the abdomen and pelvis, including the uri-
tion, and staging of renal cell cancer, including intra- nary tract. Perhaps as many as 50% of all diagnosed
venous urography, US, CT, MRI, nuclear medicine, and RCCs, therefore, are initially detected during an ultra-
positron emission tomography (PET) imaging. Invasive sound examination100 Ultrasound is sensitive for the
angiography, once a bastion of renal evaluation, now detection of renal masses, including RCC with sensitivity
plays little role in the diagnostic evaluation of RCC.

Detection and Characterization


Intravenous Urography
For over five decades the intravenous pyelogram (IVP)
has been the workhorse of imaging the urinary tract and
detecting renal neoplasms. However, its limitations,
including lack of sensitivity and specificity for renal
masses, have been well known. Many renal masses may
not be identifiable on the IVP, including those that are
small, those that do not distort the collecting system, and
those that do not create a bulge to the renal contour
(intrasubstance lesions and lesions that are anteriorly or
posteriorly oriented). Scrupulous technique, including
nephrotomography, oblique views, and bowel prep, does
improve sensitivity yet 15% of lesions greater than 3 cm
will be missed and the insensitivity rises dramatically for
smaller lesions with between 48% of masses of 2–3 cm
and 79% of masses of 1–2 cm will not be visible.97 Most Figure 10-1 4-cm RCC arising from the lower pole of the left
RCCs appear as a mass lesion on the IVP, creating a kidney.
Chapter 10 Diagnosis and Staging of Renal Cell Carcinoma 179

approaching 100% for lesions larger than 2.5 cm, normal echotexture and vascularity on color or power
although there is diminishing sensitivity for smaller Doppler often allow a confident diagnosis. It is uncom-
lesions.101 Tumors may be missed when they are isoechoic mon for RCC to appear predominately cystic on ultra-
to renal parenchyma, small, or when the examination is sound. Most of these show thick septations or chunky
technically difficult, such as with large patients; however, calcifications and are not confused with benign lesions.
new technologies, including harmonic imaging, have Indeterminate cystic lesions should generally be evalu-
resulted in dramatic improvements in image quality and ated with dedicated renal CT as occasionally benign
should result in increased sensitivity even in these set- lesions seen at CT may have a more malignant appear-
tings. Most RCCs at ultrasound appear as solid lesions ance on US. Septae may be exaggerated and appear more
(Figure 10-2). Earlier ultrasound reports suggested that ominous and intervening normal renal tissue may look
most RCCs were hypoechoic; however, these were often like tumor. Conversely, ultrasound may clarify indeter-
large lesions at an advanced stage. Although RCC may minate lesions seen at CT, including the hyperdense cyst
have variable echogenicity, the majority of currently and “too small to characterize” lesions. Hyperdense cysts
identified and smaller RCCs are in fact hyperechoic, seen at CT may appear entirely anechoic at ultrasound
sometimes markedly so, and may then be confused with confirming its benign nature.106 Similarly, “too small to
angiomyolipomas.102 Several features have been assessed characterize” lesions on CT can be shown to be simple
to try and distinguish RCC from the classically hypere- cysts in some circumstances, although the need to con-
choic angiomyolipoma.103 A very hyperechoic lesion sim- firm these lesions with ultrasound or follow these is
ilar to renal sinus fat along with posterior shadowing highly debatable.107 Despite the dramatic improvement
favors the diagnosis of an acute myelogenous leukemia in image quality, the role of ultrasound remains essen-
(AML) while a hypoechoic halo with small intratumoral tially the same in the evaluation and characterization of
cystic components suggests RCC.104 Unfortunately, the renal mass. The study is useful for excluding moder-
there is too great an overlap of findings to allow a confi- ate to larger tumors as well as identifying and distin-
dent diagnosis and it is necessary to confirm the diagno- guishing the simple cyst from the solid mass; however,
sis of angiomyolipoma with CT. Along with echogenicity the solid lesion remains nonspecific and requires further
that is variable but often increased, RCCs often have ill- evaluation, typically with CT.
defined or irregular interfaces with normal renal
parenchyma and are frequently lobulated. Although
Computed Tomography
Doppler waveform and color analysis have been analyzed
with some success in differentiating renal masses at ultra- It is safe to say that the CT now forms the backbone of
sound, solid renal lesions remain nonspecific.105 True radiologic evaluation of the urinary tract, including the
solid masses, however, must be differentiated from renal mass. The advent of multidetector spiral CT
pseudomasses and normal variants. A prominent column (MDCT) has further propelled CT to the forefront of
of Bertin, renal hypertrophy adjacent to an area of scar, urologic imaging including near isotropic imaging with
and fetal lobation can mimic a renal mass, although their scan thicknesses less than 1 mm and subsecond scanning
speeds allowing imaging of the kidneys in multiple
phases. This high spatial and temporal resolution com-
bined with quantifiable density values (Hounsfield units,
HU) and high contrast resolution makes CT excellent
for RCC detection and characterization. Finally, the wide
availability and relative safety of CT cinch its appeal. To
take full advantage of the strengths of CT, however, scan-
ning technique is critical. Virtually, all scans obtained for
renal indications should include noncontrast scans of the
kidneys. These scans can help to identify calcifications and
hemorrhage but are most important as a baseline to
assess lesion enhancement, a critical component to diag-
nosing malignancy. Four phases of enhancement may be
evaluated with CT, including arterial phase, corti-
comedullary phase, nephrographic phase, and excretory
phase. The nephrographic phase, occurring at approxi-
mately 90 seconds after contrast administration and
Figure 10-2 US demonstrating a 1.5-cm slightly hyperechoic when there is uniform cortical and medullary enhance-
renal cell carcinoma arising from the mid-pole of the right ment, is the most critical in mass detection and charac-
kidney. terization and should be included in all renal CT
180 Part III Kidney and Ureter

studies.108,109 Corticomedullary phase, seen between 30 pies. Urothelial neoplasms that invade the kidney tend to
and 90 seconds, when there is predominant cortical have an infiltrating growth pattern rather than the expan-
enhancement but little contrast in the medulla, can be sile rounded enlargement, typical of RCC.111
occasionally valuable for detecting hyperenhancing Additionally, the epicenter of the mass should be in the
lesions and tumors in end stage renal disease, but may renal pelvis. Retrograde pyelography, endoscopic biopsy,
miss centrally located masses and may underestimate and urine cytology may be used to confirm the diagnosis.
lesion enhancement.108 Excretory phase images obtained Secondary and systemic neoplastic involvement of the
after 3–5 minutes may be useful for evaluating the col- kidney can be seen with lymphoma and metastatic dis-
lecting system for invasion or urothelial lesions. Arterial ease. Similar to urothelial lesions, these entities tend to
phase scans are used for CT angiography studies to aid have an infiltrative growth pattern maintaining the reni-
surgical planning, especially when nephron sparring form shape of the kidneys.112 Also, these lesions are often
surgery is contemplated. Finally, delayed images may be multiple, bilateral and there is usually evidence of disease
used to assess lesion de-enhancement, suggesting elsewhere. Renal medullary carcinomas, first described in
enhancement and vascularity in “reverse,” when noncon- 1995 and seen in African-American males with sickle cell
trast images have not been obtained.110 trait, along with collecting duct carcinomas also show an
Detection of RCC is accomplished with high sensitiv- infiltrating growth pattern, arise centrally in the
ity by a combination of an up-to-date helical scanner, medullary region and are often large and heterogeneous
scrupulous technique, and a careful, educated review of at presentation.112 Benign neoplasms account for 10% to
the images. Sensitivities of 95% for 8–15 mm lesions and 15% of renal tumors and are of increasing frequency with
essentially 100% for larger lesions are obtainable using the now common incidental detection of smaller
helical technique and nephrographic phase images108 lesions.113 The presence of unequivocal fat (<10 HU)
(Figure 10-3). Once identified, an attempt should be within a mass generally allows a confident diagnosis of a
made to further characterize the mass and identify benign angiomyolipoma, with a few caveats. RCC can
pseudomasses and normal variants, inflammatory lesions, engulf renal sinus or perinephric fat and mimic an
benign tumors, and systemic or urothelial neoplasms. angiomyolipoma. Additionally, there have been a few
Focal renal hypertrophy, dromedary humps, and fetal case reports of adipose tissue within RCCs, although
lobulation may suggest a solid renal mass on noncontrast most of these also contained calcifications, which is dis-
images; however, they are easily recognized on multipha- tinctly rare in angiomyolipomas.114 Conversely, a small
sic images where their enhancement and homogeneity percentage, about 10%, of AMLs do not contain
matches normal renal parenchyma on all sequences. detectable fat and are indistinguishable from RCCs115
Occasionally, acute infarcts or focal infection may mimic (Figure 10-4). Oncocytoma is the most common benign
a mass but their infiltrating appearance and the clinical neoplasm and is being detected with increasing fre-
history usually allow a confident diagnosis. Urothelial
neoplasms, typically transitional cell carcinoma, account
for about 10% of renal malignancy and should be differ-
entiated from RCC due to their distinct surgical thera-

Figure 10-4 CT demonstrating a 6-cm necrotic RCC of the


Figure 10-3 Incidentally detected 1-cm RCC arising from the right kidney in a patient who also has a single 2-cm fat-
right kidney. containing AML in the left kidney.
Chapter 10 Diagnosis and Staging of Renal Cell Carcinoma 181

Figure 10-5 Large right RCC mimicking an oncocytoma.

quency.116 When large, a central scar within an otherwise


homogeneous mass may suggest the diagnosis, although
overlapping imaging findings do not allow a radiograph-
ically confident distinction from RCC and resection is
generally necessary for histologic confirmation (Figure
10-5). Smaller oncocytomas have imaging findings indis-
tinguishable from RCC. The appearance of RCC on CT
is somewhat variable depending on size, vascularity,
degree of necrosis, hemorrhage, and to some extent cell
type. Larger lesions tend to commonly have greater het- Figure 10-6 Noncontrast CT (A) demonstrating a
erogeneity with necrosis and hemorrhage, whereas homogeneous hyperdense lesion within the upper pole of the
smaller lesions tend to be much more homogeneous. left kidney mimicking a hyperdense cyst; however, after
contrast (B) there was a 45-HU increase in lesion density
Vascularity and enhancement is a key feature of RCC. A
typical of a RCC.
minimum of 10 to 15-HU increase in attenuation is seen
compared with baseline density, although many show
much greater enhancement117 (Figure 10-6A and B). require more discussion. Bosniak II lesions are minimally
Chromophobe and papillary subtypes tend to be more complicated with thin nonenhancing septations or thin,
hypovascular, whereas the more common conventional linear calcifications, whereas Bosniak III lesions are more
or clear cell types often show marked enhancement after concerning and complex with thicker septations, walls, or
contrast.118 Calcification is not uncommon in RCC, calcifications. Bosniak II lesions are very likely benign
especially in papillary and chromophobe subtypes.23 while the incidence of malignancy for class III lesions
Most renal cell cancers are rounded and expansile may approach 50%, generally necessitating surgical
although a small percentage may be infiltrating, mimick- resection. There have, however, been problems noted
ing other infiltrating malignancies. Additionally, RCC with this classification scheme. Some studies have shown
may present as a cystic lesion and the evaluation of the an unacceptable frequency of malignancy in lesions clas-
complex renal cyst is a fairly common diagnostic problem sified as category II.121 In 1993, Bosniak added an IIF
for the urologist and radiologist (Figure 10-7). In 1986, category for lesions slightly more complex and worri-
Bosniak proposed a classification scheme for cystic renal some than typical class II cysts. These IIF lesions are usu-
masses.119 Criteria are shown in Table 10-3.120 Lesions ally closely followed (thus the “F”) rather than ignored or
meeting criteria for simple cysts and obviously malignant excised. One additional problem with the Bosniak
cystic masses, Bosniak categories I and IV lesions, respec- scheme has been interobserver variability between class
tively, are rarely problematic. Bosniak II and III lesions II and class III lesions. In one study, 60 of 227 lesions
182 Part III Kidney and Ureter

originally classified as class IIF or III lesions were reclas- advance being the development of fast scanning
sified when reviewed by a different radiologist.122 sequences which allow breath-hold imaging and conse-
Despite its difficulties, however, the Bosniak classifica- quent motion artifact reduction, improved spatial resolu-
tion of cystic renal masses remains a very useful frame- tion, and increased temporal resolution, including
work for these difficult lesions, allowing evidenced-based multiphasic renal evaluation similar to CT. This com-
therapy and further study. bined with its spectacular contrast resolution and multi-
The trend of increasing incidence and improved sur- planar capabilities strengthen the role of MRI in renal
vival rate in RCC are related to several factors, perhaps mass evaluation and depiction as well as staging. High
none more important than advances in computed tomog- cost, relative lack of availability, and experience have so
raphy, which is the standard imaging technique for eval- far limited its utility. The primary role for MRI has been
uation of RCC and the solid and complex cystic renal when CT is contraindicated (renal insufficiency, contrast
mass.113 allergy), when CT is indeterminate or, in some settings,
for staging. MRI is extremely sensitive to detecting
enhancement and may therefore be especially useful in
Magnetic Resonance Imaging
certain settings, such as small lesions, complex cysts, and
Just as technical advances have improved US and CT in hypovascular neoplasms. Imaging protocols should
recent years, MRI has made great strides with a primary include T1 and T2 weighted, preferably breath-held,
images along with gadolinium-enhanced sequences. The
sensitivity of MRI for detecting renal masses is similar to
CT, or perhaps slightly improved for polar lesions due to
the ability to obtain direct coronal images.123 Like CT,
the appearance of RCC on MRI is variable depending on
various factors. Typically, the lesions are isointense to
hypointense on T1 sequences, moderately hyperintense
on T2, show enhancement after gadolinium, and demon-
strate variable degrees of hemorrhage and necrosis
(Figure 10-8). MRI, as yet, offers no significant advan-
tage compared with CT for lesion characterization with
interpretative principles as described in the CT section.
In fact, objective assessment of enhancement is more
complex and arbitrary compared to CT.124 For now, MRI
remains a problem solver, however, the unceasing tech-
nologic advances sure to come, such as in MR spec-
troscopy, may propel this modality to the forefront of
renal mass imaging.

Nuclear Medicine and PET imaging


Figure 10-7 Cystic RCC arising from the lower pole of the
Traditional nuclear medicine techniques, such as
left kidney.
renograms and renal scans, play little role in the detec-

Table 10-3 Bosniak Criteria Cystic Renal Masses


Criteria I II III IV

Wall Thin Thin More than thin Thick or nodular

Calcifications None Few, thin Irregular Coarse

Septations None Few, thin More than a few Thick, numerous

Density (HU) 0–20 0–20 0–20 More than 20

Enhancement None None None Present

From Kausik S, Segura JW, King BF Jr: Classification and management of simple and complex renal cysts.
AUA Update Ser 2002; 21:82–87.
Chapter 10 Diagnosis and Staging of Renal Cell Carcinoma 183

Figure 10-8 Axial T2 weighted MRI reveals a large


heterogeneous RCC.

tion and evaluation of renal masses. The occasional prob-


lematic pseudotumor may be confidently diagnosed as
normal renal tissue with a renal scan although CT usu-
ally has little difficulty with this situation. PET scanning
is limited in the evaluation of primary kidney lesions by
the high intrinsic renal uptake of 18-fluoro-2-deoxyglucose
(FDG), but early work has shown some promise in
lesion characterization, especially in evaluating complex
Figure 10-9 Robson classification for the staging of RCC.
cysts where FDG uptake may be able to differentiate
benign from malignant lesions.125 In another study, PET
was 94% accurate in assessing suspicious renal masses modified by Hermanek and Schrott128 in 1990 repre-
identified on another imaging modality, although there sented a major improvement because it both separated
was an inexplicable false negative in a patient with a 6-cm lymphatic from venous involvement and quantified the
RCC.126 extent of involvement of each system. After this system
was popularized, there was easy global comparison of
clinical and pathologic data that were found to correlate
Renal Cell Carcinoma—Staging
well with patient outcomes.
The most commonly used staging system for RCC prior In 1997, the International Union Against Cancer and
to the 1990s was that proposed by Holland in accordance the American Joint Committee on Cancer proposed a
with the classification system developed by Robson, change in the TNM system that was widely adopted until
Murphy, Flocks, and Kadesky (Figure 10-9).127 The lim- the most recently adopted TNM system in 2002 (Table
itations of this classification scheme have been well 10-4).129 Division between T1 and T2 tumors was origi-
defined and evidenced by the grouping in stage III that nally 2.5 cm and was changed to 7 cm because it reflected
lumps lymphatic metastases (a very poor prognostic find- the mean tumor size in a database where no prognostic
ing) with venous involvement that, even though may be significance or survival differences could be shown
extensive, can be treated with aggressive surgery and between cut points at 5, 7.5, and 10 cm.130 In 2002, the
portend a good long-term outcome. Likewise, since the widely acceptable modification was dividing stage T1 into
extent of nodal and venous involvement was not delin- T1a representing tumor size of 4 cm or less and T1b rep-
eated in this system, the prognostic significance of these resenting tumor size between 4 and 7 cm. The rationale
pathologic findings were lost. for this division is that tumors of 4 cm or less, unilateral
The tumor, nodes, and metastasis (TNM) system pro- and capsular confined, have excellent outcomes and T1a
posed by the International Union Against Cancer and tumors are very amenable to nephron sparing procedures
184 Part III Kidney and Ureter

Table 10-4 Definition of TNM


Primary tumor (T) TX Primary tumor cannot be assessed

T0 No evidence of primary tumor

T1 Tumor 7 cm or less in greatest dimension, limited to the kidney

T1a Tumor 4 cm or less in greatest dimension, limited to the kidney

T1b Tumor more than 4 cm but not more than 7 cm in greatest dimension, limited to the
kidney

T2 Tumor more than 7 cm in greatest dimension, limited to the kidney

T3 Tumor extends into major veins or invades adrenal gland or perinephric tissues but not
beyond Gerota’s fascia

T3a Tumor directly invades adrenal gland or perirenal and/or renal sinus fat but not beyond
Gerota’s fascia

T3b Tumor grossly extends into the renal vein or its segmental (muscle-containing) branches,
or vena cava below the diaphragm

T3c Tumor grossly extends into vena cava above diaphragm or invades the wall of the vena cava

T4 Tumor invades beyond Gerota’s fascia

Regional lymph nodes (N)* NX Regional lymph nodes cannot be assessed

N0 No regional lymph node metastases

N1 Metastases in a single regional lymph node

N2 Metastasis in more than one regional lymph node

Distant metastasis (M) MX Distant metastasis cannot be assessed

M0 No distant metastasis

M1 Distant metastasis
Stage grouping

Stage I T1 N0 M0

Stage II T2 N0 M0

Stage III T1 N1 M0

T2 N1 M0

T3 N0 M0

T3 N1 M0

T3a N0 M0

T3a N1 M0

T3b N0 M0

T3b N1 M0

T3c N0 M0

T3c N1 M0
Chapter 10 Diagnosis and Staging of Renal Cell Carcinoma 185

Table 10-4 Definition of TNM—cont’d


Stage 4 T4 N0 M0

T4 N1 M0

Any T N2 M0

Any T Any N M1

Histopathologic type

The predominant cancer is adenocarcinoma; subtypes are clear cell and granular cell carcinoma. The use of the following
grading system is recommended when feasible. Sarcomas and adenomas are not included. The histopathologic types are:

Conventional (clear cell) renal carcinoma


Papillary RCC
Chromophobe renal carcinoma
Collecting duct carcinoma

Histologic grade (G)

GX Grade cannot be assessed


G1 Well differentiated
G2 Moderately differentiated
G3–4 Poorly differentiated or undifferentiated

From Kidney. In Greene FL, Page DL, Fleming ID, et al (eds): AJCC Cancer Staging Manual, 6th edition, pp 323–328. New York, Springer, 2002.
*Laterality does not affect the N classification.
Note: If a lymph node dissection is performed, then pathologic evaluation would ordinarily include at least eight nodes.

with outcomes equivalent to those treated with radical alkaline phosphatase, α-glutamyltransferase, glutamic
nephrectomy.131–133 Other changes in 1997 included sim- oxaloacetic transaminase, and glutamate pyruvate
plifying the classification of the extent of nodal involve- transaminase, an elevated sedimentation rate, and signif-
ment and reclassifying the extent of venous tumor icant anemia all point to the likelihood of advanced
thrombi. It was emphasized that adrenal gland involve- disease.136,137 In the absence of either bone pain or eleva-
ment by local tumor extension should be T3a because this tions of serum alkaline phosphatase or calcium, it is
situation has a better prognosis than isolated adrenal uncommon for a patient to harbor skeletal metastases.
metastases that reflect hematogenous spread and por- The routine addition of a bone scan for the evaluation of
tends a poor prognosis.134,135 In addition, stage T3a bone metastases from RCC is not indicated. In fact, in a
includes invasion of renal sinus fat because there is no study by Koga et al.,139 the overall incidence of bone
rationale for including them in another category. Finally, metastases was 17% in a group of all stages of RCC.
it has been suggested that vena caval wall invasion has a Likewise, in all patients stages T1–T3, only 4 (1.8%) had
worse prognosis than simply the cephalad extent of the osseous metastases without other metastatic sites, lymph
tumor thrombus and should be classified as stage T3c.136 node involvement, or localized bone pain.
Clinical staging is important and has been shown to The TNM system is felt to be inadequate by some
correlate directly with prognosis as well as play a critical groups to describe prognosis in 2003. The addition of
role in appropriate preoperative planning. The clinical Fuhrman’s tumor grade and Eastern Cooperative
staging of RCC begins with a thorough history, physical Oncology Group (ECOG) patient performance status
examination, and selected laboratory tests. Symptoms (PS) has been shown to be an important prognostic
and signs of significant weight loss (greater than 10% of tool.140 Zisman et al.140 from University of California
body weight), any localized bone pain, a palpable mass, Los Angeles (UCLA) have described the UCLA
or lymphadenopathy, and/or a poor performance status Integrated Staging System (UISS). The medical records
suggest more advanced disease and a worse progno- of 477 patients undergoing nephrectomy between 1989
sis137,138 New onset of a nonreducing varicocele and and 1999 were evaluated. Median age was 61 years, male-
lower extremity edema suggest venous involvement. to-female ratio was 2.2:1, and median follow-up was 37
Likewise, other constitutional symptoms of pain, fever, months. Survival time was the primary end point
night sweats, and abnormal liver function tests, such as assessed. Sixty-four possible combinations of stage,
186 Part III Kidney and Ureter

grade, and ECOG PS were analyzed and collapsed into tation and most common staging error is determining
distinct groups. The internal validity of the categories extracapsular tumor extension into the perinephric fat
was challenged by a univariate analysis and a multivariate and as such distinguishing stages T1a, T1b, and T2 from
analysis testing for the accountability of each UISS cate- stage T3a. Perinephric stranding adjacent to renal cancers
gory against independent variable shown to have impact is a nonspecific sign and can represent edema, fibrosis,
on survival.140 prominent engorged vessels or tumor, and falsely suggest
Combining and stratifying 1997 TNM stage, Fuhrman’s capsular extension and a T3a lesion. Since, in the major-
grade and ECOG PS resulted in five stratification groups ity of cases tumor extension is microscopic, it does not
designated UISS, and numbered I–V. The overall 5-year change patient management and is of less clinical signif-
survival for the UISS groups I–V, respectively, are: 94%, icance. Perinephric stranding has been reportedly seen in
67%, 39%, 23%, and 0% (Table 10-5). UISS is an emerg- up to 50% of confirmed T1 and T2 lesions.142 On the
ing important prognostic tool for counseling patients with other hand, to confidently suggest perinephric spread
various stages of kidney cancer. It is currently being vali- requires nodular soft tissue beyond the capsule. Although
dated in two large-scale external databases.140 this finding has a 98% specificity, sensitivity is only 46%
Imaging plays a crucial role in staging and treatment with the findings usually seen with larger lesions.142
planning; however, only tests where the results affect the (Figure 10-10) Another difficult issue is for CT to distin-
patient’s management should be performed. The diagno- guish between abuttal and invasion of adjacent organs,
sis of RCC will most commonly be made incidentally by such as liver, spleen, duodenum, and pancreas.
ultrasound or CT.141 Therefore, performing excretory Obliteration of fat places suggesting invasion can be dif-
urography is of no useful value. Although ultrasound can ficult to evaluate on CT and lead to false positive results
yield significant staging information, it is rarely used as in 15% of cases.143 (Figure 10-11) In selected cases where
the sole imaging modality. Limitations are associated more careful surgical planning is necessary, an MRI may
with operator ability and interference by body habitus be helpful.144
and overlying bowel gas. Regional lymph node involvement (N1–N2) portends
In the majority of cases, radiographic staging of RCC a poor prognosis. CT must rely on anatomic size criteria
can be accomplished with the meticulous use of high to diagnose or exclude nodal disease and sensitivity and
quality abdominal CT and routine chest x-ray or chest specificity will vary based on the size used to consider a
CT. Staging accuracy for CT has been reported to be in node enlarged. Nodal size of 2 cm or greater usually is
the 90% to 91% range.117,142 Probably the greatest limi- associated with malignancy. Using a threshold of 1 cm,
false negative rates for microscopic metastases are low
Table 10-5 UCLA Integrated Staging System (UISS) (4%), while the false positive rates have ranged from 3%
Overall to 43%.136,142,145,146 These false positive rates may even
5-Year be higher in patients where venous invasion and tumor
TNM Survival necrosis are present as a result of reactive hyperplasia.
UISS Stage ECOGPS Grade (%) Because of this imaging unreliability, extirpative surgery
should not be withheld without tissue confirmation of
I I 0 1–2 94

II I 0 3–4

II Any Any 67

III 0 Any

III ≥1 1

III III ≥1 2–4 39

IV 0 1–2

IV IV 0 3–4 23

IV ≥1 1–3

V IV ≥1 4 0

From Zisman A, Pantuck AJ, Dorey F, et al: Improved


prognostication of renal cell carcinoma using an integrated staging Figure 10-10 Large right RCC with nodular soft tissue in the
system. J Clin Oncol 2001; 19:1649–1657. perinephric space consistent with transcapsular spread.
Chapter 10 Diagnosis and Staging of Renal Cell Carcinoma 187

tributaries. Enhancement of the thrombus and direct


continuity with the primary tumor are typical of neoplas-
tic thrombus. Bland thrombus, commonly seen below the
level of the renal veins in lesions with large venous exten-
sion, shows no enhancement after contrast. False nega-
tives occur in patients where a mass effect on the vein
may make tumor thrombus identification difficult (more
common on the right) and in situations where tumor
hypervascularity alone causes venous enlargement and
suggests thrombus.156 The superior most extent of
venous invasion is particularly crucial to delineate espe-
cially when the thrombus extends above the diaphragm
(T3c), as this takes careful surgical planning. Finally,
venous extension into the wall of the renal vein or IVC
may make resection impossible, but this is difficult to
Figure 10-11 Very large RCC with incontrovertible invasion diagnose unless tumor is clearly seen extending beyond
of the liver. the venous wall into the perivascular tissues.
Careful attention should also be paid on CT to the
malignancy by aspiration cytology or biopsy. MRI may remainder of the kidney as well; especially when
be more reliable for distinguishing lymphadenopathy nephron-sparing surgery is contemplated. Multifocal
from vascular structures. RCC occurs in as many as 25% of patients who are can-
Ipsilateral adrenal gland involvement by RCC has didates for partial nephrectomy.157 Although multicentric
been found in up to 10% of radial nephrectomy speci- lesions tend to be small (from “microscopic” to 1 cm), as
mens.147 With metastatic RCC, ipsilateral adrenal many as 50% of these may be detectable on preoperative
involvement has been found to be as high as 19.1% while imaging.158 Similarly, the contralateral kidney must be
the contralateral gland is affected in 11.5%.148 Patients at closely evaluated since bilateral disease is becoming more
higher risk for adrenal involvement have an enlarged commonly recognized.159
adrenal on CT, have an upper pole tumor, and/or have Metastatic disease is not uncommonly identified at the
large tumors with extensive malignant replacement of the time of staging and most often occurs in the lungs, bone,
kidney. Microscopic invasion, of course, evades detection liver, and brain. Mediastinal and mesenteric nodes, pan-
by any imaging modality; however, adrenal involvement creas, and other less common areas of distant dissemina-
with small mid- or lower pole lesions is reported to be as tion may be seen. A careful search is prudent, especially
low as 1.9%.146 Therefore, with a normal adrenal gland for small lesions. Renal cell metastatic lesions are often
on CT and small mid- and lower pole tumors, routine hypervascular and aggressive in appearance. The ques-
adrenal resection is not justified.149,150 tion of whether to routinely obtain a chest CT for staging
Tumor extension or invasion into the renal vein, vena is commonly asked. In general, a routine anterio-
cava, or into the heart is a unique finding associated with posterior and lateral plain chest film is sufficient to
RCC. The incidence of renal vein involvement and for screen for parenchymal lesions but not for mediastinal
caval extension at the time of surgical exploration has nodes less than 2 cm.160 If enlarged lymph nodes are seen
been reported to be between 10% and 39% and 4% and on abdominal CT or if lymphangitic metastases are sus-
25%, respectively.151–153 Reports of successful interior pected on plain chest film, chest CT is encouraged.
venacavotomy and tumor thrombectomy date back to Similarly, routine use of pelvic CT has been shown to pro-
1913.154 Since there are better outcomes with complete duce negligible yield and is not routinely recommended.161
surgical excision in these patients when compared to MRI has some advantages over CT for staging RCC.
those with lymphatic invasion, accurate venous staging It is noninvasive, does not subject the patient to the
with a thorough knowledge of the extent of venous risks of intravenous contrast, and provides high intrin-
involvement is necessary for optimal surgical planning sic contrast resolution permitting improved depiction
and management. The sensitivity and specificity of CT of tumor thrombus extension and invasion, invasion of
detecting vascular extension of tumor thrombus have adjacent organs, and capsular extension. This is well
been reported at 78% and 96%, respectively.143 Accuracy shown in Figure 10-12. MRI has been advocated
for caval involvement is greater (86%) than for renal vein because of its ability to directly image in multiple planes
involvement (50%).155 The CT findings suggestive of although with today’s modern CT scanners, reconstruc-
venous involvement include venous enlargement, abrupt tion in virtually all planes is possible without resolution
change in the caliber of the vein, low density intralumi- loss. A recent study by Pretorius et al.162 has shown sim-
nal filling defects, and the presence of collateral venous ilar staging accuracies for CT and MRI. In addition,
188 Part III Kidney and Ureter

high uptake in the brain, myocardium, kidneys, and blad-


der. The increased activity in the genitourinary system is
due to the inability of the nephron to resorb filtered
FDG in the convoluted tubule.164 Although PET-FDG
has detected histologically confirmed RCCs, there have
been significant false positives. A more important role for
PET-FDG (presently under investigation) is for evalua-
tion of metastatic disease and to detect recurrence or
progression and response to systemic therapies. In one
study, 10 of 10 patients were proven to have active pro-
gressive disease whereas only 7 of these 10 cases were
positive with conventional imaging. In 5 patients with no
evidence of disease or complete remission, PET-FDG
studies were negative in all whereas conventional imag-
ing was positive in three cases.165 In another study, PET-
FDG has been used to establish a response to cancer
immunotherapy with interleukin (IL)-2. The lack of
PET activity at prior active sites suggests fibrosis or
necrosis and lack of activity within a previous active renal
mass or lymph node suggests lack of viable cells and
Figure 10-12 Axial MRI demonstrating low signal tumor necrosis.166 In another recent study of the clinical role of
thrombus involving the right renal vein to the level of the IVC. PET-FDG, the detection and management of RCC,
PET-FDG accurately detected local disease spread and
metastatic disease in patients with RCC and altered treat-
several logistical and practical factors hinder MRI avail- ment plans in 40%.116 Finally, in a study comparing
ability, including long scan times, lack of a consistent PET-FDG to routine bone scan, PET-FDG sensitivity
oral contrast agent, inability to image the lungs, claus- and accuracy were 100% and 100%, respectively, com-
trophobia issues, and contraindications, such as pace- pared to bone scans that were 77.5% and 59.6%, respec-
makers. Finally, image quality can be unpredictable tively.167 Clearly, more data and experience are needed
secondary to motion and other artifacts especially before PET-FDG is recommended for routine use in
related to the inability to cooperate with multiple RCC. However, this technology with fusion imaging
sequential breath-holds. MRI’s role is a problem-solver combining CT with PET, more accurate preoperative
especially when CT findings are equivocal and in staging, and better patient care is on the horizon.
patients with renal insufficiency.
Venacavography has predominantly been abandoned.
Monitoring for Recurrence or Progression
For venous thrombus staging, it is invasive, has contrast
of Disease
risks, and risks of detaching tumor, may require superior
vena cava puncture to determine the cephalad extent of Follow-up algorithms for RCC remain controversial as
the thrombus, and is subject to the inaccuracies created there remains no systemic treatment regiment with
by slow flow and suboptimal contrast filling. In selected highly satisfactory response rates that would significantly
cases where MRI may be contraindicated and other stag- improve survival in an asymptomatic patient. Exceptions
ing modalities equivocal, transesophageal echo can be to this would be isolated renal fossa recurrences that are
very accurate for establishing the cephalad extent of rare and solitary delayed metastases (also rare).168 Levy et
tumor thrombus. Transesophageal echo, however, is al.169 from M.D. Anderson Cancer Center retrospec-
invasive.163 tively reviewed the records of 286 patients with patho-
logic stage T1–T3 RCC of which 68 relapsed. For pT1
disease only, an annual chest x-ray and liver function tests
Positron Emission Tomography
were recommended. For pT2 and pT3 tumors, it was rec-
PET permits noninvasive measurement of biochemical ommended to obtain chest radiographs every 6 months
as well as physiologic reactions. Positron-emitting for 3 years, then annually, and perform surveillance
radioisotopes labeled to endogenous biochemicals or abdominal CT scans at 24 and 60 months unless the
drugs are injected intravenously and accumulate in vari- patient becomes symptomatic. It was recommended that
ous organ tissues depending on their biochemical or bone and brain surveillance studies only be performed for
physiologic properties. Presently, the most commonly site-specific symptoms, elevated alkaline phosphatase, or
used agent is the glucose analog FDG. Normally, there is the development of metastases at other sites.169 The use
Chapter 10 Diagnosis and Staging of Renal Cell Carcinoma 189

of periodic CT and/or ultrasound for patients with post- REFERENCES


partial nephrectomy, however, is warranted.170
Evaluation of these patients can prove challenging as fre- 1. Jenal J, Murray T, Samuels A, et al: Cancer statistics,
quently there is a wedge-shaped or concave defect often 2003. CA Cancer J Clin 2003; 53:27–43.
filled with fat placed to help with hemostasis and 2. Chow WH, Devesa SS, Warren JL, Fraumeni JF Jr:
decrease urinary extravasation. Hypodense areas of old Kidney cancer incidence trends in the United States.
hemorrhage may be present as well as perfusion defects JAMA 1999; 281:1628–1631.
3. Sufrin G, Chasan S, Golio A, et al: Paraneoplastic and
secondary to devascularized surrounding tissue. For
serologic syndromes of renal adenocarcinoma. Semin
patients with renal insufficiency or contrast allergy, MRI Urol 1989; 7:158–171.
is useful. “How often to obtain imaging” is controversial. 4. Gold PJ, Fefer A, Thompson JA: Paraneoplastic
However, every 6–12 months is acceptable. Similarly, it manifestations of renal cell carcinoma. Semin Urol Oncol
has been recommended that CT scans be done every 3–6 1996; 14:216–222.
months postoperatively on patients having had “bench 5. Chow W, Devesa SS, Fraumeni JF Jr: Epidemiology
surgery” and autotransplant for RCC.171 of renal cell carcinoma. In Vogelzang NJ, Scardino
Periodic relapse monitoring is also important in fol- PT, Shipley WU, Coffey DS (eds): Comprehensive
lowing patients with small indeterminate lesions on CT Textbook of Genitourinary Oncology, 2nd edition,
or ultrasound and on patients who have increased pp 101–110. Philadelphia, Lippincott Williams &
comorbidities where intervention would only be under- Wilkins, 2000.
6. Parkin DM, Whelan SL, Ferlay J, et al: Cancer Incidence
taken if substantial growth is documented. Birnbaum et
in Five Continents, vol VII [IARC Sci. Pub. No. 143].
al.172 studied a group of patients longitudinally looking at Lyon, France, International Agency for Research on
yearly growth rates on CT. They found the growth rate Cancer, 1997.
to be between 0 and 1.6 cm in diameter in a year with a 7. Jin F, Devesa SS, Zheng W, et al: Cancer incidence trends
mean growth rate of 0.5 cm. It, therefore, seems reason- in urban Shanghai, 1972–1989. Int J Cancer 1993;
able to obtain imaging every 6 months for 1 year and if 53:764–770.
there is no change, settle for yearly follow-up. 8. McCredie M: Bladder and kidney cancers. In Doll R,
Fraumeni JF Jr, Muir CS (eds) Cancer Surveys: Rends in
Cancer Incidence and Mortality, pp 343–363. New York,
Percutaneous Needle Aspiration Cytology and Cold Spring Harbor Laboratory Press, 1994.
Biopsy for the Diagnosis of Renal Cell Carcinoma 9. Liu S, Semenciw R, Morrison H, et al: Kidney cancer in
Canada: the rapidly increasing incidence of
Percutaneous renal tumor sampling continues to be
adenocarcinoma in adults and seniors. Can J Public
frowned on except for specific indications as outlined in Health 1997; 8:99–104.
Table 10-6. Although the risk of seeding normal tissue in 10. Hock LM, Lynch J, Balaji KC: Increasing incidence of all
the needle tract is small, it is likely that the true incidence stages of kidney cancer in the last 2 decades in the United
is undeteresteimated.173 If seeding occurs, potentially States: an analysis of surveillance, epidemiology and end
curable organ-confined renal cancer is compromised. In results program data. J Urol 2002; 167:57–60.
general, a CT or ultrasound guided aspiration or biopsy 11. Kumar A, Mittal S, Buckshee K, et al: Reproductive
is quick and safe, but may be associated with bleeding or functions in obese women. Prog Food Nutr Sci 1993;
arteriovenous fistula formation. Furthermore, there are 17:89–98.
significant equivocal or false negative results due to sam- 12. Attia N, Tamborlane WV, Heptulla R, et al: The
metabolic syndrome and insulin-like growth factor I
pling error and the inability to conclusively differentiate
regulation in adolescent obesity. J Clin Endocrinol Metab
oncocytomas from low-grade RCCs.
1998; 83:1467–1471.
13. Narayan S, Roy D: Insulin-like growth factor I receptors
Table 10-6 Indications for Percutaneous Biopsy are increased in estrogen-induced kidney tumors. Cancer
or Aspiration of Renal Mass Res 1993; 53:2256–2259.
14. Ishikawa I: Development of adenocarcinoma and acquired
● When a lesion present in a patient with lymphoma cystic disease of the kidney in hemodialysis patients.
persists after systemic response to therapy Princess Takamatsu Symp 1987; 18:77–86.
15. Chandhoke PS, Torrence RJ, Clayman RV, et al: Acquired
● Distinguish between primary neoplasia and metastatic
cystic disease of the kidney: a management dilemma.
disease
J Urol 1992; 147:969–974.
● Differentiate between an infected cyst or abscess and 16. Novick AC, Campbell SC: Renal tumors. In Walsh PC,
neoplasm Retik AB, Vaughan ED Jr, Wein AJ (eds) Campbell’s
Urology, 8th edition, pp 2672–2731. Philadelphia,
● Obtain tissue diagnosis in a patient who is not an Saunders, 2002.
operative candidate 17. Ishikawa I, Saito Y, Onouchi Z, et al: A ten-year
prospective study on the development of renal cell
190 Part III Kidney and Ureter

carcinoma in dialysis patients. Am J Kidney Dis 1990; 35. Méjean M, Oudard S, Thiounn N: Prognostic factors of
16:452–458. renal cell carcinoma. J Urol 2003; 169:821–827.
18. Matson MA, Cohen EP: Acquired cystic kidney disease: 36. Tannapfel A, Hahn HA, Katalinic A, et al: Prognostic
occurrence, prevalence, and renal cancers. Medicine 1990; value of ploidy and proliferation markers in renal cell
69:217–226. carcinoma. Cancer 1996; 77:164–171.
19. Ishikawa I: Uremic acquired renal cystic disease. Nephron 37. Hofmockel G, Tsatalpas P, Muller H, et al: Significance
1991; 58:257–261. of conventional and new prognostic factors for locally
20. Levine E, Slusher SL, Grantham JJ, et al: Natural history confined renal cell carcinoma. Cancer 1995; 76:
of acquired renal cystic disease in dialysis patients. Am J 296–306.
Roentgenol 1991; 156:501–506. 38. Cronin KJ, Williams NN, Kerin MJ, et al: Proliferating
21. Levine E: Renal cell carcinoma in uremic acquired renal cell nuclear antigen: a new prognostic indicator in renal
cystic disease: incidence, detection, and management. cell carcinoma. J Urol 1994; 152:834–836.
Urologic Radiol 1992; 13:203–210. 39. Rini BI, Vogelzang NJ: Prognostic factors in renal
22. Jayson M, Sanders H: Increased of serendipitously carcinoma. Semin Oncol 2000; 27:213–220.
discovered renal cell carcinoma. Urology 1994; 40. Haitel A, Weiner H, Migschitz B, et al: Proliferating cell
51:203–205. nuclear antigen and MIB-I. An alternative to classic
23. Hanash KA: The nonmetastatic hepatic dysfunction prognostic indicators in renal cell carcinomas? Am J Clin
syndrome associated with renal cell carcinoma Pathol 1997; 107:229–235.
(hypernephroma): Stauffer’s syndrome. Prog Clin Biol Res 41. Katagiri A, Wantanabe R, Tomita Y: E-cadherin
1982; 100:310–316. expression in renal cell cancer and its significance in
24. Blay JY, Rossi JF, Eijdenes J, et al: Role of interleukin-6 metastasis and survival. Br J Urol 1995; 71:376–379.
in the paraneoplastic inflammatory syndrome associated 42. Shimazui T, Giroldi L, Bringuier P, et al: Complex
with renal cell carcinoma. Int J Cancer 1997; cadherin expression in renal cell carcinoma. Can Res
72:424–430. 1996; 56:3234–3237.
25. Gotoh A, Kitazawa S, Mizuno Y, et al: Common 43. Shimazui T, Oosterwijk E, Akaza H, et al: Expression of
expression of parathyroid hormone-related protein and no cadherin-6 as a novel diagnostic tool to predict prognosis
correlation of calcium levels in renal cell carcinoma. of patients with E-cadherin-absent renal cell carcinoma.
Cancer 1993; 71:2803–2806. Clin Can Res 1998; 4:2419–2424.
26. McDougal WS, Garnick MB: Clinical signs and 44. Paul RF, Ewing C, Robinson J, et al: Cadherin 6, a cell
symptoms of renal cell carcinoma. In Vogelzang NJ, adhesion molecule specifically expressed in the proximal
Scardino PT, Shipley WU, Coffey DS (eds): tubule and renal cell carcinoma. Can Res 1997;
Comprehensive Textbook of Genitourinary Oncology, 57:2741–2748.
2nd edition, pp 111–115. Philadelphia, Lippincott 45. MacLennan G, Bostwick D: Microvessel density in renal
Williams & Wilkins, 2000. cell carcinoma: lack of prognostic significance. Urol 1995;
27. Esen A, Ozen H, Ayhan A, et al: Serum ferritin: a tumor 46:27–30.
marker for renal cell carcinoma. J Urol 1991; 46. Kuerbitz S, Plunkett B, Walsh W, et al: Wild-type p53 is
145:1134–1137. a cell cycle checkpoint determinant following irradiation.
28. Sufrin G, Mirand EA, Moore RH: Hormones in renal Proc Natl Acad Sci U S A 1992; 89:7491–7495.
cancer. J Urol 1977; 117:433–438. 47. Hollstein M, Sidransky D, Vogelstein B, et al: p53
29. Northway RO, Ritenour CWM, Marshall FF: Renal cell mutations in human cancers. Science 1991; 253:49–53.
carcinoma: an overview. AUA Update Ser 2001; 48. Lane D, Benchimol S: p53 oncogene of anti-oncogene?
20:194–199. Genes Dev 1990; 4:1–8.
30. Symbas NP, Townsend MF, El-Galley R, et al: Poor 49. Uhlman D, Nguyen P, Manivel C, et al: Association
prognosis associated with thrombocytosis in patients with of immunohistochemical staining for p53 with
renal cell carcinoma. BJU Int 2000; 86:203–207. metastatic progression and poor survival in patients
31. O’Keefe SC, Marshall FF, Issa MM, et al: with renal cell carcinoma. J Natl Can Inst 1994;
Thrombocytosis is associated with a significant increase in 86:1470–1474.
the cancer specific death rate after radical nephrectomy. 50. Shina H, Igawa M, Urakami S, et al: Clinical significance
J Urol 2002; 168:1378–1380. of immunohistochemically detectable p53 protein in renal
32. Yu C, Chen K, Chen M, et al: Serum iron as a tumor cell carcinoma. Eur Urol 1997; 31:73–80.
marker in renal cell carcinoma. Eur Urol 1991; 19:58–58. 51. Bui MHT, Seligson D, Han K, et al: Carbonic anhydrase
33. Yasunaga Y, Shin M, Miki T, et al: Prognostic factors of IX is an independent predictor of survival in advanced
renal cell carcinoma: a multivariate analysis. J Surg Oncol renal clear cell carcinoma: implications for prognosis and
1998; 68:11–18. therapy. Clin Cancer Res 2003; 9:802–811.
34. Delahunt B, Bethwaite PB, Nacey JN, et al: Proliferating 52. Pastorek J, Pastorekova S, Callebaut I, et al: Cloning and
cell nuclear antigen (PCNA) expression as a prognostic characterization MN, a human tumor-associated protein
indicator for renal cell carcinoma: comparison with with a domain homologous to carbonic anhydrase and a
tumour grade, mitotic index, and silver-staining nucleolar putative helix-loop-helix DNA binding segment.
organizer region numbers. J Pathol 1993; 170:471–477. Oncogene 1994; 9:2877–2888.
Chapter 10 Diagnosis and Staging of Renal Cell Carcinoma 191

53. Wykoff CC, Pugh CW, Maxwell PH, Harris AL, Ratcliffe 2nd edition, pp 116–128. Philadelphia, Lippincott
PJ: Identification of novel hypoxia dependent and Williams & Wilkins, 2000.
independent target genes of the von Hippel–Lindau 69. Maxwell PH, Wiesener MS, Chang GW, et al: The
(VHL) tumour suppressor by mRNA differential tumour suppressor protein VHL targets hypoxia-inducible
expression profiling. Oncogene 2000; 19:6297–6305. factors for oxygen-dependent proteolysis. Nature 1999;
54. Ivanov S, Liao SY, Ivanova A, et al: Expression of 399:271–275.
hypoxia-inducible cell-surface transmembrane carbonic 70. Iliopoulos O, Levy AP, Jiang C, et al: Negative regulation
anhydrases in human cancer. Am J Pathol 2001; of hypoxia-inducible genes by the von Hippel–Lindau
158:905–919. protein. Proc Natl Acad Sci U S A 1996; 93:
55. Ivanov SV, Kumin I, Wei MH, et al: Down-regulation of 10595–10599.
transmembrane carbonic anhydrases in renal cell 71. Zbar B, Tory K, Merino M, et al: Hereditary papillary
carcinoma cell lines by wild-type von Hippel–Lindau renal cell carcinoma. J Urol 1994; 151:561–566.
transgenes. Proc Natl Acad Sci U S A 1998; 72. Zbar B, Glenn G, Lubensky I, et al: Hereditary papillary
95:12596–12601. renal cell carcinoma. Clinical studies in 10 families. J Urol
56. Wykoff CC, Beasley NJ, Watson PH, et al: Hypoxia- 1995; 153:907–912.
inducible expression of tumor-associated carbonic 73. Schmidt L, Duh FM, Kishida T, et al: Germline and
anhydrases. Cancer Res 2000; 60:7075–7083. somatic mutations in the tyrosine kinase domain of the
57. Shi Q, Abbruzzese JL, Huang S, et al: Constitutive and MET proto-oncogene in papillary renal carcinomas. Nat
inducible interleukin 8 expression by hypoxia and acidosis Genet 1997; 16:68–73.
renders human pancreatic cancer cells more tumorigenic 74. Zhuang Z, Park WS, Pack S, et al: Trisomy 7-harbouring
and metastatic. Clin Cancer Res 1999; 5:3711–3721. non-random duplication of the mutant MET allele in
58. Giatromanolaki A, Koukourakis MI, Sivridis E, et al: hereditary papillary renal carcinomas. Nat Genet 1998;
Expression of hypoxia-inducible carbonic anhydrase-9 20:66–69.
relates to angiogenic pathways and independently to poor 75. Schmidt LS, Warren MB, Nickerson ML, et al:
outcome in non-small cell lung cancer. Cancer Res 2001; Birt–Hogg–Dube syndrome, a genodermatosis associated
6l:7992–7998. with spontaneous pneumothorax and kidney neoplasia,
59. Parkkila S, Rajaniemi H, Parkkila AK, et al: Carbonic maps to chromosome 17p11.2. Am J Hum Genet 2001;
anhydrase inhibitor suppresses invasion of renal cancer 69:876–882.
cells in vitro. Proc Natl Acad Sci U S A 2000; 76. Herring JC, Enquist EG, Chernoff A, et al: Parenchymal
97:2220–2224. sparing surgery in patients with hereditary renal cell
60. Kovacs G, Erlandsson R, Boldog F, et al: Consistent carcinoma: 10-year experience. J Urol 2001; 165:777–781.
chromosome 3p deletion and loss of heterozygosity in 77. Roth JS, Rabinowitz AD, Benson M, et al: Bilateral renal
renal cell carcinoma. Proc Natl Acad Sci U S A 1988; cell carcinoma in the Birt–Hogg–Dube syndrome. J Am
85:1571–1575. Acad Dermatol 1993; 20:1055–1058.
61. Kovacs G, Frisch S: Clonal chromosome abnormalities in 78. Pavlovich CP, Walther MM, Eyler RA, et al: Renal
tumor cells from patients with sporadic renal cell tumors in the Birt–Hogg–Dube syndrome. Am J Surg
carcinomas. Cancer Res 1989; 49:651–659. Pathol 2002; 26:1542–1552.
62. Seizinger BR, Rouleau GA, Ozeius LJ, et al: von 79. Crotty TB, Lawrence KM, Moertel CA, et al:
Hippel–Lindau disease maps to the region of Cytogenetic analysis os six renal oncocytomas and a
chromosome 3 associated with renal cell carcinoma. chromophobe cell renal carcinoma. Evidence that −Y, −1
Nature 1988; 332:268–269. may be a characteristic anomaly in renal oncocytomas.
63. Gnarra JR, Tory K, Weng Y, et al: Mutations of the VHL Cancer Genet Cytogenes 1992; 61:61–66.
tumour suppressor gene in renal carcinoma. Nat Genet 80. Fuzesi L, Gunawan B, Braun S, et al: Renal oncocytoma
1994; 7:85–90. with a translocation t(9;11)(p23;q13). J Urol 1994;
64. Herman JG, Latif F, Weng Y, et al: Silencing of the VHL 152:471–472.
tumor-suppressor gene by DNA methylation in renal 81. Neuhaus C, Dijkhuizen T, van den Berg E, et al:
carcinoma. Proc Natl Acad Sci U S A 1994; Involvement of the chromosomal region 11q13 in renal
91:9700–9704. oncocytoma: case report and literature review. Cancer
65. Linehan WM, Lerman MI, Zbar B: Identification of the Genet Cytogenes 1997; 94: 95–98.
von Hippel–Lindau (VHL) gene: its role in renal cancer. 82. Walter TA, Pennington RD, Decker HJ, et al:
JAMA 1995; 273:564–570. Translocation t(9;11)(p23;q12): a primary
66. Zbar B, Brauch H, Talmadge C, et al: Loss of alleles of chromosomal change in renal oncocytoma. J Urol
loci on the short arm of chromosome 3 in renal cell 1989; 142:117–119.
carcinoma. Nature 1987; 327:721–724. 83. Weirich G, Glenn G, Junker K, et al: Familial renal
67. Zbar B: von Hippel–Lindau disease and sporadic renal cell oncocytoma: clinicopathological study of 5 families.
carcinoma. Cancer Surv 1995; 25:219–232. J Urol 1998; 160: 335–340.
68. Walther MM, Enquist EG, Jennings SB, et al: Molecular 84. Choyke PL, Glenn GM, Walther MM, et al: Hereditary
genetics of renal cell carcinoma. In Vogelzang NJ, renal cancers. Radiology 2003; 226:33–46.
Scardino PT, Shipley WU, Coffey DS (eds): 85. Störkel S, Ebie JN, Adlakha K, et al: Classification of
Comprehensive Textbook of Genitourinary Oncology, renal cell carcinoma: workgroup no. 1, Union
192 Part III Kidney and Ureter

Internationale Contre le Cancer (UICC) and the 103 Yamashita Y, Ueno S, Makita O, et al: Hyperechoic renal
American Joint Committee on Cancer (AJCC). Cancer tumors: anechoic rim and intratumoral cysts in US
1997; 80:987–989. differentiation of renal cell carcinoma from
86. Weiss LM, Gelb AB: Adult renal epithelial neoplasms. angiomyolipoma. Radiology 1993; 188:179–182.
Am J Clin Pathol 1995; 103:624–634. 104. Siegel CL, Middleton WD, Teefey SA, et al:
87. Oyasu R: Renal cancer: histologic classification update. Angiomyolipoma and renal cell carcinoma: US
Int J Clin Oncol 1998; 3:125–133. differentiation. Radiology 1996; 198:789.
88. Foster K, Crossey P, Cairns P, et al: Molecular genetic 105. Kuijper D, Kruyt RH, Oudkerk M: Renal masses: value
investigation of sporadic renal cell carcinoma: analysis of of duplex Doppler ultrasound in the differential
allele loss on chromosomes 3p, 5q, 11p, 17 and 22. Br J diagnosis. J Urol 1994; 151:326.
Cancer 1994; 69:230–234. 106. Foster WL, Roberts L, Halvorsen RA, et al: Sonography
89. Ritenour CWM, Northway RO, Marshall FF: Renal of small renal masses with indeterminate density
tumors. In Gillenwater JY, Grayhack JT, Howards SS, characteristics on computed tomography. Urol Radiol
Mitchell ME (eds): Adult and Pediatric Urology, 4th 1988; 10:59.
edition, pp 611–639. Philadelphia, Lippincott Williams 107. Davidson AJ, Hartman DS, Choyke PL, Wagner BJ.
& Wilkins, 2002. Radiologic assessment of renal masses: implications for
90. Crotty T, Farrow G, Lieber M: Chromophobe cell renal patient care. Radiology 1997; 202:297–305.
carcinoma: clinicopathological features of 50 cases. J 108. Szolar DH, Kammerhuber F, Altziebler S, et al:
Urol 1995; 154: 964–967. Multiphasic helical CT of the kidney: increased
91. Campbell SC, Russo P, Hamed G, et al: Chromophobe conspicuity for detection and characterization of small
cell carcinoma of the kidney: a clinicopathologic study. (<3 cm) renal masses. Radiology 1997; 202:211–217.
Proc Am Urol Assoc 1996b; 155:299. 109. Kopka L, Fischer U, Zoeller G, et al: Dual-phase helical
92. Renshaw AA, Henske EP, Loughlin KR, et al: Aggressive CT of the kidney: value of the corticomedullary and
variants of chromophobe renal cell carcinoma. Cancer nephrographic phase for evaluation of renal lesions and
1996; 78:1756–1761. preoperative staging of renal cell carcinoma. Am J
93. Polascik TJ, Cairns P, Epstein JI: Distal nephron renal Roentgenol 1997; 169:1573–1578.
tumors: microsatellite allelotype. Cancer Res 1996; 110. Macari M, Bosniak MA. Delayed CT to evaluate renal
56:1892–1895. masses incidentally discovered at contrast-enhanced CT:
94. Steiner M, Goldman S, Fishman E, et al: The natural demonstration of vascularity with de-enhancement.
history of renal angiomyolipoma. J Urol 1993; Radiology 1999; 213:674–680.
150:1782–1786. 111. Gash J, Zagoria RJ, Dyer RB, Assimos DG: Imaging
95. Davis C Jr, Mostofi F, Sesterhenn I: Renal medullary features of infiltrating renal lesions. Crit Rev Diagn
carcinoma. The seventh sickle cell nephropathy. Am J Imaging 1992; 33:293–310.
Surg Pathol 1995; 19:1–11. 112. Pickhardt PF, Lonergan GF, Davis CF Jr, Kashitani N,
96. Homma Y, Kawabe K, Kitamura T, et al: Increased Wagner BF. Infiltrative renal lesions: radiologic-
incidental detection and reduced mortality in renal pathologic correlation. Radiographics 2000; 20:215–243.
cancer – recent retrospective analysis at eight 113. Zagoria RJ. Imaging of small renal masses. Am J
institutions. Int J Urol 1995; 2:77–80. Roentgenol 2000; 175:945–955.
97. Warshauer DM, McCarthy SM, Street L, et al: 114. DeAngelo P, Gash, JR, Horn AW, et al: Fat in renal cell
Detection of renal masses: sensitivities and specificities carcinoma that lacks associated calcification. Am J
of excretory urography/linear tomography, US and CT. Roentgenol 2002; 178:931.
Radiology 1988; 169:363–365. 115. Davidson AJ, Hartman DS, Choyke PL, Wagner BJ:
98. Daniel WW, Hartman GW, Witten DM, et al: Calcified Radiologic assessment of renal masses: implications for
renal masses: a review of ten years’ experience at the patient care. Radiology 1997; 202:297–305.
Mayo Clinic. Radiology 1972; 103:503. 116. Dechet CB, Sebo T, Farrow G, et al: Prospective
99. Einstein DM, Herts BR, Weaver R, et al: Evaluation of analysis of intraoperative frozen needle biopsy of
renal masses detected by excretory urography: cost- solid renal masses in adults. J Urol 1999;
effectiveness of sonography versus CT. Am J Roentgenol 162:1282–1285.
1995; 164:371–375. 117. Sheth S, Scatarige JC, Horton KM, et al: Current
100. Spouge AR, Wilson SR, Wooley B: Abdominal concepts in the diagnosis and management of renal cell
sonography in asymptomatic executives: prevalence of carcinoma: role of multidetector CT and three-
pathologic findings, potential benefits, and problems. dimensional CT. Radiographics 2001; 21:S237.
J Ultrasound Med 1996; 15:763. 118. Kim JK, Kim TK, Ahn HJ, et al: Differentiation of
101. Jamis-Dow CA, Choyke PL, Jennings SB, et al: Small subtypes of renal cell carcinoma on helical CT scan. Am
(≤3-cm) renal masses: detection with CT versus US J Roentgenol 2002; 178:1499.
and pathologic correlation. Radiology 1996; 198: 119. Bosniak MA. The current radiological approach to renal
785–788. cysts. Radiology 1986; 158:1–10.
102. Forman HP, Middleton WD, MElson GL, et al: 120. Kausik S, Segura JW, King BF Jr: Classification and
Increasing frequency of detection of hyperechoic renal management of simple and complex renal cysts. AUA
cell carcinomas. Radiology 1993; 188:431. Update Ser 2002; 21:82–87.
Chapter 10 Diagnosis and Staging of Renal Cell Carcinoma 193

121. Wilson TE, Doelle EA, Cohan RH, Wojno K, Korobkin and the American Joint Committee on Cancer (AJCC).
M: Cystic renal masses: a reevaluation of the usefulness Cancer 1997; 80:994–996.
of the Bosniak classification system. Acad Radiol 1996; 139. Koga S, Tsuda S, Nishikid M, et al: The diagnostic value
3:564–570. of bone scan in patients with renal cell carcinoma. J Urol
122. Lang EK, Macchia RJ, Gayle B, et al: CT-guided biopsy 2001; 166(6):2126–2128.
of indeterminate renal cystic masses (Bosniak 3 and 2F): 140. Zisman A, Pantuck AJ, Dorey F, et al: Improved
accuracy and impact on clinical management. Eur Radiol prognostication of renal cell carcinoma using an
2002; 12:2518–2524. integrated staging system. J Clin Oncol 2001;
123. Rofsky NM, Bosniak MA: MR imaging in the 19:1649–1657.
evaluation of renal masses. MRI Clin North Am 141. Homma Y, Kawabe K, Kitamura T, et al: Increased
1997; 5:67–81. incidental detection and reduced mortality in renal
124. Ho VB, Allen SF, Hood MN, et al: Renal masses: cancer – recent retrospective analysis at eight
quantitative assessment of enhancement with dynamic institutions. Int J Urol 1995; 2:77–80.
MR imaging. Radiology 2002; 224:695–700. 142. Johnson CD, Donnick NR, Cohan RH, et al: Renal
125. Goldberg MA, Mayo-Smith WW, Papanicolaou N, et al: adenocarcinoma: CT staging of 100 tumors. Am J
FDG PET characterization of renal masses: preliminary Roentgenol 1987; 148(1):59–63.
experience. Clin Radiol 1997; 52(7):510–515. 143. Bectol RE, Zagorie RJ: Imaging approach to staging of
126. Ramdave S, Thomas GW, Berlangieri SU, et al: Clinical renal cell carcinoma. Urol Clin North Am 1997;
role of F-18 fluorodeoxyglucose positron emission 24:507–522.
tomography for detection and management of renal cell 144. Choyke PL: Detection and staging of renal cancer. Magn
carcinoma. J Urol 2001; 166:825–830. Reson Imaging Clin North Am 1997; 5:29–47.
127. Holland JM: Cancer of the kidney: natural history and 145. Studer WE, Scherz S, Scheidegger J, et al: Enlargement
staging. Cancer 1973; 32:1030. of regional lymph nodes in renal cell carcinoma is
128. Hermanek P, Schrott KM: Evaluation of the new tumor, often not due to metastases. J Urol 1990; 144(2/1):
nodes, and metastases classification of renal cell 243–245.
carcinoma. J Urol 1990; 144:238–242. 146. Fein AB, Lee JKT, Balte DM, et al: Diagnosis and
129. Thompson IM Jr, Andriole GL, Blumenstein B, et al: staging of renal cell carcinoma: a comparison of MR
Kidney. In Greene FL, Page DL, Fleming ID, et al (eds): imaging and CT. Am J Roentgenol 1987;
AJCC Cancer Staging Manual, 6th edition, pp 323–328. 148(4):749–753.
New York, Springer, 2002. 147. Angervall L, Wahlquist L: Follow-up and prognosis of
130. Guinan PD, Vogelzant NJ, Frengen AM, et al: Renal cell renal carcinoma in a series operated by perifascial
carcinoma: tumor size, stage and survival. J Urol 1995; nephrectomy combine with adrenalectomy and
153:901–903. retroperitoneal lymphadenectomy. Eur Urol 1978;
131. Lerner SE, Hawkins CA, Blute ML, et al: Disease 4(1):13–17.
outcome in patients with low stage renal cell carcinoma 148. Saitoh H, Nakayama M, Nakamurak K, et al: Distant
treated with nephron sparing or dial surgery. J Urol metastasis of renal adenocarcinoma in nephrectomized
1996; 155:1868–1873. cases. J Urol 1982; 127(6):1092–1095.
132. Hafez KS, Fergary AF, Nocick AC: Nephron sparing 149. Russo P: Renal cell carcinoma: presentation, staging, and
surgery for localized renal cell carcinoma: impact of surgical treatment. Semin Oncol 2000; 27:160–176.
tumor size on patient survival, tumor recurrence and 150. Gill IS, McClennan BH, Kenbl K, et al: Adrenal
TNM staging. J Urol 1999; 162(6):1930–1933. involvement from renal cell carcinoma: predictive valve
133. Lee CT, Katz J, Shi W, et al: Surgical management of of computerized tomography. J Urol 1994;
renal tumors 4 cm or less in a contemporary cohort. 152:1082–1085.
J Urol 2000; 163:730–736. 151. Horan JJ, Robertson CN, Choyke PL, et al: The
134. Sagalowsky AK, Kadesky KT, Ewatt DM et al: Factors detection of renal carcinoma extension into the renal
influencing adrenal metastasis in renal cell carcinoma. vein and interior vena cava: a prospective comparison of
J Urol 1994; 151:1181–1184. venacavography and magnetic resonance imaging. J Urol
135. Sardock DS, Settel AP, Resnick MI: A new protocol for 1989; 142(4):943–948.
follow-up of renal cell carcinoma based on pathological 152. Ney C: Thrombosis of the interior vena cava
stage. J Urol 1995; 154:28. associated with malignant tumors. J Urol 1946;
136. Hatcher PA, Anderson EE, Paulson DF, et al: Surgical 55(6):583–590.
management and prognosis of renal cell carcinoma 153. Arkless R: Renal carcinoma: how it metastasizes.
invading the vena cava. J Urol 1991; 145:20–24. Radiology 1965; 84(3):496–501.
137. Gelb AB: Renal cell carcinoma. Current prognostic 154. Berg A: Malignant hypernephroma of the kidney, its
factors. Union Internationale Contre le Cancer (UICC) clinical course and diagnosis, with a description of the
and the American Joint Committee on Cancer (AJCC). author’s method of radial operative cure. Surg Gynecol
Cancer 1997; 80:981–986. Obstet 1913; 17(4):463–471.
138. Srigley JR, Hutter RV, Gelb AB, et al: Current 155. Hietala S, Ekelund L, JungGerg B: Venous invasion in
prognostic factors – renal cell carcinoma: work group renal cell carcinoma: a correlate clinical and radiologic
no. 4, Union Internationale Contre le Cancer (UICC) study. Urol Radiol 1998; 9(4):210–216.
194 Part III Kidney and Ureter

156. de Schepper A: “Nutcracker” phenomenon of the renal 166. Markoff DA, Thompson JA, Gold P, et al: Identification
vein and venous pathology of the left kidney. J Belge of Interleukin-2-induced complete response in metastatic
Radiol 1972; 55(5):508–511. renal cell carcinoma by FDG-PET despite radiographic
157. When M, O’Toole K, Bixon R, et al: The incidence of evidence suggesting persistent tumor. Am J Roentgenol
multifocal renal cell carcinoma in patients who are 1997; 169:1049–1058 (abstract).
candidates for partial nephrectomy. Urology 1995; 154:968. 167. Wu HC, Yen RF, Shen YY: Comparing whole body 18F-
158. Kletscher BA, Qian J, Bostwick PG, et al: Prospective 1-deoxyglucose positron emission tomography and
analysis of multifocality in renal cell carcinoma: influence technetium-99m methylene diphosphate bone scan to
of histological pattern, grade, number, size, volume and detect bone metastases in patients with renal cell
DNA ploidy. J Urol 1995; 153:904. carcinomas – a preliminary report. J Cancer Res Clin
159. Henriksson C, Geterwd K, Aldergorg F, et al: Bilateral Oncol 2002; 128(9):503–506.
asynchronous renal cell carcinoma: computed 168. Esrig P, Ahlering TE, Lieskovsky G, et al: Experience
tomography at the contralateral kidney 10–43 years after with fossa recurrence of renal cell carcinoma. J Urol
nephrectomy. Eur Urol 1992; 22:209. 1992; 147:1491–1494.
160. Zagoria RJ, Bechtold RE, Dyer RB: Staging of renal 169. Levy PA, Slaton JW, Swanson PA, et al: Stage specific
adenocarcinoma: role of various imaging procedures. Am guidelines for surveillance after radical nephrectomy for
J Roentgenol 1995; 184:363–370. local renal cell carcinoma. J Urol 1998; 159(4):1163–2267.
161. Fielding JR, Aliabadi N, Renshaw A, et al: Staging of 170. Kovick AC: Renal-sparing surgery for renal cell
119 patients with renal cell carcinoma: the yield and carcinoma. Urol Clin North Am 1993; 10:277–282.
cost-effectiveness of pelvic CT. Am J Roentgenol 1999; 171. Stormant TJ, Bilhartz DL, Zincke H: Pitfalls of berch
172(1):23–25. surgery and auto-transplantation for renal cell
162. Pretorius EJ, Wickstrom L, Siegelman S: MR imaging of carcinoma. Mayo Clinic Proc 1992; 62:621–628.
renal neoplasms. MRI Clin North Am 2000; 4:813. 172. Birnbaum BA, Bosniak MA, Megibow AJ, et al:
163. Glazer A, Novick AC: Preoperative transesophageal Observations on the growth of renal neoplasms.
echocardiography for assessment of resonance imaging. Radiology 1990; 176:695–701.
Urology 1997; 49:32–34. 173. Sheroy PP, Lakhkar BN, Ghosh MK, et al: Cutaneous
164. Itoh CK, Seltzer MA, Frank WJ, et al: Positron emission seeding of renal oncocytoma by Chiba needle aspiration
tomography in urologic oncology. J Urol 1998; biopsy. Case Report. Acta Radiol 1991; 32(1):50–52.
159:347–356.
165. Bachor R, Kotzerke J, Gottfried HW, et al: Positron
emission tomography in diagnosis of renal cell
carcinoma. Urologe A 1996; 35:146–150.
C H A P T E R

11 Renal Cell Carcinoma: Localized


Disease
Kenneth Ogan, MD, and Fray F. Marshall, MD

Renal cell carcinoma (RCC) is a unique neoplasm because American Cancer Society (ACS), based on the rates from
currently there is no effective systemic therapy once it has the National Cancer Institute’s (NCI) surveillance,
progressed beyond the kidney. Therefore, even more so Epidemiology and End Results (SEER) program, the
than in other malignancies, prompt and effective treatment estimated number of new cases and deaths from kidney
of localized RCC is of paramount importance. Over the cancer in the United States for 2004 was 12,480. Chow
past several decades, in part due to the diagnosis of smaller et al.8 have reported a rapidly increasing incidence of
tumors with improved imaging modalities, there has been RCC between 1975 and 1995 in the United States.
a paradigm shift in the treatment of localized renal tumors. While the greatest increase was seen for localized
First described by Robson et al.1 in 1968, open radical tumors, the incidence also increased for advanced
nephrectomy (ORN), including removal of the ipsilateral tumors. Thus, it is unlikely that this increased incidence
adrenal gland and an extended regional lymphadenectomy, is solely due to an increased detection rate, but it more
became the standard of care for surgical extirpation of renal likely represents a true increased incidence of RCC. The
tumors for the following several decades. In the 1980s, incidence rate in males is approximately twice that of
Hohenfellner, Novick and others initiated nephron-spar- females, with the peak age of diagnosis in the seventh and
ing surgery (NSS), removing the renal tumor with a mar- eighth decades of life. While Caucasians are affected
gin of normal parenchyma and sparing the remaining more commonly than African Americans, the incidence
normal parenchyma.2,3 While originally reserved for and mortality rates are increasing at a greater pace in the
patients that would be rendered functionally anephric with African-American population.8
radical nephrectomy, nephron-sparing surgical procedures In the past, presentation with the classic triad of flank
have been extended to the majority of patients with small pain, abdominal mass, and hematuria was rarely associ-
cortical renal tumors (<4 cm), even in patients with a nor- ated with localized disease. Due to the protected location
mal contralateral kidney. Over the past decade, the intro- of the kidney within the retroperitoneum, the majority of
duction of laparoscopic radical nephrectomy (LRN)4 and symptomatic tumors present at an advanced stage.
laparoscopic partial nephrectomy (LPN)5 has provided a Fortunately, in the current day of routine ultrasonogra-
minimally invasive treatment modality to the urologist’s phy (US), computed tomography (CT) imaging, and
surgical armamentarium for the treatment of localized magnetic resonance imaging (MRI), this presentation is
renal tumors. Finally, in an effort to even further reduce the rare, and in fact, the majority of tumors are discovered
morbidities of open and laparoscopic procedures, energy- incidentally.9 Incidental tumors have also been shown to
based ablative techniques have been developed in which be of a lower-stage than their symptomatic counter-
small renal tumors are destroyed and left in situ.6,7 parts.10,11 Thompson and Peek11 reported that 82% of
incidental tumors were stage I or stage II, whereas only
32% of symptomatic tumors were at early stage.
EPIDEMIOLOGY AND CLINICAL PRESENTATION
Consequently, following treatment these incidental
Malignant tumors of the kidney account for greater than tumors also demonstrated improved 10-year survival
2% of cancer incidence and mortality. According to the compared to the symptomatic tumors (90% versus 30%).

195
196 Part III Kidney and Ureter

Importantly, these smaller, lower-staged tumors are more cumulative survival compared to conventional clear cell
amenable to nephron-sparing and minimally invasive RCC.18 Collecting duct carcinoma is a rare type of RCC,
procedures. accounting for only 0.4% to 2.6% of renal neo-
plasms.14,19,20 These tumors are characterized by an
aggressive course with a high incidence of locoregional
CLASSIFICATION AND HISTOPATHOLOGY
spread and metastases.21 Similarly, Davis et al. were the
The majority of kidney tumors originating in the renal first to describe renal medullary carcinoma, which
cortex are adenocarcinomas, commonly called RCCs. appears to be a variant of collecting duct carcinoma. This
The first attempt to classify renal tumors was by type of RCC is unique because it is exclusively found in
Grawitz12 when he attributed the origin of renal tumors black patients with either sickle trait or sickle cell hemo-
to an ectopic adrenal rest and, therefore, called them globin disease.22,23 As with its collecting duct counter-
“hypernephroma.” Since that time numerous classifica- part, this is a very aggressive disease with a dismal mean
tion systems have been developed to characterize renal survival of 3.5±2.4 months following diagnosis.23–25
tumors. Currently, most people adhere to the classifica- Finally, sarcomatoid RCC is no longer considered as a
tion system developed in 1997 by the World Health distinct histopathologic category but rather an ominous
Organization (WHO) in combination with the Union subtype. In the largest series to date, Mian et al.26 from
Internationale Contre le Cancer (UICC) and the American M.D. Anderson Cancer Center reported a dismal median
Joint Committee of Cancer (AJCC) (Table 11-1),13 which overall survival of only 9 months in 108 patients classified
separates adult renal tumors into either benign or malig- with sarcomatoid RCC. Thus, it is apparent that the his-
nant categories. tologic type of RCC strongly affects prognosis and may
Of the malignant primary tumors of the kidney cortex, act as a guide to the institution of adjuvant systemic ther-
conventional clear cell carcinoma accounts for 70% to apies. Conversely, the future of classification or identifi-
80% of renal neoplasms.13,14 Papillary RCC is the second cation of renal tumors will most likely be based on
most common carcinoma, accounting for 10% to15% of molecular, rather than histopathologic findings.27
renal neoplasms.15,16 Compared to clear cell carcinoma,
papillary RCC has been associated with a better progno-
RISK FACTORS
sis. The overall 5-year survival rates for papillary com-
pared to conventional clear cell RCC have ranged The increased incidence of RCC has been noted across
between 82% to 90% and 44% to 54%, respectively.16,17 all stages and cannot be explained solely by the increased
Chromophobe RCC accounts for approximately 5% of detection of incidental tumors. RCC incidence rates
renal neoplasms. As with papillary RCC, chromophobe increased steadily between 1975 and 1995, by 2.3%
has been found to be associated with a better 5-year among white men, 3.1% among white women, 3.9%
among black men, and 4.3% among black women.8,28
While the greatest increase was seen for localized
Table 11-1 Classification of Renal Cell Carcinoma
tumors, there was also an increase in kidney cancer-spe-
Malignant neoplasms cific mortality during this time period. Thus, this sug-
Conventional (clear cell) carcinoma
gests that there is a true increased incidence of renal
cancer rather than just an increased rate of detection.
Papillary renal carcinoma Despite numerous epidemiologic studies, the etiology
of RCC remains largely unknown. There are definitive
Chromophobe renal carcinoma genetic, dietary, and environmental risk factors for the
Collecting duct carcinoma
development of RCC. Hereditary RCC syndromes place
family members at a tremendous increased risk of RCC
RCC unclassified and should likely prompt early screening. Table 11-2 lists
the most common inherited syndromes and the affected
Benign neoplasms chromosomes associated with increased risks of develop-
Oncocytoma
ing RCC.29 Also, acquired renal cystic disease secondary
to chronic hemodialysis has been associated with an
Papillary adenoma increased risk of RCC.30 Similarly, renal transplantation,
with its necessary immunosuppression, has been demon-
Metanephric adenoma strated to confer up to a 80-fold increased risk of devel-
oping RCC.31
From Storkel S, Eble JN, Adlakha K, et al: Classification of renal cell
carcinoma: Workgroup No. 1. Union Internationale Contre le Cancer There are numerous studies that consistently impli-
(UICC) and the American Joint Committee on Cancer (AJCC). cate smoking,32 hypertension,33 asbestos exposure,34
Cancer 1997; 80:987–989, with permission. petroleum exposure,35 and frequent analgesic use36 as
Chapter 11 Renal Cell Carcinoma: Localized Disease 197

Table 11-2 Inherited Syndromes with an Increased Risk of RCC


Inherited Disease Chromosome Characteristics

von Hippel-Lindau 3p25-p26 Hemangioblastoma of CNS/retina, pheochromocytoma, RCC


(clear cell type), renal cysts, pancreatic cysts, and cystadenomas

Hereditary papillary c-MET proto-oncogene 7q31 High incidence of bilateral papillary RCC
renal carcinoma

Tuberous sclerosis 1 TSC1 on 9q34 Facial angiofibromas, peringual fibromas, renal angiomyolipomas

Tuberous sclerosis 2 TSC2 on 16p13.3 Facial angiofibromas, peringual fibromas, renal angiomyolipomas

Birt-Hogg-Dube 17p12-q11.2 Fibrofolliculomas, different histologic renal tumors, and


pulmonary cysts associated with increased risk of pneumothorax

Modified from Godley P, Kim SW: Curr Opin Oncol 2002; 14:280–285, with permission.

TUMOR BIOPSY
risk factors for the development of RCC. There is con-
flicting evidence on the risks of specific dietary sub- While the majority of solid renal tumors represent RCC,
stances on the increased risk of RCC. However, obesity there are certain situations in that the preoperative diag-
and a high body mass index (BMI) have been consistently nosis is less certain. Review of large radical nephrectomy
noted in several studies to connote an increased risk of series reports benign lesions in up to 20% of cases. Thus,
RCC.37–39 in an effort to avoid needless nephrectomies for benign
tumors, the debate arises whether preoperative biopsy
is warranted in a proportion of these patients.
DIAGNOSIS
Percutaneous biopsies can be performed by fine needle
Localized RCC rarely causes symptoms or physical find- aspiration (FNA) or with hollow-bore needles under
ings on examination. What used to be called as the radiologic guidance. The FNA is less traumatic but must
“internist’s tumor” is now found more commonly by the be sent for cytologic versus pathologic analysis.
radiologist during evaluation of other ailments. Opponents of renal tumor biopsies note the risk of tumor
Radiologic imaging of the kidneys for a suspected renal spread due to needle tract seeding and the possibility of
tumor can be performed with ultrasonography (US), obtaining false negative biopsies. With newer biopsy
intravenous pyelography (IVP), CT, or MRI. needles, this occurrence is rare.
Angiography, which was routinely performed in the past, Wood et al.41 retrospectively reviewed 79 biopsies in
is currently a rare component of the radiologic evalua- 73 patients with renal masses. All tumors initially under-
tion. However, angiography may be a useful adjunct in went FNA (22 gauge) with immediate cytologic analysis,
preoperative planning prior to a complex partial nephrec- followed by core biopsy (17 to 20 gauge). Biopsy samples
tomy and during angioinfarction of large tumors. The were adequate for diagnosis in 74 of 79 (94%) cases. Of
advantages of ultrasound include low cost, low morbidity, 49 positive biopsies, 15 (31%) involved non-RCCs. Of
lack of potentially nephrotoxic contrast agents, and the the 79 biopsies, 5 (6%) were false negative and all corre-
ability to differentiate solid from cystic masses. These lated to samples with insufficient tissue for diagnosis.
characteristics make ultrasonography the imaging The ultimate diagnosis was made on repeat biopsy or at
modality of choice for a potential screening program. surgery. Biopsy results altered treatment in 32 (44%)
IVP has been the traditional imaging study of the upper patients who did not undergo surgery because of biopsy
urinary tract used in the evaluation of hematuria. The results. There were no major complications or tumor
main advantage of IVP is the ability to detect small track seeding reported at a mean follow-up or 30.4
urothelial tumors. However, due to similar costs, months. Finally, the efficacy and safety of FNA was com-
increased time to perform the study, and lack of detailed parable to that of core biopsy specimens.
cortical imaging, this study is being replaced by CT.40 Lechevallier et al.42 reported on 63 consecutive
The main advantage of MRI is the ability to determine patients that underwent 18-gauge core biopsies for suspi-
the cranial extent of tumor thrombus extension and the cious renal masses. Biopsy material was insufficient for
presence of contiguous organ involvement. Finally, analysis, suspicious for RCC and diagnostic in 15 (19%),
positron emission tomography (PET) is an emerging 2 (3%), and 56 (78%) of cases, respectively. Biopsy
technology with potential applications for staging and revealed a benign lesion in 8 (13%) patients and a non-
detection of tumor recurrences. surgical malignant tumor in 21 (33%) of patients. Thus,
198 Part III Kidney and Ureter

in theory, 29 (46%) of patients could have avoided sur- tumor-bearing kidney and adrenal gland within Gerota’s
gery because of biopsy results. Finally, for those patients fascia along with an extended lymph node dissection.
who had a nephrectomy (26 patients) for malignant dis- Until recently, ORN, as described by Robson, was the
ease, the accuracy of biopsy for histopathologic and standard of care for localized RCC. This “gold standard”
Fuhrman nuclear grade evaluations was 89% and 78%, has been challenged by the introduction of two concepts:
respectively. Importantly, there were no complications elective NSS for small renal tumors and laparoscopic
necessitating intervention from the biopsies and no renal surgery, including radical and partial nephrectomy.
evidence of tumor seeding at a mean follow-up of Elective NSS is now considered by many to represent the
29 ± 11 months. standard of care for the management of small renal
Finally, Harisinghani et al.43 looked at the accuracy of tumors, with 10-year cancer-specific survival rates similar
percutaneous renal biopsy of Bosniak III renal cysts, to radical nephrectomy.47 LRN was first performed by
which are diagnostic dilemmas for the surgeon and Clayman et al.4 in 1991 for a tumor-bearing kidney. Since
patient. In their series of 28 biopsied category III lesions, then, LRN has gained wide acceptance because of its
17 (60.7%) were malignant (16 RCCs and 1 lymphoma) decreased postoperative pain, decreased hospital stay and
and 11 (39.3%) were benign (6 hemorrhagic cysts, 3 shortened convalescence compared to the open
inflammatory cysts, 1 metanephric adenoma, and 1 cystic approach.48–50 Only recently a significant follow-up has
oncocytoma). Seventeen of the 28 lesions (16 RCCs and been made available to demonstrate the laparoscopic
1 inflammatory cyst) underwent surgical resection after approach to be oncologically equivalent to open surgery.
the biopsy. All resected lesions had pathologic diagnoses Finally, ablative technologies are coming to fruition, in
identical to the percutaneous imaging-guided biopsy that tumors are destroyed with heat or cold energy and
results. The remaining 11 patients, who did not have sur- then left in situ following treatment.
gery, had radiologic follow-up for a minimum of 1 year,
with benign lesions showing no interval change. Thus, in
OPEN RADICAL NEPHRECTOMY
these studies, surgery could have been avoided in 39% to
Technique
46% of cases, which begs the question of whether renal
biopsies should be performed more frequently in selected For details on “technique” see “Textbook of Operative
patients. Urology” by Marshall.51 Briefly, the kidney can be
approached through several different incisions (flank,
transperitoneal, thoracoabdominal), depending on the
INDICATIONS FOR SURGERY
size and location of the tumor, the patient’s body habitus,
Radiation and systemic therapy (chemotherapy or and the surgeon’s preference. Whichever approach is
immunotherapy) are both ineffective treatment of local- taken, early ligation of the renal pedicle and removal of
ized RCC. Similarly, the results of systemic treatment for the kidney and tumor with adequate margins are manda-
metastatic disease are poor. Thus, surgery is the treat- tory (Figure 11-1).
ment of choice for locally confined or locally advanced The flank approach is the most commonly performed
disease without regional or distant metastasis. There is incision for radical nephrectomy. For the flank incision,
recent evidence to suggest that surgical removal of the the patient is placed laterally on the operating table with
renal primary (cytoreductive surgery) in the midst of the table flexed and the kidney rest elevated. The incision
metastatic disease may also improve survival rates in is usually made over the 11th or 12th rib, and a rib may
combination with systemic immunotherapy.44,45 need to be removed for exposure of the kidney. The goal
Therefore, one could make the argument that any is to stay extraperitoneal, as well as extrapleural. Early
resectable renal tumor, regardless of size, or presence of identification, access and ligation of the renal hilar ves-
metastases, is a candidate for surgical removal if the sels, and avoidance of the intraperitoneal cavity are the
patient has a high performance status (PS). For those advantages of this approach. Disadvantages include risk
patients with especially poor prognostic factors, some of pneumothorax, which can be easily repaired without
investigators have suggested testing tumor responsive- leaving a chest tube if identified intraoperatively, and rel-
ness to immunotherapy prior to cytoreductive nephrec- atively increased pain compared to other incisions
tomy to avoid the morbidity of surgery in patients because of division of the flank muscles.
unlikely to respond to systemic treatment.46 The transperitoneal approach is accessed via a sub-
costal, midline upper abdominal or chevron incision.
The main advantages of the transperitoneal approach are
SURGICAL MANAGEMENT OF LOCAL DISEASE
the ability to examine surrounding intraabdominal struc-
Robson et al.,1 in 1968, were the first to describe their tures, including the contralateral kidney, and the
results of radical nephrectomy for RCC. In this classic improved access to the hilar vessels for larger tumors.
article, the authors describe the radical extirpation of the Disadvantages include poor exposure of the upper pole of
Chapter 11 Renal Cell Carcinoma: Localized Disease 199

Figure 11-1 Radical nephrectomy with division of hilar vessels.

the kidney and the increased risks of postoperative ileus (HAL). The original description by Clayman and col-
and adhesions. leagues4 was a transperitoneal approach. The retroperi-
Finally, the thoracoabdominal approach, while not toneal approach was developed to mimic the open flank
often necessary, arguably provides the optimal exposure nephrectomy and allow the hilar vessels to be accessed
for large upper pole tumors and tumor thrombus into the and controlled quickly without violating the peritoneal
vena cava. Entrance into the pleural cavity also allows for cavity. Lastly, hand-assisted nephrectomy was developed
concomitant metastasectomy of isolated ipsilateral pul- to facilitate the procedure for the novice surgeon, to
monary lesions. Disadvantages include the need for a provide an alternative to open conversion and for large
chest tube postoperatively and the risks of postoperative renal tumors and specimens, where HAL may help to
atelectasis. While it has been argued that the thoracoab- expedite the procedure. Surgeon’s preference, laparo-
dominal approach is associated with an increased risk of scopic procedure, patient anatomy, and history of prior
postoperative pain, Kumar et al.52 demonstrated no surgery all impact the approach that is taken, but the
increase in postoperative pain or analgesic requirements ultimate goal of all three is to perform an adequate
in patients with a thoracoabdominal versus a flank inci- oncologic operation while minimizing morbidity.
sion. Thus, there are numerous approaches to the kidney A preoperative consent form is signed, detailing the
for a radical nephrectomy, and the urologic surgeon must possible complications. Specifically for laparoscopic
be familiar with each to tailor the approach to best suit surgery, patients are instructed about the risks of open
the situation. conversion, hypercarbia, and neuromuscular injury
from prolonged positioning. For each of the three
techniques, bowel preparation for the day prior to sur-
LAPAROSCOPIC RADICAL NEPHRECTOMY
gery is optional to prevent colonic distension from
The three laparoscopic approaches to LRN are transperi- obscuring visualization. Patients are administered a
toneal, retroperitoneal, and hand-assisted laparoscopy second-generation cephalosporin intravenously and
200 Part III Kidney and Ureter

intraoperatively for routine prophylaxis. In case of the upper pole of the kidney is dissected free and the
rapid blood loss, the anesthesiologist must have ade- ureter and lateral attachments are divided to free the
quate intravenous access with 2 large bore IV lines and, entire kidney. The kidney is placed in a specimen retrieval
at the surgeon’s discretion, patients are typed and bag for intact removal or morcellation.
cross-matched for 2 units of packed red blood cells The major advantage of the transperitoneal approach
(RBCs). Of note, the anesthesiologist should be edu- is the increased working space and multiple landmarks to
cated not to use nitrous inhalant as it can cause bowel guide the surgeon. In addition, the transperitoneal
distention and that temporary oliguria with insufflation approach allows for extraction via a Pfannenstiel incision
is common and expected. providing an excellent cosmetic result. Disadvantages
include the need to reflect multiple abdominal structures
to gain access to the kidney, including the spleen, pan-
Technique
creas, colon, liver, and duodenum. Manipulation of the
Transperitoneal
bowel has been thought by some investigators to increase
A Foley catheter and orogastric tube are first placed the risks of postoperative ileus compared to retroperi-
while the patient is in the supine position. Patients are toneal approaches.
then positioned in a modified 45-degree flank position
with meticulous care taken to pad all pressure points.
Retroperitoneal
The kidney rest is not elevated, and the bed is minimally
flexed. The patients are then firmly secured to the oper- The preoperative preparation is the same for the
ating table with 3-inch cloth tape. Once secured, prior to retroperitoneal as the transperitoneal approach. As
covering the patient with sterile drapes, the bed is tilted described by Gill,53 the patient is placed in a 90-degree
completely to the right and left sides to assure that the full flank position with the table flexed and the kidney
patient’s position does not change. For emergent conver- rest elevated. Initial access is obtained through a 1 to
sion to an open procedure the patient is rotated on the 2-cm transverse incision located just below the tip of the
bed into a supine position to make a subcostal incision 12th rib. The tissue layers are bluntly dissected down to
and obtain immediate access to bleeding. the thoracolumbar fascia, which is incised sharply, gain-
Intraperitoneal access can be accomplished either with ing access to the retroperitoneum. Blunt finger dissection
the Veress needle or an open Hassan technique. is performed between the psoas muscle and the posterior
However, the authors use both; the Hassan technique is aspect of Gerota’s fascia. A balloon dilator (Origin
used exclusively in patients with multiple abdominal scars MedSystems, Menlo Park, CA) may be positioned into
or when the Veress technique fails. We typically gain this space and inflated to approximately 800 cm3 of air. A
access at the umbilicus where the abdominal wall is the laparoscope is then inserted into the transparent balloon
thinnest. The abdomen is temporarily insufflated to 20 to confirm adequate placement within the retroperi-
mm Hg for trocar placement under direct laparoscopic toneum. The balloon is removed, a 10-mm blunt trocar
visualization. We typically use a 3 or 4 port configuration is introduced through the skin incision, and the pneu-
for LRN. Once all trocars have been placed, the pneu- moretroperitoneum is established at 15 mm Hg. Two
moperitoneum is decreased to 15 mm Hg to reduce the additional trocars are placed under direct visualization at
possible complications of hypercarbia and decreased the lateral border of the erector spinae muscle just below
venous return. the 12th rib and between the mid- and anterior axillary
Initial dissection involves incising the posterior peri- line, 3 cm cephalad to the iliac crest.
toneum at the line of Toldt and reflecting the colon medi- Dissection begins with longitudinal incision of
ally to gain access to the retroperitoneum. In doing so the Gerota’s fascia anterior to the psoas muscle in the region
renocolic ligaments are divided to expose the kidney. On of the renal hilum. The renal artery is then easily visual-
the left side, the splenocolic ligaments are divided and ized, circumferentially dissected, clipped/stapled, and
care is taken not to injure the tail of the pancreas. On the divided. The renal vein is anterior to the artery, exposed
right side, the duodenum is reflected medially and the and divided using an Endo-GIA vascular stapler.
hepatocolic ligaments are divided to adequately expose Suprahilar dissection is performed along the medial
the renal hilum. The ureter is then identified and mobi- aspect of the upper pole. Finally, the ureter and gonadal
lized superiorly with the gonadal vessels. The renal hilum vessels are divided and the remainder of the kidney is
is identified and then the artery and vein are sequentially mobilized outside of Gerota’s fascia. As with the
divided using an Endo-GIA stapler (US Surgical, transperitoneal approach the specimen is placed in a
Norwalk, CT; Ethicon Endosurgical, Cincinnati, OH) or retrieval bag for either intact removal or morcellation.
Weck locking polymer clips (Weck Closure Systems, The proponents of retroperitoneal LRN have claimed
Research Triangle Park, NC). We no longer use a tita- that this approach is associated with less morbidity com-
nium disposable clip appliers to control the artery. Finally, pared to transperitoneal LRN secondary to rapid access
Chapter 11 Renal Cell Carcinoma: Localized Disease 201

to the hilum and avoidance of the peritoneal cavity. In the new hand devices protect the extraction wound with
theory, avoiding the peritoneal cavity should decrease the an impermeable barrier. Thus, there is no need for an
incidence of postoperative ileus and inadvertent injury to extraction bag device. There is no role for morcellation
intraperitoneal contents. McDougall et al.54 reported a since the hand port incision is large enough for almost all
faster return to oral intake in a retrospective comparison specimens.
of patients undergoing retroperitoneal versus transperi- Though not embraced by all surgeons, HAL has
toneal nephrectomy for benign disease. Conversely, gained popularity within the private sector secondary to
Gill et al.55 recently prospectively compared the out- the short learning curve and quick operative times. For
comes of transperitoneal versus retroperitoneal LRN in the “pure” laparoscopists, HAL may provide an alterna-
88 consecutive patients. Although the retroperitoneal tive to conversion. Finally, having the hand in the opera-
approach was associated with less surgical time, there was tive field may provide distinct advantages in patients with
no significant difference in blood loss, complications, large renal specimens or with prior abdominal surgery.
pain requirements, or hospital stay. The main theoretic Disadvantages to HAL include less working space sec-
disadvantage of this technique is the limited working ondary to the hand in the intraabdominal cavity. This is
space allotted. To the novice surgeon inexperienced with especially true in thin, small patients. The extraction site
this technique, this technique may initially make orienta- is limited to where the hand device is placed, typically
tion and dissection challenging. In addition, large renal lower midline, but rarely in a Pfannenstiel position.
specimens may be difficult to handle. Abbou et al.56 still Finally, similar to the transperitoneal technique, HAL
recommend limiting retroperitoneal nephrectomy to requires multiple abdominal structures to be reflected
T1 lesions. prior to exposing the kidney, which theoretically may
lead to an increased incidence of injury to adjacent
organs and prolonged ileus.
Hand-Assisted
Hand-assisted LRN is the latest addition to the laparo-
Comparison of Laparoscopic Radical Nephrectomy
scopic armamentarium. It has been criticized by “pure”
to Open Radical Nephrectomy
laparoscopists because of the presumed increased mor-
Intraoperative and Postoperative Parameters
bidity associated with the incision necessary for hand
insertion. However, Nelson et al.57 recently reviewed 22 In general, the benefits of LRN have been in patient
hand-assisted and 16 standard laparoscopic radical recovery from surgery. Numerous series have demon-
nephrectomies and demonstrated that there was no dif- strated that compared to ORN, laparoscopic patients
ference in pain scores, hospital stay, or return to normal incur less postoperative pain and need for analgesics,
activity. Importantly, the operative time was significantly shorter hospitalization, and a reduced convalescence.
less in the hand-assist group (205 versus 270 minutes) Intraoperatively, ORN is advantageous, with shorter
and less of a learning curve to overcome. operative times, but also with an increased blood loss.
The HAL technique utilizes the same presurgical Finally, opponents of laparoscopy criticize the increased
preparation as the transperitoneal technique, including costs associated with these procedures, secondary to
patient positioning. Typically the nondominant hand is costly equipment and prolonged operative times.
placed into the abdomen via an incision placed below the However, Lotan et al., in a cost comparison analysis of
umbilicus with 2 to 3 accessory trocars for laparoscopic LRN versus ORN, found that the laparoscopic approach
instruments. The initial steps of exposing the kidney are was actually less costly due to reduced hospitalization
identical to that described for the transperitoneal tech- costs secondary to shorter hospital stays. The laparo-
nique. The kidney is then mobilized except for the upper scopic approach, with its shorter convalescence, would be
pole, using both blunt and sharp dissection techniques. even more cost advantageous if cost from lost wages was
The ureter is divided prior to dividing the hilar vessels. included in the cost analysis (Table 11-3).
This allows the surgeons hand to grasp the kidney and
place the hilum on stretch. The fingers are then able to
Oncologic Adequacy
palpate, present, and if need be, compress the hilum dur-
ing dissection. The hilar vessels are separately controlled The goal of any laparoscopic procedure should be to
and divided using an Endo-GIA stapler (US Surgical, reproduce the efficacy of its open counterpart, while pro-
Norwalk, CT; Ethicon Endosurgical, Cincinnati, OH) or viding the benefits of the minimally invasive approach.
Weck locking polymer clips (Weck Closure Systems, One of the earliest surrogate endpoints to compare the
Research Triangle Park, NC). Finally, the upper pole oncologic adequacy of LRN to ORN was specimen
attachments are released, and if the situation dictates, the weight and margin status. The specimen weight follow-
adrenal gland is removed with the specimen en bloc. The ing LRN and ORN should be equivalent if the specimen
specimen is delivered through the hand incision. All of is removed intact and approximately 20% less if the
Chapter 11 Renal Cell Carcinoma: Localized Disease 203

Table 11-4 Laparoscopic Radical Nephrectomy Oncologic Results


Number Clinical Tumor Port Site or Renal 5-Year Cancer Mean Follow-Up
Study of Patients Stage Fossa Recurrence Specific Survival (%) (months)

Cadeddu et al.62 157 T1-2,N0,M0 0 91 19.2

Dunn et al.48 61 T1-2,N0,M0 0 — 25

Gill et al.59 53 T1-2,N0,M0 0 — 13

Chan et al.63 67 T1-2,N0,M0 0 95 35.6

Portis et al.60 64 T1-2,N0,M0 1 98 54

Ono et al.58 147 T1-2,N0,M0 — 96 30 (median)

ogy, grade, and stage were accurately determined follow- stay. However, Gettman et al.77 showed no significant
ing morcellation. Only tumor size was not assessed. differences in subjective pain and activity scores, and
However, as noted, with modern imaging tumor size can time to return to normal activity. Ono et al.58 also
be accurately measured preoperatively and incorporated demonstrated no significant differences in analgesic
into the tumor staging process. Meng et al.74 found that requirements (29 mg versus 29 mg analgesics) and con-
india ink covering of the specimens prior to morcellation valescence (22.7 days versus 23.3 days) in the intact
allows for accurate determination of surgical margins. extraction group and morcellated group, respectively.
Therefore, accurate staging of morcellated renal tumors These conflicting results call into question the benefits
is possible following morcellation and should not be a of specimen morcellation. This in combination with
concern. prolonged operative times, expenses associated with
It is well established that specimen morcellation is a morcellation and risk of tumor spillage; all combine to
safe alternative to intact removal as long as the morcel- make intact specimen removal our technique of choice.
lation is performed in an impermeable entrapment sack.
Urban et al.75 tested the permeability of 18 LapSacs
(Cook Urologic, Spencer, IN) following porcine laparo- TUMOR THROMBUS
scopic nephrectomy or nephroureterectomy with a Venal caval tumor thrombus has been reported in 4% to
high-speed electrical tissue morcellator. None of the 10% of all patients with RCC.78–80 Venous extension is
sacks demonstrated permeability to bovine serum albu- classified by the cranial extent of the tumor thrombus.
min, indigo carmine or mouse bladder tumor cells fol- While there are numerous classification schemes avail-
lowing morcellation. These results were confirmed by able, the one proposed by Skinner et al.80 is simple to use
Landman et al.73 in 14 of 15 LapSacs that remained and has implications on the surgical approach and ulti-
impermeable following morcellation. The one case of mate prognosis (Table 11-5).
perforation occurred with a specimen that had been In 1913, Berg et al.81 were the first to remove an intra-
preserved in formalin. Clinically, Dunn et al.48 reported caval thrombus in a patient with RCC. Sosa et al.82
a series of 39 cases where the specimen was morcellated reported dismal results with no survivors at 1 year in
with no local or port site recurrences over 9 years. patients undergoing nephrectomy alone, without
However, Fentie et al.72 have reported a case with a removal of the vena caval thrombus. Conversely, numer-
clinical stage T3 tumor with an isolated port site recur-
rence following laparoscopic nephrectomy and morcel-
lation. Thus, while morcellation has been demonstrated Table 11-5 Classification of superior extent of RCC
to be safe, meticulous care must be taken to avoid tumor in the IVC
contamination during morcellation. In particular, to Level 3 Level 2 Level 1
avoid entrapment sac violation and tumor spillage,
laparoscopic visualization during morcellation is Intra-atrial Extension within Extension below the
tumor the intrahepatic insertion of the
mandatory.
extension IVC but not into hepatic veins
Whether there is a clinical advantage to morcellation the atrium
remains to be determined. Walther et al.76 compared
specimen morcellation to intact removal and described From Skinner DG, Pritchett TR, Lieskovsky G, et al: Ann Surg 1989;
decreased analgesic requirements and a shorter hospital 210:387–392. (Discussion 392–394.), with permission.
204 Part III Kidney and Ureter

ous investigators have demonstrated long-term survival warranted in patients with tumor thrombi, regardless of
rates with removal of tumor thrombus at the time of rad- cranial extent, without evidence of metastatic disease.
ical nephrectomy, even with cranial extension into the
atrium (Figure 11-2).
ROLE OF ADRENALECTOMY
There is considerable debate as to whether the pres-
ence of tumor thrombus independently portends a poor The adrenal gland is easily removed en bloc using an
prognosis. Numerous reports have demonstrated ORN or LRN technique. However, most LRN series
encouraging 5-year survival rates of 32% to 64% in reserved ipsilateral adrenalectomy for patients with evi-
patients undergoing radical nephrectomy with removal dence of adrenal involvement on preoperative studies or
of inferior vena caval thrombi.80,83,84 The survival rates large upper pole lesions where removal was required to
decrease dramatically when tumor thrombus is associated ensure a negative margin.62 The major report that sup-
with positive lymph nodes, distant metastasis and when ports this practice comes from Tsui et al.,86 in that the
the tumor thrombus invades into the wall of the inferior records of 511 patients undergoing ORN with ipsilateral
vena cava (IVC). The prognostic significance of the cra- adrenalectomy were reviewed. The incidence of adrenal
nial extent of tumor thrombus has also been the point of metastasis was 5.7%. Importantly, tumor stage correlated
contention. Skinner et al.,80 in a series of 53 patients, with the probability of adrenal involvement, with T4,
demonstrated 5-year survival rates for subhepatic, T3, and T1 to T2 tumors accounting for 40%, 7.8%, and
hepatic, and atrial thrombi of 35%, 18%, and 0%, 0.6% of cases, respectively. Also, preoperative CT imag-
respectively. Conversely, Staehler et al.85 found no differ- ing demonstrated 99.6% specificity and a 94.4% negative
ence in survival rates among patients with levels I to IV predictive value. Similarly, Paul et al.87 evaluated 866
thrombi. Therefore, an aggressive surgical approach is consecutive patients undergoing ORN with ipsilateral

Figure 11-2 Tumor thrombus with vena caval extension into the atrium.
Chapter 11 Renal Cell Carcinoma: Localized Disease 205

adrenalectomy. In their series, a total of 27 (3.1%) of lymph nodes draining the kidney, an extended lym-
adrenal metastasis were found. Of the 27 patients, 21 had phadenectomy would be necessary to potentially remove
multiple metastases at diagnosis and only 6 (0.7%) pre- all involved lymph nodes.
sented with solitary ipsilateral adrenal metastasis. Regional lymph node dissection for the treatment of
Univariate and multivariate analysis revealed tumor size RCC may be unnecessary in patients with no evidence of
and M stage as best predictors of adrenal involvement. lymph node enlargement on preoperative studies or dur-
They concluded that adrenalectomy was not necessary if ing surgery. Minervini et al.90 showed no difference in
the tumor was less than 8 cm in size on preoperative CT 5-year survival between a group of 108 patients who
imaging and there was no evidence of metastasis or underwent radical nephrectomy alone versus 49 patients
extension from adjacent renal tissue. who underwent radical nephrectomy plus a regional
lymph node dissection. Of the 49 patients with no suspi-
cion of lymph node involvement, only 1 (2%) had a his-
ROLE OF LYMPHADENECTOMY
tologically confirmed positive node. This is in agreement
The incidence of lymph node metastasis in RCC ranges with the results of the EORTC Genitourinary Group in
from 13% to 32%, with increasing incidence with higher that only 1.0% of patients with no suspicion of involve-
stage tumors.88 The regional nodal drainage is different ment had nodal disease.88 There is often variability in the
in the right and left sides.89 The left kidney most often surgical dissection and pathologic examination of nodal
drains to the para-aortic nodes in the lumbar region, tissue. In patients with no evidence of nodal enlargement,
whereas the right kidney drains primarily to the interaor- a regional lymph node dissection (open or laparoscopic)
tocaval and paracaval nodes.89 Figure 11-3 demonstrates is not frequently beneficial.
the borders of a lymph node dissection. Regretfully, sec- Whether patients with evidence of suspicious lymph
ondary to the frequently aberrant drainage pattern of the nodes garner any benefits from an extended dissection at

Figure 11-3 Lymph node dissection for RCC.


206 Part III Kidney and Ureter

the time of nephrectomy also remains controversial. prior to immunotherapy versus immunotherapy alone in
Improved pathologic staging does provide information patients with metastatic RCC.105 They found that while
for guiding subsequent systemic immunotherapy. there was no significant difference in the radiologic
However, the data demonstrating improved survival rates response rates of metastatic deposits between the two
in patients undergoing lymphadenectomy are based groups, the nephrectomy group had a statistically signif-
solely on nonrandomized retrospective studies,91,92 with icant improved survival (medial survival of 11.1 versus 8.1
multiple studies arguing against its utility.93,94 The only months). Importantly, there was only one operative mor-
prospective trial comparing radical nephrectomy with tality, low operative morbidity (4.9%), and negligible
and without lymphadenectomy (EORTC 30881) demon- delay to the institution of immunotherapy (median 19.9
strated no increase in morbidity with lymphadenectomy, days). Similarly, the EORTC Genitourinary Group
but the results are too premature to determine any sur- prospectively demonstrated a delay in time to disease
vival benefits.88 Importantly, a complete laparoscopic progression and an improved median survival in patients
lymph node dissection can be performed if the surgeon treated with cytoreductive nephrectomy prior to sys-
desires. Several investigators reporting their results with temic immunotherapy.45 The only criticism with these
laparoscopic retroperitoneal lymph node dissections for two studies is that they lack a group of patients that only
teste tumors have validated this.95,96 received nephrectomy, without immunotherapy. Thus, it
is impossible to assess the true benefit of immunotherapy
in the patients’ improved survival.
ROLE OF NEPHRECTOMY WITH ADVANCED
Finally, to avoid the potential surgical morbidity of
DISEASE
nephrectomy in patients who will not respond to
While RCC is being diagnosed at an earlier stage, immunotherapy, several investigators have proposed
approximately 30% of patients still present with metasta- neoadjuvant immunotherapy, and reservation of surgery
tic disease, and 30% to 50% of initially localized tumors only for patients that demonstrate a response.106,107
eventually progressing to metastatic disease.91,97 RCC is Rackley et al.108 demonstrated a slightly higher objective
a unique cancer, in that removal of the primary tumor has response rate and longer median survival rates for
been associated with rare spontaneous regression of patients treated with initial immunotherapy followed
metastasis.98 However, review of this phenomenon has by surgery compared to patients who underwent pri-
demonstrated an overall incidence of only 0.7% com- mary nephrectomy and adjuvant immunotherapy.
pared to a mortality rate with nephrectomy of between Krishnamurthi et al.46 reported on a selected population
1% and 5%.99,100 Thus, nephrectomy alone in the face of of 14 patients with metastatic RCC who underwent ini-
metastatic disease is not generally curative and must be tial immunotherapy followed by surgical resection of pri-
considered palliative. The debate arises whether mary and metastatic lesions. All 14 patients either had an
nephrectomy with or without metastasectomy in combi- objective response to immunotherapy (9) or stable
nation with adjuvant immunotherapy improves response disease (5) following immunotherapy. Their 3-year can-
rates, and ultimately, patient survival. cer-specific survival rate was an impressive 81.5%, sug-
Surgical removal of the primary tumor along with iso- gesting that patients that respond favorable to initial
lated metastatic lesions has been demonstrated to be immunotherapy may represent a biologically more favor-
effective for colon cancer metastatic to the liver.101 able group.
Numerous retrospective studies have suggested a survival
benefit for surgical extirpation of metastatic RCC
PARTIAL NEPHRECTOMY (OPEN OR
lesions. van der Poel et al.102 reported on 152 resections
LAPAROSCOPIC)
of RCC metastases performed in 101 patients. Patients
demonstrated a median survival of 28 months after the The first report of a partial nephrectomy was by Wells in
initial metastasectomy. Improved survival correlated with 1884 when he described this technique for a perirenal
pulmonary metastases and a tumor-free interval of more fibrolipoma.109 NSS was initially proposed for the surgi-
than 2 years between primary tumor and metastasis. cal management of patients in that radical nephrectomy
Surprisingly, the number of metastases and additional would render them functionally anephric. This consisted
immuno- or radiation therapy did not influence survival. of patients with bilateral renal tumors, tumors in a soli-
Numerous studies have suggested that cytoreductive tary kidney, and patients with preexisting renal insuffi-
nephrectomy prior to immunotherapy for unresectable ciency. Due to the excellent results seen in these patient
metastatic RCC may improve response rates.103,104 populations, the indications for NSS have expanded to
Regretfully, these studies are retrospective and nonran- include all patients with small renal tumors (<4 cm).
domized, making them subject to negative selection bias. Fergany et al.47 reported on their long-term results of
Recently, however, the SWOG conducted a randomized NSS for sporadic RCC in 107 patients treated with par-
prospective study examining the role of nephrectomy tial nephrectomy prior to 1988. They reported 5- and
Chapter 11 Renal Cell Carcinoma: Localized Disease 207

10-year cancer-specific survival rates of 88.2% and 73%, negative margin, the extent of peritumoral normal
respectively. Isolated recurrences in the ipsilateral kidney parenchyma excised is not significant.
were seen in only 2 patients (4%) at a mean follow-up of
104 ± 57 months. These results are even more impressive
RENAL TUMOR ABLATION
considering that tumors were symptomatic in 68%
of cases, bilateral in 50%, and more than 4 cm in 45%. With the advent of improved imaging modalities, the
A subset of patients (29) that met currently accepted vast majority of renal tumors detected are of low grade
indications for elective NSS (unilateral tumors <4 cm) and stage. In the past, options were limited to observa-
demonstrated a 100% cancer-specific survival at 10 years. tion or partial/radical nephrectomy. While tumors less
Importantly, although almost 40% of patients had preop- than 3-cm rarely metastasize, the natural history of RCC
erative renal insufficiency (creatinine ≥1.5 mg/dl), renal is unpredictable and observation may not be in the best
function was preserved in the majority of patients, with interest of the patient for a disease with no effective sys-
100 patients (93%) maintaining adequate renal function temic treatment.117,118 Thus, ablative therapy provides a
to avoid dialysis. minimally invasive modality for the treatment of these
small tumors that minimizes the risks and morbidity tra-
ditionally associated with open or LPN.
Technique
Open3 and LPN adhere to the same operative principle;
Cryoablation
removal of the tumor with a margin of normal
parenchyma. The main complications of this procedure Cryoablation involves the destruction of cells by rapid-
are bleeding and urinoma formation. Bleeding can be min- freeze thaw cycles, with complete necrosis of renal
imized with hilar clamping or with the application of parenchyma occurring at temperatures of −19.4 ˚C or
parenchymal compression and renal injury can be reduced lower.119 Delworth et al.120 first described open renal
through cold ischemia. Urinoma formation can be avoided cryoablation of an RCC in 1996, and since then it has
through direct suture repair of any entry into the collect- been performed by several investigators laparoscopi-
ing system. Due to the inherent limitations of intracorpo- cally6,121 and percutaneously.122 Gill et al.7 and
real suturing and renal cooling, numerous adjunctive Shingleton and Sewell123 recently reported on two large
techniques have been developed to facilitate LPN.110–113 series of laparoscopic and percutaneous renal cryoabla-
Nonetheless, this is an advanced laparoscopic procedure, tion, respectively.
which is best reserved for smaller exophytic tumors. With Gill et al.7 reported short-term follow-up of 32 patients
the advent of improved instrumentation and experience undergoing laparoscopic renal cryoablations who had exo-
with the laparoscopic approach, Gill et al.114 have reported phytic enhancing renal masses less than 4 cm in size.
on an increasing experience treating larger, more complex Anteriorly located tumors were approached via a
central tumors, mimicking the open technique. transperitoneal route, while posterior and lateral tumors
were approached retroperitoneally. Once exposed and ver-
ified with color Doppler ultrasound, the renal tumor was
Surgical Margin
biopsied prior to cryoablation. Under direct laparoscopic
Historically, the surgical dictum has always been to excise and real-time ultrasonic visualization the tumor was punc-
at least a 1-cm margin of normal peritumoral renal tured with a 4.8-mm cryoprobe, and the tumor was treated
parenchyma during partial nephrectomy to decrease the with a double freeze–thaw cycle. Treatment was stopped
risk of local tumor recurrences. This principle was based on when the ice ball was seen to extend approximately 1 cm
scant evidence and has recently been challenged. Lerner beyond the edge of the tumor. Follow-up imaging con-
et al.115 reported similar local recurrence and survival rates sisted of MRI scans obtained on postoperative day 1 and
in patients that underwent tumor enucleation compared to then at 1, 2, 3, 6, and 12 months. CT-guided core needle
standard partial nephrectomy. Sutherland et al.116 reported biopsies were obtained at 3 or 6 months.
on 41 patients, with a mean postoperative follow-up of All procedures were performed laparoscopically with-
49 patients, who underwent partial nephrectomy for small out the need for open conversion. Mean tumor size by
renal tumors (mean size = 3.2 cm). With a median surgical intraoperative ultrasound was 2 cm with an average blood
margin of only 0.2 cm (range 0.05 to 0.70), no patient with loss of 66.8 ml (range 10 to 200). There were no major
a negative parenchymal surgical margin had a local recur- intraoperative complications and two minor postopera-
rence at the resection area. Thus, for low stage tumors, tive complications that did not require surgical interven-
margin size was irrelevant as long as the surgical bed was tion. Among 20 patients imaged with MRI at 1 year,
free of residual tumor. Importantly, multiple biopsies from 5 patients had no cryolesion identified and 15 patients
the bed of the tumor resection should be performed to con- had a 66% reduction in cryolesion size. None of the
fer margin status. These results suggest that in lieu of a 23 patients who had CT-guided biopsies at 3 or 6 months
208 Part III Kidney and Ureter

had evidence of residual cancer on histopathologic analy- Recently, Shingleton and Sewell125 reported on
sis. The authors noted that while CT-guided biopsies longer-term follow-up of their series. A total of 65
have a reported false-negative rate of 16%,124 the absence patients were treated with percutaneous cryoablation and
of cancer on all of the 23 biopsies is encouraging. followed for a mean of 18 months (range 2 to 30). While
However, they appropriately cautioned that long-term 9 patients required more than one treatment for com-
follow-up is warranted to assess the true efficacy of plete ablation, 60 patients were alive without evidence of
cryoablation for small renal tumors. local disease recurrence at last follow-up. As longer fol-
Shingleton and Sewell123 initially reported their series low-up becomes available we will be better able to assess
of 20 patients treated with MRI-guided percutaneous the true efficacy of this technology (Table 11-6).
cryoablation with a mean follow-up of 9.1 months (range
3 to 14). For this procedure, a high Telsa open MRI unit
Radiofrequency Ablation
is necessary. The patients were administered a general
anesthetic (18) or intravenous sedation (2) and placed Much of the experience using radiofrequency (RF)
prone on the interventional MRI docking table. Axial fast energy for ablative therapy has come from the treatment
spin echo images were obtained to determine probe entry of liver tumors.131,132 Zlotta et al.133 were the first to
site and angle of insertion. The cryoprobe, which is 2 or describe the effects of RF ablation on human renal
3 mm in diameter and 15 cm in length (Galil Medical, Tel tumors prior to nephrectomy in 1997. Since then, RF
Aviv, Israel) was then activated for a total of 3 energy has been utilized both laparoscopically111 and
freeze–thaw cycles. Periodic reimaging was performed to percutaneously130,134 for the treatment of small renal
monitor ice ball propagation and to ensure treatment 5 tumors. RF energy employs alternating electric current
mm beyond the edge of the mass while avoiding damage to agitate tissue ions in proximity to the probe, resulting
to the collecting system and surrounding structures. in frictional heating of the tissue around the RF elec-
Following probe removal after the third cycle, the access trodes. Heating of tissues to 50 to 55 ˚C for 4 to 6 min-
sheath was packed with absorbable knitted fabric or utes produces irreversible cell damage while
absorbable gelatin sponge pledgets to facilitate hemosta- temperatures between 60 and 100 ˚C result in tissue
sis. Follow-up included MRI or CT scans at 1 week, and coagulation and almost instantaneous cell death.135
1, 3, 6, and 12 months. Walther et al.136 demonstrated necrosis in 10 of 11
The mean procedure time was 97 minutes (range 56 to tumors treated intraoperatively by RF ablation prior to
172) and there were no intraoperative complications. surgical removal.
The only postoperative complication was a wound Gervais et al.134 were the first to report their experi-
abscess. On follow-up CT scan or MRI imaging, only ence with percutaneous RF ablation of 9 tumors in
one mass of the 22 treated demonstrated continued con- 8 patients. The mean tumor size was 3.3 cm (range 1.2 to
trast enhancement. This patient was subsequently 5.0) and all tumors showed contrast enhancement on
retreated and 6-month follow-up imaging showed no preablation CT or MRI imaging. Seven of 9 tumors
residual enhancement. Overall 20 of the 22 masses either treated had biopsy proven RCC prior to ablation.
completely resolved or decreased in size. While routine Patients were anesthetized with intravenous sedation,
post ablation biopsies were not performed, two core and tumors were localized with ultrasound or CT guid-
biopsies of tumors with rim enhancement were both neg- ance. RF ablation was performed with an RF generator
ative for malignancy. As with Gill’s series, the mean fol- (Cosman Coagulator CC-1; Radionics, Burlington, MA)
low-up was short at 9.1 months (range 3 to 14). and single (one 2.0 to 3.0-cm tip) or cluster of (three 2.5-

Table 11-6 Clinical Tumor Ablation Series


Mean Tumor Successful Mean Follow-up
Author Technique Patients Size (cm) Ablation* (months)

Sung et al.126 Laparoscopic cryoablation 50 2.1 97% (30/31)† 18.8

Shingleton et al.127 Percutaneous cryoablation 35 3.7 86% (30/35) 12

Jacomides et al.128 Laparoscopic RF ablation 11 2.1 100% (11/11) 9.8

McGovern et al.129 Percutaneous RF ablation 17 1–5.5 84% (15/18) 6–36

Pavlovich et al.130 Percutaneous RF ablation 21 2.4 79% (21/24) 2.0

*Successful ablation defined as no contrast enhancement on follow-up CT imaging.


†Negative CT-directed biopsy at 3–6 months.
Chapter 11 Renal Cell Carcinoma: Localized Disease 209

cm tip) cooled tip electrodes. Once the electrodes were demonstrate persistent contrast enhancement, 4 did not
appropriately positioned, the tumor was treated for reach target treatment temperature at each of the elec-
12 minutes at a current of 1500 to 1800 mA. The elec- trodes for the entire treatment period. While results are
trodes were repositioned in larger tumors to ensure com- promising, 79% successful ablation is less than ideal.
plete tumor ablation, and additional treatments at a Recently, we have reported successful RF ablation in 12
separate setting were necessary in 4 patients (total of of 13 tumors (93%) at a short-term follow-up of only 4.9
14 treatments). Unlike cryoablation, real-time ultra- months.
sound imaging is not effective in monitoring the RF Laparoscopic RF ablation can also be performed for
lesions acutely.137 Therefore, the only true measure of renal tumors that are not amenable to percutaneous abla-
treatment success is diligent follow-up imaging with or tion or for tumors that cannot be safely treated with LPN
without image-guided biopsies. Gervais et al.134 per- because of location. Yohannes et al.138 were the first to
formed CT or MRI at 1, 3, and 6 months, and then every describe retroperitoneoscopic RF ablation of a solid renal
6 months thereafter. mass in an 83-year old man. The tumor was 2.0 cm in
Percutaneous RF treatments are well tolerated, and 12 size and located on the anterior surface of the kidney.
of the 14 treatments reported by Gervais et al.134 were Due to extensive comorbidities and renal insufficiency,
conducted on an outpatient basis. There was one compli- laparoscopic RF ablation was performed rather than par-
cation attributable to the RF ablation in a patient that tial nephrectomy. The advantages of the laparoscopic
sustained a large paranephric and renal pelvis hematoma approach are that it allows biopsy of the renal mass and
necessitating a ureteral stent and blood transfusion. overlying fat prior to ablation, and also provides direct
Follow-up imaging at 6 months demonstrated no tumor visualization of the surrounding structures minimizing
enhancement in all 5 exophytic tumors and all 3 tumors potential injury.
less than 3 cm. Conversely, 2 of the 3 central tumors Jacomides et al.128 have performed laparoscopic
measuring 4.4 and 5.0 cm, respectively, demonstrated transperitoneal RF ablation of 11 tumors in 8 patients.
persistent contrast enhancement necessitating repeat The mean tumor size was 2.1 cm (range 1.0 to 3.6) with
treatments. While these short-term results (mean follow- an average blood loss of only 44 cc (range 20 to 100).
up = 10.3 months) are encouraging, the authors recom- Biopsy of the tumors following ablation revealed RCC in
mend this treatment only for exophytic tumors less than 6 patients, acute myelogenous leukemia (AML) in
3 cm in size. 1 patient and 4 oncocytomas in a single patient who
Pavlovich et al.130 reported their experience at The underwent multiple ablations. The only complication
National Cancer Institute with percutaneous renal was transient elevation in serum creatinine to 1.8 mg/dl
tumor ablation in patients with either von Hippel- in the patient with multiple ablations. Follow-up CT
Lindau clear cell tumors or hereditary papillary RCC. imaging at 6 weeks lack of contrast enhancement in all of
Patients with CT scan demonstrating solid renal tumors the tumors treated. The advantages of the laparoscopic
3 cm or less than enlarged during the previous year were approach are that anterior tumors can be safely treated,
considered candidates for ablative therapy. In conjunc- surrounding structures can be directly visualized and
tion with an interventional radiologist, the authors local- avoided, and adequate tissue specimens can be obtained
ized and targeted the tumors with either ultrasound for pathologic analysis. There have been reports of vary-
or CT guidance. Then under intravenous sedation or ing efficacy of RF, but this likely represents engineering
general anesthesia, the RITA probe (RITA Medical and technical problems rather than a primary deficiency
Systems, Mountain View, CA) was positioned into the of RF.
tumor and the electrodes deployed until the tumor vol-
ume was completely covered. The tumors were then
High-Intensity Focused Ultrasound
treated to a target temperature of 70 ˚C for two or three
10 to 12-minute cycles, depending on the size and loca- High-intensity focused ultrasound (HIFU) is a nonin-
tion of the tumors. vasive technique of delivering ultrasonic energy to a
A total of 21 patients underwent RF ablation of 24 specific area within the body. The ultrasound energy
tumors. The mean diameter of the tumors treated was can be focused into a small volume (1 cc) so that tem-
2.4 cm (range 1.5 to 3.0). All patients tolerated the pro- perature at the treatment site reaches 90 ˚C and the
cedure and were medically stable for discharge from the intervening tissue between the transducer and target is
hospital within 24 hours. There were no major compli- theoretically unaffected. As with the other ablative
cations, though 2 patients suffered flank pain on ipsilat- technologies, the cells at the treatment site undergo
eral hip flexion and 2 complained of skin numbness on coagulative necrosis and cell death. HIFU was first
the ipsilateral flank. Follow-up CT imaging at 2 months investigated as a nonsurgical treatment for experimen-
revealed absence of contrast enhancement in 19 of the 24 tal liver tumors in the rabbit model.139 Soon afterward,
(79%) tumors treated. Of the 5 tumors that continued to Foster et al.140 investigated the creation of prostatic
210 Part III Kidney and Ureter

lesions in the canine prostate with transrectally deliv- Table 11-7 2002 TNM Classification
ered HIFU. Subsequently, HIFU has been utilized
Primary Tumor (T)
clinically for the treatment of benign141 and malignant
diseases142 of the prostate. Tx: Primary tumor cannot be assessed
Chapelon et al.143 were the first to explore the tissue
effects of transcutaneous delivery of HIFU to kidneys T0: No evidence of primary tumor
in the canine model. Adams et al.144 subsequently
T1: Tumor 7 cm or less in greatest dimension, limited to
investigated the feasibility of treating renal tumors with the kidney
HIFU in the rabbit model. In their study, renal tumors
were induced and then treated with HIFU by placing T2: Tumor extends more than 7 cm in greatest dimension,
the transducer either on the kidney or transcuta- limited to the kidney
neously. With the transducer on the kidney surface
T3: Tumor extends into major veins or invades adrenal
opposite the tumor location, the HIFU was able to cre-
gland or perinephric tissues but not beyond Gerota’s
ate well-defined areas of necrosis of the tumor and fascia
renal parenchyma. Transcutaneous delivery resulted in
less well-defined treatment areas, with only 7 of 9 rab- T3a: Tumor directly invades adrenal gland or perirenal
bits demonstrating ablation of the tumors. Four of the and/or renal sinus fat but not beyond Gerota’s fascia
rabbits suffered 1 cm skin burns in areas where the
probe contacted the skin. Of note, there were no T3b: Tumor grossly extends into the renal vein or its
segmental (muscle-containing) branches, or vena cava
injuries to adjacent organs. Similar problems of incon-
below the diaphragm
sistent treatment zones and injuries to the skin have
been noted in transcutaneous HIFU in the porcine T3c: Tumor grossly extends into vena cava diaphragm or
model.145 invades the wall of the vena cava
In a phase 1 study in humans, Vallancien et al.146
treated 4 patients with T2 and T3 kidney tumors with T4: Tumor invades beyond Gerota’s fascia
pyrotherapy (HIFU) 2, 6, 8, and 15 days prior to Regional Lymph Nodes (N)
nephrectomy. In all cases, histology demonstrated coag-
ulative necrosis in the targeted tumor areas. As in the Nx: Regional lymph nodes cannot be assessed
previous animal studies, there was one case of a superfi-
cial skin burn attributed to an error in dosimetry calcula- N0: No regional lymph node metastases
tion. While HIFU represents the ultimate in minimally N1: Metastases in a single regional lymph node
invasive treatment of small renal tumors, the problems of
imprecise targeting and thermal burns to the overlying N2: Metastasis in more than one regional lymph node
skin have prevented its widespread use. The Cyberknife,
which has been extensively used for the treatment of Distant Metastasis (M)
brain tumors, is an additional extracorporeal means of Mx: Distant metastasis cannot be assessed
renal tissue ablation currently being evaluated in animal
models.147 M0: No distant metastasis

M1: Distant metastasis


PROGNOSTIC FACTORS
From Javidan J, Stricker HJ, Tamboli P, et al: Prognostic significance
A prognostic factor is a marker that can be used to of the 1997 TNM classification of renal cell carcinoma. J Urol 1999;
determine progression of a disease or its arrest. 162:1277–1281, with permission.
Reliable prognostic factors for RCC would allow
physicians to avoid ineffective treatment and enroll
patients earlier in either conventional treatments or RCC. In 1997, the classification system was modified so
therapeutic trials. A recent review article by Mejean that the transition point between T1 and T2 tumors was
et al.148 separated prognostic factors into three sepa- changed from 2.5 cm to the current size of 7 cm. This
rated subgroups of those associated with the tumor, the change was initially undertaken because multivariant
patient, and the treatment. analysis demonstrated improved survival stratification
Tumor stage, which reflects the anatomic extent of the with the 7 cm cutoff.149,150 Subsequent analyses have sug-
tumor, is the most widely utilized prognostic factor for gested that 7 cm may be too high and that the T1 classi-
RCC. While there have been numerous staging systems fication should be subclassified into T1a and T1b groups.
developed, the TNM classification system (Table 11-7) However, the exact cutoff between these subgroups is
has become the gold standard for most tumors, including debated, with studies proposing values of 4.0,151 5.5,152
Chapter 11 Renal Cell Carcinoma: Localized Disease 211

and 6.6 cm,150 respectively. A range between 4.0 and Eastern Cooperative Oncology Group (ECOG) PS of 2
4.5 cm seems most reasonable from pathologic, techni- or greater has been associated with a poorer response to
cal, and prognostic considerations. These issues will most immunotherapy171,172 and shorter survival.153,159,173
likely be represented by modifications in The American Biologic factors, such as an increased erythrocyte sedi-
Joint Committee on Cancer 6th Edition of TNM mentation rate,174 elevated platelet counts,175 and hemo-
Staging. globin concentration of less than 10 mg/dl,176 have been
Venous involvement with tumor thrombus is relatively found to be poor prognostic factors.
common in RCC compared to most tumors. The prog- Finally, numerous investigators have attempted to
nostic significance of tumor thrombi has been debated, combine multiple prognostic factors to further improve
most likely due to difficulties in differentiating tumor the overall accuracy. Kattan et al.177 from Memorial
thrombus from blood clot and quantifying vascular inva- Sloane-Kettering Cancer Center developed a nomogram
sion. Some investigators believe that tumor thrombus, no to predict recurrence after surgery for RCC combining
matter the level, is independently associated with a the following predictor variables: patient symptoms,
decreased prognosis.153,154 Others have demonstrated no including incidental, local or systemic, histology, includ-
differences in overall survival with the presence of tumor ing chromophobe, papillary or conventional, tumor size,
thrombi.155,156 The presence of gross, as well as and pathologic stage. Similarly, Zisman et al.178 at the
microvascular, vascular invasion has been unanimously University of California at Los Angeles developed the
associated with an increased incidence of metastasis and UCLA Integrated Staging System (UISS) to stratify
reduced cancer-specific survival.157 patients into survival groups after surgery. Combining
Metastatic disease is associated with a poor prognosis. and stratifying TNM staging, Fuhrman’s grade and
Prognostic factors identified by multivariant analysis ECOG PS resulted in five separate survival groups. The
include PS, number of sites, time to appearance, and projected 2- and 5-year survival rates for the five groups
location. Poor PS has been demonstrated to be inde- were, respectively: I, 96% and 94%; II, 89% and 67%;
pendently associated with a decreased cancer-specific III, 66% and 39%; IV, 42% and 23%; V, 9% and 0%.
survival.158 Multiple metastatic sites have a worse prog- Both of these systems, compared to individual prognostic
nosis compared to single metastatic deposits.159,160 The factors, provide the physician and patient a more accurate
appearance of metastases within 12 months from the means of predicting survival. This will help better deter-
original diagnosis is associated with a poor progno- mine candidates for adjuvant therapy and enrollment in
sis.161,162 Finally, isolated metastatic disease within the experimental trials.
lung tends to have a better prognosis compared to other
organs.102 SCREENING
Complex histologic and molecular analysis of tumor
specimens is becoming a routine part of determining The high proportion of incidentally discovered renal
tumor biology and ultimate prognosis. Histologic tumor tumors combined with improved survival of these lower
type is readily available, with poor prognosis associated stage tumors suggests the possibility of screening for
with collecting duct carcinoma,163 renal medullary carci- RCC. However, it must be remembered that RCC
noma,24 and the presence of sarcomatoid features.164 accounts for only 2% of solid renal tumors, and even in
Grading as described by Fuhrman and colleagues, which selected populations of patients with hematuria, the
is based on nucleolar aspect, size and content, has been prevalence is only 3% to 6%.179,180 The US Preventative
shown to be an independent determinant of prognosis in Services Task Force (1996) and the Canadian Task Force
multiple studies.165,166 Nuclear morphometric studies on the Periodic Health Examination both recommend
have been demonstrated to correlate with prognosis, but against routinely screening asymptomatic patients for
most investigators have been disappointed with the abil- hematuria to identify those with urologic malignan-
ity of these parameters to offer independent prognostic cies.181 Tosaka et al.182 performed screening ultrasounds
information.167–169 Finally, molecular markers are an in a population of 41,364 asymptomatic people and
exciting area of research but none have reached common found only 19 (0.04%) renal adenocarcinomas. They cal-
use in the clinical setting. culated that for every cancer detected, 2177 ultrasounds
Patient factors seem to be very important to the over- had to be performed. Thus, they concluded that the
all prognosis. As would be expected, patients present with immense cost incurred would be prohibitive, and that
“incidentalomas” were found to have tumors of lower screening should be reserved for patients at risk, such as
stage and grade, with a lower incidence of metastases and von Hippel-Lindau disease, hereditary RCC, and
longer survival compared to patients presenting with acquired renal cystic disease. Therefore, population-
symptomatic tumors.170 As stated above, PS, which com- based screening for renal cancer with urinalysis and renal
bines symptoms and comorbidities, has been noted as an imaging can only be recommended for patients at risk for
independent prognostic variable in most series. The a renal malignancy.
212 Part III Kidney and Ureter

SUMMARY 12. Grawitz P: Die Enstehung von Nierentumoren aus


Nebennierengewebe. Arch Klin Chir 30:8241883.
The treatment of RCC has undergone a dramatic change 13. Storkel S, Eble JN, Adlakha K, et al: Classification of
over the past several decades. While the systemic treat- renal cell carcinoma: Workgroup No. 1. Union
ment of advanced disease has made relatively little Internationale Contre le Cancer (UICC) and the
progress, surgical treatment of local RCC is continuously American Joint Committee on Cancer (AJCC). Cancer
evolving. Recent studies have indicated that radical 1997; 80:987–989.
nephrectomy is warranted for certain patients in the face 14. Kovacs G, Akhtar M, Beckwith BJ, et al: The Heidelberg
of metastatic disease. For all but the largest of renal tumors classification of renal cell tumours. J Pathol 1997;
or the presence of renal vein thrombi, ORN has been 183:131–133.
replaced with LRN. Even these cases are starting to be 15. Kovacs G: Molecular differential pathology of renal cell
tumours. Histopathology 1993; 22:1–8.
tackled laparoscopically by some surgeons. Additionally,
16. Mancilla-Jimenez R, Stanley RJ, Blath RA: Papillary
small renal tumors are being treated with nephron-sparing renal cell carcinoma: a clinical, radiologic, and
surgery, despite normal contralateral kidneys. Even open pathologic study of 34 cases. Cancer 1976;
partial nephrectomy is now being challenged by exciting 38:2469–2480.
new laparoscopic and ablative techniques. While the long- 17. Guinan PD, Vogelzang NJ, Fremgen AM, et al: Renal
term results of these techniques are still maturing, the ini- cell carcinoma: tumor size, stage and survival. Members
tial results are very promising. of the Cancer Incidence and End Results Committee.
J Urol 1995; 153:901–903.
18. Thoenes W, Storkel S, Rumpelt HJ: Human
chromophobe cell renal carcinoma. Virchows Arch B
REFERENCES Cell Pathol Incl Mol Pathol 1985; 48:207–217.
19. Fleming S, Lewi HJ: Collecting duct carcinoma of the
1. Robson CJ, Churchill BM, Anderson W: The results of kidney. Histopathology 1986; 10:1131–1141.
radical nephrectomy for renal cell carcinoma. Trans Am 20. Thoenes W, Storkel S, Rumpelt HJ: Histopathology and
Assoc Genitourin Surg 1968; 60:122–129. classification of renal cell tumors (adenomas,
2. Chan DY, Marshall FF: Partial nephrectomy for oncocytomas and carcinomas). The basic cytological and
centrally located tumors. Urology 1999; 54:1088–1091. histopathological elements and their use for diagnostics.
(Discussion 1091–1092.) Pathol Res Pract 1986; 181:125–143.
3. Polascik TJ, Pound CR, Meng MV, et al: Partial 21. Kennedy SM, Merino MJ, Linehan WM, et al:
nephrectomy: technique, complications and pathological Collecting duct carcinoma of the kidney. Hum Pathol
findings. J Urol 1995; 154:1312–1318. 1990; 21:449–456.
4. Clayman RV, Kavoussi LR, Soper NJ, et al: Laparoscopic 22. Davidson AJ, Choyke PL, Hartman DS, et al:
nephrectomy: initial case report. J Urol 1991; Renal medullary carcinoma associated with sickle
146:278–282. cell trait: radiologic findings. Radiology 1995;
5. Winfield HN, Donovan JF, Lund GO, et al: 195:83–85.
Laparoscopic partial nephrectomy: initial experience and 23. Friedrichs P, Lassen P, Canby E, et al: Renal
comparison to the open surgical approach. J Urol 1995; medullary carcinoma and sickle cell trait. J Urol 1997;
153:1409–1414. 157:1349.
6. Bishoff JT, Chen RB, Lee BR, et al: Laparoscopic renal 24. Avery RA, Harris JE, Davis CJ Jr, et al: Renal medullary
cryoablation: acute and long-term clinical, radiographic, carcinoma: clinical and therapeutic aspects of a newly
and pathologic effects in an animal model and described tumor. Cancer 1996; 78:128–132.
application in a clinical trial. J Endourol 1999; 25. Davis CJ Jr, Mostofi FK, Sesterhenn IA: Renal
13:233–239. medullary carcinoma. The seventh sickle cell
7. Gill IS, Novick AC, Meraney AM, et al: Laparoscopic nephropathy. Am J Surg Pathol 1995; 19:1–11.
renal cryoablation in 32 patients. Urology 2000; 26. Mian BM, Bhadkamkar N, Slaton JW, et al: Prognostic
56:748–753. factors and survival of patients with sarcomatoid renal
8. Chow WH, Devesa SS, Warren JL, et al: Rising cell carcinoma. J Urol 2002; 167:65–70.
incidence of renal cell cancer in the United States. JAMA 27. Young AN, de Oliveira Salles PG, Lim SD, et al: Beta
1999; 281:1628–1631. defensin-1, parvalbumin, and vimentin: a panel of
9. Jayson M, Sanders H: Increased incidence of diagnostic immunohistochemical markers for renal
serendipitously discovered renal cell carcinoma. Urology tumors derived from gene expression profiling studies
1998; 51:203–205. using cDNA microarrays. Am J Surg Pathol 2003;
10. Konnak JW, Grossman HB: Renal cell carcinoma as an 27:199–205.
incidental finding. J Urol 1985; 134:1094–1096. 28. Marshall FF, Stewart AK, Menck HR: The National
11. Thompson IM, Peek M: Improvement in survival of Cancer Data Base: report on kidney cancers. The
patients with renal cell carcinoma—the role of the American College of Surgeons Commission on Cancer
serendipitously detected tumor. J Urol 1988; and the American Cancer Society. Cancer 1997;
140:487–490. 80:2167–2174.
Chapter 11 Renal Cell Carcinoma: Localized Disease 213

29. Godley P, Kim SW: Renal cell carcinoma. Curr Opin 46. Krishnamurthi V, Novick AC, Bukowski RM: Efficacy of
Oncol 2002; 14:280–285. multimodality therapy in advanced renal cell carcinoma.
30. Schillinger F: Acquired cystic kidney disease in renal Urology 1998; 51:933–937.
insufficiency: a multicentre study. Group of 47. Fergany AF, Hafez KS, Novick AC: Long-term results of
Nephrologists of the East of France. Eur J Med 1993; nephron sparing surgery for localized renal cell
2:457–460. carcinoma: 10-year follow-up. J Urol 2000; 163:442–445.
31. Hoshida Y, Nakanishi H, Shin M, et al: Renal neoplasias 48. Dunn MD, Portis AJ, Shalhav AL, et al: Laparoscopic
in patients receiving dialysis and renal transplantation: versus open radical nephrectomy: a 9-year experience.
clinico-pathological features and p53 gene mutations. J Urol 2000; 164:1153–1159.
Transplantation 1999; 68:385–390. 49. Parra RO, Perez MG, Boullier JA, et al: Comparison
32. Kuper H, Boffetta P, Adami HO: Tobacco use and between standard flank versus laparoscopic nephrectomy
cancer causation: association by tumour type. J Intern for benign renal disease. J Urol 1995; 153:1171–1173.
Med 2002; 252:206–224. (Discussion 1173–1174.)
33. Chow WH, Gridley G, Fraumeni JF Jr, et al: Obesity, 50. Ratner LE, Kavoussi LR, Sroka M, et al: Laparoscopic
hypertension, and the risk of kidney cancer in men. assisted live donor nephrectomy—a comparison with the
N Engl J Med 2000; 343:1305–1311. open approach. Transplantation 1997; 63:229–233.
34. Mandel JS, McLaughlin JK, Schlehofer B, et al: 51. Marshall FF: Radical nephrectomy: flank approaches. In
International renal-cell cancer study. IV. Occupation. Int Marshall FF (ed): Textbook of Operative Urology,
J Cancer 1995; 61:601–605. pp 248–255. Philadelphia, Pennsylvania, WB Saunders,
35. Lynge E, Andersen A, Nilsson R, et al: Risk of cancer 1996.
and exposure to gasoline vapors. Am J Epidemiol 1997; 52. Kumar S, Duque JL, Guimaraes KC, et al: Short and
145:449–458. long-term morbidity of thoracoabdominal incision for
36. Gago-Dominguez M, Yuan JM, Castelao JE, et al: nephrectomy: a comparison with the flank approach.
Regular use of analgesics is a risk factor for renal cell J Urol 1999; 162:1927–1929.
carcinoma. Br J Cancer 1999; 81:542–548. 53. Gill IS: Retroperitoneal laparoscopic nephrectomy. Urol
37. Lindblad P, Wolk A, Bergstrom R, et al: The role of Clin North Am 1998; 25:343–360.
obesity and weight fluctuations in the etiology of renal 54. McDougall EM, Clayman RV: Laparoscopic
cell cancer: a population-based case-control study. nephrectomy for benign disease: comparison of the
Cancer Epidemiol Biomarkers Prev 1994; 3:631–639. transperitoneal and retroperitoneal approaches.
38. Mattioli S, Truffelli D, Baldasseroni A, et al: J Endourol 1996; 10:45–49.
Occupational risk factors for renal cell cancer: a case— 55. Gill IS, Strzempkowski B, Kaouk J, et al: Prospective
control study in northern Italy. J Occup Environ Med randomized comparison: transperitoneal vs
2002; 44:1028–1036. retroperitoneal laparoscopic radical nephrectomy. J Urol
39. Shapiro JA, Williams MA, Weiss NS: Body mass index 2002; 167:19.
and risk of renal cell carcinoma. Epidemiology 1999; 56. Abbou CC, Cicco A, Gasman D, et al: Retroperitoneal
10:188–191. laparoscopic versus open radical nephrectomy. J Urol
40. Lang EK, Macchia RJ, Thomas R, et al: Improved 1999; 161:1776–1780.
detection of renal pathologic features on multiphasic 57. Nelson CP, Wolf JS Jr: Comparison of hand assisted versus
helical CT compared with IVU in patients presenting standard laparoscopic radical nephrectomy for suspected
with microscopic hematuria. Urology 2003; 61:528–532. renal cell carcinoma. J Urol 2002; 167:1989–1994.
41. Wood BJ, Khan MA, McGovern F, et al: Imaging 58. Ono Y, Kinukawa T, Hattori R, et al: Laparoscopic
guided biopsy of renal masses: indications, accuracy and radical nephrectomy for renal cell carcinoma: a five-year
impact on clinical management. J Urol 1999; experience. Urology 1999; 53:280–286.
161:1470–1474. 59. Gill IS, Schweizer D, Hobart MG, et al: Retroperitoneal
42. Lechevallier E, Andre M, Barriol D, et al: Fine-needle laparoscopic radical nephrectomy: the Cleveland clinic
percutaneous biopsy of renal masses with helical CT experience. J Urol 2000; 163:1665–1670.
guidance. Radiology 2000; 216:506–510. Lotan Y, Gettman MT, Roehrborn CG, et al:
43. Harisinghani MG, Maher MM, Gervais DA, et al: Laparoscopic nephrectomy is cost effective compared to
Incidence of malignancy in complex cystic renal masses open nephrectom in a large country hospital. JSLS 2003;
(Bosniak category III): should imaging-guided biopsy 7:111–15
precede surgery? Am J Roentgenol 2003; 60. Portis AJ, Yan Y, Landman J, et al: Long-term follow-up
180:755–758. after laparoscopic radical nephrectomy. J Urol 2002;
44. Flanigan RC, Salmon SE, Blumenstein BA, et al: 167:1257–1262.
Nephrectomy followed by interferon alfa-2b compared 61. Gill IS, Meraney AM, Schweizer DK, et al: Laparoscopic
with interferon alfa-2b alone for metastatic renal-cell radical nephrectomy in 100 patients: a single center
cancer. N Engl J Med 2001; 345:1655–1659. experience from the United States. Cancer 2001;
45. Mickisch GH, Garin A, van Poppel H, et al: Radical 92:1843–1855.
nephrectomy plus interferon-alfa-based immunotherapy 62. Cadeddu JA, Ono Y, Clayman RV, et al: Laparoscopic
compared with interferon alfa alone in metastatic renal- nephrectomy for renal cell cancer: evaluation of efficacy
cell carcinoma: a randomised trial. Lancet 2001; and safety: a multicenter experience. Urology 1998;
358:966–970. 52:773–777.
214 Part III Kidney and Ureter

63. Chan DY, Cadeddu JA, Jarrett TW, et al: Laparoscopic 78. Hatcher PA, Anderson EE, Paulson DF, et al: Surgical
radical nephrectomy: cancer control for renal cell management and prognosis of renal cell carcinoma
carcinoma. J Urol 2001; 166:2095–2099. (Discussion invading the vena cava. J Urol 1991; 145:20–23.
2099–2100.) (Discussion 23–24.).
64. Kruitwagen RF, Swinkels BM, Keyser KG, et al: 79. Nesbitt JC, Soltero ER, Dinney CP, et al: Surgical
Incidence and effect on survival of abdominal wall management of renal cell carcinoma with inferior vena
metastases at trocar or puncture sites following cava tumor thrombus. Ann Thorac Surg 1997;
laparoscopy or paracentesis in women with ovarian 63:1592–1600.
cancer. Gynecol Oncol 1996; 60:233–237. 80. Skinner DG, Pritchett TR, Lieskovsky G, et al: Vena
65. Lacy AM, Garcia-Valdecasas JC, Delgado S, et al: caval involvement by renal cell carcinoma. Surgical
Postoperative complications of laparoscopic-assisted resection provides meaningful long-term survival. Ann
colectomy. Surg Endosc 1997; 11:119–122. Surg 1989; 210:387–392. (Discussion 392–394.)
66. Schaeff B, Paolucci V, Thomopoulos J: Port site 81. Berg AA: Malignant hypernephroma of the kidney. Its
recurrences after laparoscopic surgery. A review. Dig clinical course and diagnosis, with a description of the
Surg 1998; 15:124–134. author’s method of radical operative cure. Surg Gynecol
67. Vukasin P, Ortega AE, Greene FL, et al: Wound Obstet 1913; 17:463.
recurrence following laparoscopic colon cancer resection. 82. Sosa RE, Muecke EC, Vaughan ED Jr, et al: Renal cell
Results of the American Society of Colon and Rectal carcinoma extending into the inferior vena cava: the
Surgeons Laparoscopic Registry. Dis Colon Rectum prognostic significance of the level of vena caval
1996; 39:S20–S23. involvement. J Urol 1984; 132:1097–1100.
68. Bangma CH, Kirkels WJ, Chadha S, et al: Cutaneous 83. Glazer AA, Novick AC: Long-term follow-up after
metastasis following laparoscopic pelvic surgical treatment for renal cell carcinoma extending
lymphadenectomy for prostatic carcinoma. J Urol 1995; into the right atrium. J Urol 1996; 155:448–450.
153:1635–1636. 84. Montie JE, el Ammar R, Pontes JE, et al: Renal cell
69. Stolla V, Rossi D, Bladou F, et al: Subcutaneous carcinoma with inferior vena cava tumor thrombi. Surg
metastases after coelioscopic lymphadenectomy for Gynecol Obstet 1991; 173:107–115.
vesical urothelial carcinoma. Eur Urol 1994; 26:342–343. 85. Staehler G, Brkovic D: The role of radical surgery for
70. Rassweiler JJ, Paz A, Gaitonde KD, Seemann O, Frede renal cell carcinoma with extension into the vena cava.
T: Laparoscopic surgery for urologic malignancies—the J Urol 2000; 163:1671–1675.
risk of local recurrence and port site metastasis. J Urol 86. Tsui KH, Shvarts O, Barbaric Z, et al: Is adrenalectomy
2002; 167, Abstract 84. a necessary component of radical nephrectomy? UCLA
71. Castilho LN, Fugita OE, Mitre AI, et al: Port site tumor experience with 511 radical nephrectomies. J Urol 2000;
recurrences of renal cell carcinoma after 163:437–441.
videolaparoscopic radical nephrectomy. J Urol 2001; 87. Paul R, Mordhorst J, Busch R, et al: Adrenal sparing
165:519. surgery during radical nephrectomy in patients with
72. Fentie DD, Barrett PH, Taranger LA: Metastatic renal cell cancer: a new algorithm. J Urol 2001;
renal cell cancer after laparoscopic radical 166:59–62.
nephrectomy: long-term follow-up. J Endourol 2000; 88. Blom JH, van Poppel H, Marechal JM, et al: Radical
14:407–411. nephrectomy with and without lymph node dissection:
73. Landman J, Lento P, Hassen W, et al: Feasibility of preliminary results of the EORTC randomized phase III
pathological evaluation of morcellated kidneys after protocol 30881. EORTC Genitourinary Group. Eur
radical nephrectomy. J Urol 2000; 164:2086–2089. Urol 1999; 36:570–575.
74. Meng MV, Koppie TM, Duh QY, et al: Novel method 89. Marshall FF, Powell KC: Lymphadenectomy for renal
of assessing surgical margin status in laparoscopic cell carcinoma: anatomical and therapeutic
specimens. Urology 2001; 58:677–681. considerations. J Urol 1982; 128:677–681.
75. Urban DA, Kerbl K, McDougall EM, et al: Organ 90. Minervini A, Lilas L, Morelli G, et al: Regional lymph
entrapment and renal morcellation: permeability studies. node dissection in the treatment of renal cell
J Urol 1993; 150:1792–1794. carcinoma: is it useful in patients with no suspected
76. Walther MM, Lyne JC, Libutti SK, et al: Laparoscopic adenopathy before or during surgery? BJU Int 2001;
cytoreductive nephrectomy as preparation for 88:169–172.
administration of systemic interleukin-2 in the treatment 91. Giuliani L, Giberti C, Martorana G, et al: Radical
of metastatic renal cell carcinoma: a pilot study. Urology extensive surgery for renal cell carcinoma: long-term
1999; 53:496–501. results and prognostic factors. J Urol 1990; 143:468–473.
77. Gettman MT, Napper C, Corwin TS, et al: (Discussion 473–474.)
Laparoscopic radical nephrectomy: prospective 92. Herrlinger A, Schrott KM, Schott G, et al: What are the
assessment of impact of intact versus fragmented benefits of extended dissection of the regional renal
specimen removal on postoperative quality of life. lymph nodes in the therapy of renal cell carcinoma.
J Endourol 2002; 16:23–26. J Urol 1991; 146:1224–1227.
Chapter 11 Renal Cell Carcinoma: Localized Disease 215

93. Ditonno P, Traficante A, Battaglia M, et al: Role of 110. Cadeddu JA: Re: Cable tie compression to facilitate
lymphadenectomy in renal cell carcinoma. Prog Clin laparoscopic partial nephrectomy. J Urol 2001; 166:219.
Biol Res 1992; 378:169–174. 111. Gettman MT, Bishoff JT, Su LM, et al: Hemostatic
94. Siminovitch JP, Montie JE, Straffon RA: laparoscopic partial nephrectomy: initial experience with
Lymphadenectomy in renal adenocarcinoma. J Urol the radiofrequency coagulation-assisted technique.
1982; 127:1090–1091. Urology 2001; 58:8–11.
95. Karayiannakis AJ, Makri GG, Mantzioka A, et al: Systemic 112. Gill IS, Munch LC, Clayman RV, et al: A new renal
stress response after laparoscopic or open cholecystectomy: tourniquet for open and laparoscopic partial
a randomized trial. Br J Surg 1997; 84:467–471. nephrectomy. J Urol 1995; 154:1113–1116.
96. Rassweiler JJ, Frede T, Lenz E, et al: Long-term 113. Hubert J, Mourey E, Suty JM, et al: Water-jet dissection
experience with laparoscopic retroperitoneal lymph node in renal surgery: experimental study of a new device in
dissection in the management of low-stage testis cancer. the pig. Urol Res 1996; 24:355–359.
Eur Urol 2000; 37:251–260. 114. Gill IS, Desai MM, Kaouk JH, et al: Laparoscopic partial
97. Henriksson C, Haraldsson G, Aldenborg F, et al: Skeletal nephrectomy for renal tumor: duplicating open surgical
metastases in 102 patients evaluated before surgery for renal techniques. J Urol 2002; 167:469–475 (Discussion
cell carcinoma. Scand J Urol Nephrol 1992; 26:363–366. 475–476.)
98. Nishiyama K, Shirahama T, Yoshimura A, et al: 115. Lerner SE, Hawkins CA, Blute ML, et al: Disease
Expression of the multidrug transporter, P-glycoprotein, outcome in patients with low stage renal cell carcinoma
in renal and transitional cell carcinomas. Cancer 1993; treated with nephron sparing or radical surgery. J Urol
71:3611–3619. 1996; 155:1868–1873.
99. de Riese W, Goldenberg K, Allhoff E, et al: Metastatic 116. Sutherland SE, Resnick MI, Maclennan GT, et al: Does
renal cell carcinoma (RCC): spontaneous regression, the size of the surgical margin in partial nephrectomy for
long-term survival and late recurrence. Int Urol Nephrol renal cell cancer really matter? J Urol 2002; 167:61–64.
1991; 23:13–25. 117. Bosniak MA: Observation of small incidentally detected
100. Oliver RT, Nethersell AB, Bottomley JM: Unexplained renal masses. Semin Urol Oncol 1995; 13:267–272.
spontaneous regression and alpha-interferon as 118. Rendon RA, Stanietzky N, Panzarella T, et al: The
treatment for metastatic renal carcinoma. Br J Urol natural history of small renal masses. J Urol 2000;
1989; 63:128–131. 164:1143–1147.
101. Ueno H, Mochizuki H, Hatsuse K, et al: Indicators for 119. Chosy SG, Nakada SY, Lee FT Jr, et al: Monitoring
treatment strategies of colorectal liver metastases. Ann renal cryosurgery: predictors of tissue necrosis in swine.
Surg 2000; 231:59–66. J Urol 1998; 159:1370–1374.
102. van der Poel HG, Roukema JA, Horenblas S, et al: 120. Delworth MG, Pisters LL, Fornage BD, et al:
Metastasectomy in renal cell carcinoma: a multicenter Cryotherapy for renal cell carcinoma and angiomyolipoma.
retrospective analysis. Eur Urol 1999; 35:197–203. J Urol 1996; 155:252–254. (Discussion 254–255.)
103. Franklin JR, Figlin R, Rauch J, et al: Cytoreductive surgery 121. Gill IS, Novick AC, Soble JJ, et al: Laparoscopic renal
in the management of metastatic renal cell carcinoma: the cryoablation: initial clinical series. Urology 1998;
UCLA experience. Semin Urol Oncol 1996; 14:230–236. 52:543–551.
104. Robertson CN, Linehan WM, Pass HI, et al: Preparative 122. Uchida M, Imaide Y, Sugimoto K, et al: Percutaneous
cytoreductive surgery in patients with metastatic renal cryosurgery for renal tumours. Br J Urol 1995;
cell carcinoma treated with adoptive immunotherapy 75:132–136. (Discussion 136–137.)
with interleukin-2 or interleukin-2 plus lymphokine 123. Shingleton WB, Sewell PE Jr: Percutaneous renal tumor
activated killer cells. J Urol 1990; 144:614–617. cryoablation with magnetic resonance imaging guidance.
(Discussion 617–618.) J Urol 2001; 165:773–776.
105. Flanigan RC, Yonover PM: The role of radical 124. Zincke H, Dechet CB, Blute ML: Prospective analysis of
nephrectomy in metastatic renal cell carcinoma. Semin needle biopsy of solid renal masses. J Urol 1999;
Urol Oncol 2001; 19:98–102. 161(45):169.
106. Kim B, Louie AC: Surgical resection following 125. Shingleton WB, Sewell PW: Percutaneous renal
interleukin 2 therapy for metastatic renal cell carcinoma cryoablation—24 and 36 month follow-up. J Endourol
prolongs remission. Arch Surg 1992; 127:1343–1349. 2002; 16:A133.
107. Sella A, Swanson DA, Ro JY, et al: Surgery following 126. Sung GT, Meraney AM, Schweizer DK, et al:
response to interferon-alpha-based therapy for residual Laparoscopic renal cryoablation in 50 patients:
renal cell carcinoma. J Urol 1993; 149:19–21. intermediate follow-up. J Urol 2001; 165(S):158.
(Discussion 21–22.) 127. Shingleton WS, Sewell PE: Percutaneous renal
108. Rackley R, Novick A, Klein E, et al: The impact of cryoablation for renal tumors: one-year follow-up. J Urol
adjuvant nephrectomy on multimodality treatment of 2001; 165(S):186.
metastatic renal cell carcinoma. J Urol 1994; 128. Jacomides L, Ogan K, Watumull L, et al: Laparoscopic
152:1399–1403. application of radio frequency energy enables in situ
109. Novick AC: Nephron-sparing surgery for renal cell renal tumor ablation and partial nephrectomy. J Urol
carcinoma. Annu Rev Med 2002; 53:393–407. 2003; 169:49–53. (Discussion 53.)
216 Part III Kidney and Ureter

129. McGovern FJ, McDougal WS, Gervais D, et al: 146. Vallancien G, Chartier-Kastler E, Harouni M, et al:
Percutaneous radiofrequency ablation of human renal Focused extracorporeal pyrotherapy: experimental study
cell carcinoma. J Urol 2001; 165(S):157. and feasibility in man. Semin Urol 1993; 11:7–9.
130. Pavlovich CP, Walther MM, Choyke PL, et al: 147. Ponsky LE, Crownover RL, Rosen MJ, et al: The initial
Percutaneous radio frequency ablation of small renal evaluation of the cyberknife technology for
tumors: initial results. J Urol 2002; 167:10–15. extracorporeal renal tissue ablation. Urology 2003;
131. Curley SA, Izzo F, Ellis LM, et al: Radiofrequency 61(3):498–501
ablation of hepatocellular cancer in 110 patients with 148. Mejean A, Oudard S, Thiounn N: Prognostic factors of
cirrhosis. Ann Surg 2000; 232:381–391. renal cell carcinoma. J Urol 2003; 169:821–827.
132. Wood TF, Rose DM, Chung M, et al: Radiofrequency 149. Javidan J, Stricker HJ, Tamboli P, et al: Prognostic
ablation of 231 unresectable hepatic tumors: indications, significance of the 1997 TNM classification of renal cell
limitations, and complications. Ann Surg Oncol 2000; carcinoma. J Urol 1999; 162:1277–1281.
7:593–600. 150. Gettman MT, Blute ML, Spotts B, et al: Pathologic
133. Zlotta AR, Wildschutz T, Raviv G, et al: staging of renal cell carcinoma: significance of tumor
Radiofrequency interstitial tumor ablation (RITA) is a classification with the 1997 TNM staging system.
possible new modality for treatment of renal cancer: ex Cancer 2001; 91:354–361.
vivo and in vivo experience. J Endourol 1997; 151. Hafez KS, Fergany AF, Novick AC: Nephron sparing
11:251–258. surgery for localized renal cell carcinoma: impact of
134. Gervais DA, McGovern FJ, Wood BJ, et al: Radio- tumor size on patient survival, tumor recurrence and
frequency ablation of renal cell carcinoma: early clinical TNM staging. J Urol 1999; 162:1930–1933.
experience. Radiology 2000; 217:665–672. 152. Kinouchi T, Saiki S, Meguro N, et al: Impact of tumor
135. Rhim H, Goldberg N, Dodd GD, et al: Essential size on the clinical outcomes of patients with Robson
techniques for successful radiofrequency thermal State I renal cell carcinoma. Cancer 1999; 85:689–695.
ablation of malignant hepatic tumors. Radiographics 153. Giberti C, Oneto F, Martorana G, et al: Radical
2001; 21:S17–S39. nephrectomy for renal cell carcinoma: long-term results
136. Walther MC, Choyke PL, Libutti SK: A phase 2 study of and prognostic factors on a series of 328 cases. Eur Urol
radio frequency interstitial tissue ablation of localized 1997; 31:40–48.
renal tumors. J Urol 2000; 163:1424. 154. Tongaonkar HB, Dandekar NP, Dalal AV, et al: Renal
137. Dupuy DE, Goldberg SN: Image-guided radiofrequency cell carcinoma extending to the renal vein and inferior
tumor ablation: challenges and opportunities—part II. vena cava: results of surgical treatment and prognostic
J Vasc Interv Radiol 2001; 12:1135–1148. factors. J Surg Oncol 1995; 59:94–100.
138. Yohannes P, Pinto P, Rotariu P, et al: 155. Elfving P, Mandahl N, Lundgren R, et al: Prognostic
Retroperitoneoscopic radiofrequency ablation of a solid implications of cytogenetic findings in kidney cancer.
renal mass. J Endourol 2001; 15:845–849. Br J Urol 1997; 80:698–706.
139. Yang R, Reilly CR, Rescorla FJ, et al: High-intensity 156. Medeiros LJ, Gelb AB, Weiss LM: Renal cell carcinoma.
focused ultrasound in the treatment of experimental liver Prognostic significance of morphologic parameters in
cancer. Arch Surg 1991; 126:1002–1009. (Discussion 121 cases. Cancer 1988; 61:1639–1651.
1009–1010.) 157. Griffiths DF, Verghese A, Golash A, et al: Contribution
140. Foster RS, Bihrle R, Sanghvi N, et al: Production of of grade, vascular invasion and age to outcome in
prostatic lesions in canines using transrectally clinically localized renal cell carcinoma. BJU Int 2002;
administered high-intensity focused ultrasound. Eur 90:26–31.
Urol 1993; 23:330–336. 158. Palmer PA, Vinke J, Philip T, et al: Prognostic factors for
141. Bihrle R, Foster RS, Sanghvi NT, et al: High intensity survival in patients with advanced renal cell carcinoma
focused ultrasound for the treatment of benign prostatic treated with recombinant interleukin-2. Ann Oncol
hyperplasia: early United States clinical experience. 1992; 3:475–480.
J Urol 1994; 151(5):1271–1275. 159. Elson PJ, Witte RS, Trump DL: Prognostic factors for
142. Gelet A, Chapelon JY, Bouvier R: Treatment of organ survival in patients with recurrent or metastatic renal cell
confined prostatic cancer by high intensity ultrasound carcinoma. Cancer Res 1988; 48:7310–7313.
(HIFU) delivered by transrectal route: pilot study. J Urol 160. Miyao N, Oda T, Shigyou M, et al: Pre-operatively
1995; 153: 629A. determined prognostic factors in metastatic renal cell
143. Chapelon JY, Margonari J, Theillere Y, et al: Effects of carcinoma. Eur Urol 1997; 31:292–296.
high-energy focused ultrasound on kidney tissue in the 161. Kavolius JP, Mastorakos DP, Pavlovich C, et al:
rat and the dog. Eur Urol 1992; 22:147–152. Resection of metastatic renal cell carcinoma. J Clin
144. Adams JB, Moore RG, Anderson JH, et al: High- Oncol 1998; 16:2261–2266.
intensity focused ultrasound ablation of rabbit kidney 162. Negrier S, Escudier B, Lasset C, et al: Recombinant
tumors. J Endourol 1996; 10:71–75. human interleukin-2, recombinant human interferon
145. Watkin NA, Morris SB, Rivens IH, et al: High-intensity alfa-2a, or both in metastatic renal-cell carcinoma.
focused ultrasound ablation of the kidney in a large Groupe Francais d’Immunotherapie. N Engl J Med
animal model. J Endourol 1997; 11:191–196. 1998; 338:1272–1278.
Chapter 11 Renal Cell Carcinoma: Localized Disease 217

163. Chao D, Zisman A, Pantuck AJ, et al: Collecting duct in renal cell carcinoma. Quantitative assessment of tumor
renal cell carcinoma: clinical study of a rare tumor. J heterogeneity. Cancer 1993; 72:2667–2674.
Urol 2002; 167:71–74. 174. Ljungberg B, Grankvist K, Rasmuson T: Serum acute
164. Grabowski M, Huzarski T, Lubinski J, et al: Survival in phase reactants and prognosis in renal cell carcinoma.
patients with rare subtypes of renal cell carcinoma. BJU Cancer 1995; 76:1435–1439.
Int 2002; 89:599–600. 175. O’Keefe SC, Marshall FF, Issa MM, et al:
165. Bretheau D, Lechevallier E, de Fromont M, et al: Thrombocytosis is associated with a significant increase
Prognostic value of nuclear grade of renal cell in the cancer specific death rate after radical
carcinoma. Cancer 1995; 76:2543–2549. nephrectomy. J Urol 2002; 168:1378–1380.
166. Thrasher JB, Paulson DF: Prognostic factors in renal 176. Yasunaga Y, Shin M, Miki T, et al: Prognostic factors of
cancer. Urol Clin North Am 1993; 20:247–262. renal cell carcinoma: a multivariate analysis. J Surg
167. Carducci MA, Piantadosi S, Pound CR, et al: Nuclear Oncol 1998; 68:11–18.
morphometry adds significant prognostic information to 177. Kattan MW, Reuter V, Motzer RJ, et al: A postoperative
stage and grade for renal cell carcinoma. Urology 1999; prognostic nomogram for renal cell carcinoma. J Urol
53:44–49. 2001; 166:63–67.
168. Pound CR, Partin AW, Epstein JI, et al: Nuclear 178. Zisman A, Pantuck AJ, Dorey F, et al: Improved
morphometry accurately predicts recurrence in clinically prognostication of renal cell carcinoma using an integrated
localized renal cell carcinoma. Urology 1993; 42:243–248. staging system. J Clin Oncol 2001; 19:1649–1657.
169. Ruiz JL, Hernandez M, Martinez J, et al: Value of 179. Carter WC III, Rous SN: Gross hematuria in 110 adult
morphometry as an independent prognostic factor in urologic hospital patients. Urology 1981; 18:342–344.
renal cell carcinoma. Eur Urol 1995; 27:54–57. 180. Warshauer DM, McCarthy SM, Street L, et al:
170. Tsui KH, Shvarts O, Smith RB, et al: Renal cell Detection of renal masses: sensitivities and specificities
carcinoma: prognostic significance of incidentally of excretory urography/linear tomography, US, and CT.
detected tumors. J Urol 2000; 163:426–430. Radiology 1988; 169:363–365.
171. Fossa SD, Kramar A, Droz JP: Prognostic factors and 181. Grossfeld GD, Litwin MS, Wolf JS, et al: Evaluation of
survival in patients with metastatic renal cell carcinoma asymptomatic microscopic hematuria in adults: the
treated with chemotherapy or interferon-alpha. Eur J American Urological Association best practice policy—
Cancer 1994; 30A:1310–1314. part I: definition, detection, prevalence, and etiology.
172. Lissoni P, Barni S, Ardizzoia A, et al: Prognostic factors Urology 2001; 57:599–603.
of the clinical response to subcutaneous immunotherapy 182. Tosaka A, Ohya K, Yamada K, et al: Incidence and
with interleukin-2 alone in patients with metastatic renal properties of renal masses and asymptomatic renal cell
cell carcinoma. Oncology 1994; 51:59–62. carcinoma detected by abdominal ultrasonography.
173. van der Poel HG, Mulders PF, Oosterhof GO, et al: J Urol 1990; 144:1097–1099.
Prognostic value of karyometric and clinical characteristics
C H A P T E R

12 Surgery of Renal Cell Carcinoma,


Including Partial Nephrectomy
Andrew C. Novick, MD

Notwithstanding recent advances in our understanding Involvement of the inferior vena cava (IVC) with
of the genetics and biology of renal cell carcinoma RCC occurs in 3% to 7% of cases and renders the task
(RCC), surgery remains the mainstay of curative treat- of complete surgical excision more complicated. Yet,
ment for this disease. Nevertheless, the role of surgery is operative removal offers the only hope for cure and,
changing with respect to both localized RCC and when there are no metastases, an aggressive approach is
patients with metastatic disease. Nephron-sparing sur- justified. Five-year survival rates of 40% to 58% have
gery has assumed an increasing role in the management been reported after complete surgical excision.2–5 The
of localized tumors. The advent of promising best results have been achieved when the tumor does
immunotherapy regimens and their adjunctive use with not involve the perinephric fat and regional lymph
surgery offers new hope for patients with disseminated nodes. The cephalad extent of vena caval involvement
malignancy. This chapter will review the contemporary is not prognostically important and, even with intraa-
role of surgery and specific operative techniques in the trial tumor thrombi, extended cancer-free survival is
management of patients with RCC. possible following surgical treatment when there is no
nodal or distant metastasis.6 In planning the appropri-
ate operative approach for tumor removal, it is essen-
RADICAL NEPHRECTOMY
tial for preoperative radiographic studies to define
Indications and Evaluation
accurately the distal (craniad) limits of a vena caval
Radical nephrectomy is the treatment of choice for tumor thrombus.
patients with localized RCC.1 The preoperative evalua- RCC involving the IVC should be suspected in
tion of patients with RCC has changed considerably in patients who have lower extremity edema, a varicocele,
recent years due to the advent of new imaging modalities, dilated superficial abdominal veins, proteinuria, pul-
such as ultrasonography, computed tomography (CT) monary embolism, a right atrial mass, or nonfunction of
scanning, and magnetic resonance imaging (MRI). In the involved kidney. Currently, MRI is the preferred
many patients, a complete preliminary evaluation can be diagnostic study for demonstrating both the presence
performed using these noninvasive modalities. Renal and distal extent of IVC involvement.7 Transesophageal
arteriography is no longer routinely necessary prior to echocardiography (TEE) and transabdominal color flow
performing radical nephrectomy. All patients should Doppler ultrasonography have also proven to be useful
undergo a metastatic evaluation including a chest X-ray, diagnostic studies in this regard. Inferior vena cavogra-
abdominal CT scan, and occasionally a bone scan; the phy is reserved for patients in whom an MRI or ultra-
latter is only necessary in patients with bone pain or an sound study is either nondiagnostic or contraindicated.
elevated serum alkaline phosphatase. Radical nephrec- Renal arteriography is particularly helpful in patients
tomy is occasionally done in patients with metastatic dis- with RCC involving the IVC since in 35% to 40% of
ease to palliate severe associated local symptoms, to allow cases, distinct arterialization of a tumor thrombus is
entry into a biologic response modifier protocol, or con- observed. When this finding is present, preoperative
comitant with resection of a solitary metastatic lesion. embolization of the kidney often causes shrinkage of the

218
Chapter 12 Surgery of Renal Cell Carcinoma, Including Partial Nephrectomy 219

Surgical Anatomy
thrombus that facilitates its intraoperative removal.
When adjunctive cardiopulmonary bypass with deep The anatomic relationship of the kidneys to surrounding
hypothermic circulatory arrest is considered, coronary structures is illustrated in Figure 12-1. The kidneys are
angiography is also performed preoperatively.4 If signif- located on either side of the vertebral column in the lum-
icant obstructing coronary lesions are found, these can bar fossa of the retroperitoneal space. Each kidney is sur-
be repaired simultaneously during cardiopulmonary rounded by a layer of perinephric fat that is in turn
bypass. covered by a distinct fascial layer termed Gerota’s fascia.
Radical nephrectomy encompasses the basic princi- Posteriorly, both kidneys lie on the psoas major and
ples of early ligation of the renal artery and vein, quadratus lumborum muscles. Posteriorly and superiorly,
removal of the kidney outside Gerota’s fascia, removal the upper pole of each kidney is in contact with the
of the ipsilateral adrenal gland, and performance of a diaphragm.
complete lymphadenectomy from the crus of the A small segment of the anterior medial surface of the
diaphragm to the aortic bifurcation.1 Perhaps the most right kidney is in contact with the right adrenal gland.
important aspect of radical nephrectomy is removal of However, the major anterior relationships of the right
the kidney outside Gerota’s fascia, because capsular kidney are the liver, which overlies the upper two-
invasion with perinephric fat involvement occurs in thirds of the anterior surface, and the hepatic flexure of
25% of patients. Removal of the ipsilateral adrenal the colon, which overlies the lower one-third. The sec-
gland is not routinely necessary unless the malignancy ond portion of the duodenum covers the right renal
either extensively involves the kidney or is located in the hilum.
upper portion of the kidney.8 Although lymphadenec- A small segment of the anterior medial surface of the
tomy allows for more accurate pathologic staging, the left kidney is also covered by the left adrenal gland. The
therapeutic value remains controversial. A study from major anterior relationships of the left kidney are
Giuliani and associates suggests that a subset of patients the spleen, body of the pancreas, stomach, and splenic
with micrometastatic lymph node involvement may flexure of the colon.
benefit from performance of a lymphadenectomy.9 At
the present time, the need for routine performance of a
Surgical Incisions
complete lymphadenectomy in all cases is unresolved,
and there remains a divergence of clinical practice The surgical approach for radical nephrectomy is deter-
among urologists with respect to this aspect of radical mined by the size and location of the tumor as well as by
nephrectomy. the habitus of the patient.10 The operation is usually per-

Figure 12-1 The anatomic relationship of the kidneys to the surrounding structures. The
liver is retracted superiorly in this illustration.
220 Part III Kidney and Ureter

formed through a transperitoneal incision to allow


abdominal exploration for metastatic disease and early
access to the renal vessels with minimal manipulation of
the tumor. Occasionally, an extraperitoneal flank incision
is employed in elderly patients or in patients with small
tumors who are also classified as poor risks.
The author prefers an extended subcostal or a bilateral
subcostal incision for most patients undergoing radical
nephrectomy (Figure 12-2). This incision provides better
access to the lateral and superior portion of the kidney
than a midline abdominal incision. When employing an
anterior subcostal incision, the patient is in the supine
position with a rolled sheet beneath the upper lumbar
spine. The incision begins approximately one to two fin-
gerbreadths below the costal margin in the anterior axil-
lary line and then extends with a gentle curve across the Figure 12-2 Patient positioning for anterior subcostal
midline, ending at the midportion of the opposite rectus transperitoneal incision.
muscle. The incision is carried through the subcutaneous
tissues to the anterior fascia, which is divided in the
direction of the incision. In the lateral aspect of the inci- from the tumor mass. The patient is placed in a semi-
sion, a portion of the latissimus dorsi muscle is divided. oblique position, with a rolled sheet placed longitudinally
The external oblique muscle is divided, exposing the beneath the flank. The incision is begun in the 8th inter-
fibers of the internal oblique muscle (Figure 12-3A). The costal space, near the angle of the rib, and is carried
rectus, internal oblique, and transversus abdominous across the costal margin to the midpoint of the opposite
muscles are divided along with the posterior rectus rectus muscle, above the umbilicus. The latissimus dorsi,
sheath (see Figure 12-3B and C). The peritoneal cavity is external oblique, rectus, and intercostal muscles are
entered in the midline, and the ligamentum teres is divided in the direction of the incision (Figure 12-5A).
divided (see Figure 12-3D). The pleura is then opened to obtain complete exposure of
The thoracoabdominal approach is preferable for per- the diaphragm (see Figure 12-5B and C).
forming radical nephrectomy in patients with large The diaphragmatic incision is made at the periphery
tumors involving the upper portion of the kidney (Figure about 2 cm inside its attachment at the chest wall with
12-4). It is particularly advantageous on the right side, the incision then being carried around circumferentially
where the liver and its venous drainage into the upper to the posterior aspect of the diaphragm (see Figure 12-5D).
vena cava can limit exposure and impair vascular control Circumferential incision of the diaphragm obviates dam-
as the tumor mass is being removed. Less need exists for age to the phrenic nerve and also creates a diaphragmatic
a thoracoabdominal incision on the left side because the flap, which can be pushed into the chest to provide com-
spleen and pancreas can usually be readily elevated away plete exposure of the liver, which is then retracted

Figure 12-3 A and B, The various steps in performing an anterior subcostal transperitoneal
incision are illustrated (see text for details).
Chapter 12 Surgery of Renal Cell Carcinoma, Including Partial Nephrectomy 221

Figure 12-3 cont’d C and D, The various steps performing an anterior subcostal
transperitoneal incision are illustrated (see text for details).

upward (see Figure 12-5E). If further mobilization of the rior peritoneum lateral to the left colon is incised verti-
liver is needed, the right triangular ligament and coro- cally and the incision is carried upward to divide the
nary ligament can be incised to mobilize the entire right lienorenal ligament. Care must be taken to avoid tearing
lobe of the liver upward. This maneuver provides excel- the delicate capsule of the spleen. The plane between the
lent additional exposure of the suprarenal vena cava. kidney and adrenal gland posteriorly, and the pancreas
Medial to the ribs, the internal oblique and transversus and spleen anteriorly, is developed by blunt dissection.
abdominous muscles are divided and the peritoneal cav- The left colon and duodenum are reflected medially, and
ity is entered. The kidney and great vessels may then be the pancreas and spleen are reflected cephalad, with care
exposed by upward retraction of the liver and medial vis- taken not to injure the spleen or the pancreas (Figure
ceral mobilization (see Figure 12-5F ). 12-6). When adequate exposure of the kidney and great
vessels has been obtained, a self-retaining ring retractor
is inserted to maintain the operative field (Figure 12-7).
Left Radical Nephrectomy
The operation is initiated with dissection of the renal
After the peritoneal cavity is entered, a thorough explo- pedicle. The left renal vein is quite long as it passes over
ration is done to rule out metastatic disease. The poste- the aorta. The vein is mobilized completely by ligating
and dividing gonadal, adrenal, and lumbar tributaries.
The vein can be retracted to expose the artery posteri-
orly, which is then mobilized toward the aorta (Figure
12-8). The renal artery is ligated with 2.0 silk ligatures
and divided, and the renal vein is then similarly managed.
The kidney is mobilized outside Gerota’s fascia with
blunt and sharp dissection as needed (Figure 12-9).
Remaining vascular attachments are secured with nonab-
sorbable sutures or metal clips. Visualization of the upper
vascular attachments is facilitated by downward retrac-
tion of the kidney. The ureter is then ligated and divided
to complete the removal of the kidney and adrenal gland
(Figure 12-10).
The classic description of radical nephrectomy
includes the performance of a complete regional lym-
phadenectomy. The lymph nodes can be removed en
Figure 12-4 Patient positioning for a right thoracoabdominal bloc with the kidney and adrenal gland or separately, fol-
incision. lowing the nephrectomy. The lymph node dissection is
222 Part III Kidney and Ureter

Figure 12-5 The various steps in performing a thoracoabdominal incision are illustrated (see
accompanying text).

begun at the crura of the diaphragm just below the origin all the periaortic lymphatic tissue. Care must be taken to
of the superior mesenteric artery. A readily definable avoid injury to the origins of the celiac and superior
periadventitial plane is seen close to the aorta that can be mesenteric arteries superiorly, as they arise from the
entered. The dissection may then be carried along the anterior surface of the aorta. The dissection of the
aorta and onto the origin of the major vessels, to remove periaortic and pericaval lymph nodes is then carried
Chapter 12 Surgery of Renal Cell Carcinoma, Including Partial Nephrectomy 223

downward en bloc to the origin of the inferior mesenteric obtain exposure and control of the suprarenal vena cava
artery. The sympathetic ganglia and nerves are removed above the level of the tumor thrombus. If necessary, per-
together with the lymphatic tissue. The cisterna chyli is forating veins to the caudate lobe of the liver are secured
identified medial at the right crus. Entering lymphatic and divided to allow separation of the caudate lobe from
vessels are secured to prevent the development of chylous the vena cava. This maneuver can allow an additional 2 to
ascites. 3 cm length of vena cava to be exposed superiorly. The
infrarenal vena cava is then occluded below the thrombus
with a Satinsky venous clamp, and the opposite renal vein
Right Radical Nephrectomy
is gently secured with a small bulldog vascular clamp.
On the right side, after entering the peritoneal cavity, the Finally, in preparation for tumor thrombectomy, a curved
posterior peritoneum lateral to the right colon is incised Satinsky clamp is placed around the suprarenal vena cava
vertically and the incision is carried high up along the above the level of the thrombus (see Figure 12-14D).
vena cava to the level of the hepatic veins. The right The anterior surface of the renal vein is then incised
colon and duodenum are reflected medially, and the liver over the tumor thrombus and the incision is continued
and gallbladder are retracted upward (Figure 12-11). posteriorly with scissors, passing just beneath the throm-
Care is taken to avoid trauma to the delicate hepatic bus (see Figure 12-14E). In most cases, there is no attach-
veins, which may enter the vena cava at this level. When ment of the thrombus to the wall of the vena cava. After
adequate exposure of the kidney and adrenal gland is the renal vein has been circumscribed, gentle downward
obtained, a self-retaining ring retractor is inserted to traction is exerted on the kidney to extract the tumor
maintain the operative field. thrombus from the vena cava (see Figure 12-14F ). After
The vena cava and renal vein are retracted medially removal of the gross specimen, the suprarenal vena caval
and downward to expose the right renal artery. clamp may be released temporarily as the anesthetist
Alternatively, with a large medial tumor, the renal artery applies positive pulmonary pressure; this maneuver can
may be exposed between the vena cava and the aorta ensure that any small remaining fragments of thrombus
(Figure 12-12). Ligation of the renal artery and vein is are flushed free from the vena cava. When the tumor
performed as described on the left side with 2.0 silk liga- thrombectomy is completed, the cavotomy incision is
tures. Since the right renal vein is usually short, ligation repaired with a continuous 5-0 vascular suture (see
should take place at the level of its entrance to the vena Figure 12-14G).
cava. The remainder of the radical nephrectomy is per- In occasional cases, there is direct caval invasion of the
formed as described for left-sided tumors. tumor at the level of the entrance of the renal vein and
for varying distances. This requires resection of a portion
of the vena caval wall. Narrowing of the caval lumen by
Radical Nephrectomy with Infrahepatic Vena Caval
up to 50% will not adversely affect maintenance of caval
Involvement
patency. If further narrowing appears likely, caval recon-
There are four levels of vena caval involvement in RCC struction can be performed with a free graft of peri-
that are characterized according to the distal extent of the cardium.
tumor thrombus (Figure 12-13). A bilateral subcostal In some patients, more extensive direct growth of
transperitoneal incision usually provides excellent expo- tumor into the wall of the vena cava is found at surgery.
sure for performing radical nephrectomy and removal of The prognosis for these patients is generally poor, par-
a perirenal or infrahepatic IVC thrombus. For extremely ticularly when hepatic venous tributaries are also
large tumors involving the upper pole of the kidney, a involved, and the decision to proceed with radical surgi-
thoracoabdominal incision may alternatively be used. cal excision must be carefully considered. Several impor-
After the abdomen is entered, the colon is reflected tant principles must be kept in mind when undertaking
medially and a self-retaining ring retractor is inserted to en bloc vena caval resection. Resection of the infrarenal
maintain exposure of the retroperitoneum (Figure portion of the vena cava usually can be done safely,
12-14A). The renal artery and the ureter are ligated and because an extensive collateral venous supply will be
divided, and the entire kidney is mobilized outside developed in most cases. With right-sided kidney
Gerota’s fascia leaving the kidney attached only by the tumors, resection of the suprarenal vena cava is also pos-
renal vein (see Figure 12-14B,C ). During the initial dis- sible provided the left renal vein is ligated distal to the
section, care is taken to avoid unnecessary manipulation gonadal and adrenal tributaries, which then provide col-
of the renal vein and vena cava. lateral venous drainage from the left kidney with left-
The vena cava is then completely dissected from sur- sided kidney tumors. The suprarenal vena cava cannot be
rounding structures above and below the renal vein, and resected safely owing to the paucity of collateral venous
the opposite renal vein is also mobilized. It is essential to drainage from the right kidney. In such cases, right renal
224 Part III Kidney and Ureter

Figure 12-6 After entering the peritoneal cavity, the colon is Figure 12-8 The left renal vein is mobilized by ligating its
reflected medially to expose the left kidney and great vessels. major branches to expose the artery posteriorly.

Figure 12-7 A self-retaining ring retractor is inserted to Figure 12-9 After securing the pedicle and dividing the
maintain exposure of the operative field. ureter, the kidney is mobilized outside Gerota’s fascia.
Chapter 12 Surgery of Renal Cell Carcinoma, Including Partial Nephrectomy 225

Figure 12-12 The right renal artery may be mobilized either


lateral to the vena cava (below) or between the vena cava
and the aorta (above).
Figure 12-10 Remaining medial vascular attachments are
secured and divided to complete the nephrectomy.

Radical Nephrectomy with Intrahepatic


or Suprahepatic Vena Caval Involvement
In patients with RCC and an intrahepatic or suprahepatic
IVC thrombus, the difficulty of surgical excision is sig-
nificantly increased. In such cases, the operative tech-
nique must be modified because it is not possible to
obtain subdiaphragmatic control of the vena cava above
the tumor thrombus. Several different surgical maneu-
vers have been used to provide adequate exposure, pre-
vent severe bleeding, and achieve complete tumor
removal in this setting.4
One described technique for obtaining vascular con-
trol involves temporary occlusion of the suprahepatic and
intrapericardial portion of the IVC. To reduce hepatic
venous congestion and troublesome backbleeding, the
porta hepatis and superior mesenteric artery are also
temporarily occluded. A disadvantage of this approach is
that occlusion of the latter vessels can be safely tolerated
for only 20 minutes. This approach is also not applicable
Figure 12-11 After entering the peritoneal cavity, the right in cases of tumor extension into the right atrium. At the
colon and duodenum are reflected medially to expose the Cleveland Clinic, we have preferred to employ car-
right kidney and great vessels. diopulmonary bypass with deep hypothermic circulatory
arrest for many patients with supradiaphragmatic tumor
venous drainage can be maintained by preserving a thrombi and for all patients with right atrial tumor
tumor-free strip of vena cava augmented, if necessary, thrombi. We initially reported a favorable experience with
with a pericardial patch; alternatively, the right kidney can this approach in 43 patients,4 and a subsequent study has
be autotransplanted to the pelvis or an interposition graft shown excellent long-term cancer-free survival following
of saphenous vein may be placed from the right renal vein its use in patients with right atrial thrombi.6 The relevant
to the splenic, inferior mesenteric, or portal vein. technical aspects are subsequently described.
226 Part III Kidney and Ureter

Figure 12-13 Classification of inferior vena caval thrombi according to the distal extent of
the thrombus as perineal, infrahepatic, intrahepatic, and suprahepatic.

A bilateral subcostal incision is used for the abdominal and crystalloid cardioplegic solution is infused. Under
portion of the operation. After confirming resectability, a circulatory arrest, deep hypothermia is initiated by reduc-
median sternotomy is made (Figure 12-15). Intraoperative ing arterial inflow blood temperature as low as 10 ˚ C .
monitoring is accomplished with an arterial line, a multiple- The head and abdomen are packed in ice during the cool-
lumen central venous pressure catheter, and a pulmonary ing process. After approximately 15 to 30 minutes, a core
artery catheter. Nasopharyngeal and bladder temperatures temperature of 18 to 20 ˚C is achieved. At this point, flow
are monitored. Anesthesia is induced with either fentanyl, through the perfusion machine is stopped and 95% of the
sufentanil, or thiopental and maintained with a narcotic blood volume is drained into the pump with no flow to
inhalation agent.11 any organ.
The kidney is completely mobilized outside Gerota’s The tumor thrombus can now be removed in an
fascia with division of the renal artery and ureter, such essentially bloodless operative field. An incision is made
that the kidney is left attached only by the renal vein. The in the IVC at the entrance of the involved renal vein, and
infrarenal vena cava and contralateral renal vein are also the ostium is circumscribed. When the tumor extends
exposed. Extensive dissection and mobilization of the into the right atrium, the atrium is opened at the same
suprarenal vena cava are not necessary with this approach. time (Figure 12-17). If possible, the tumor thrombus is
Adequate exposure is somewhat more difficult to achieve removed intact with the kidney. Frequently, this step is
for a left renal tumor. Simultaneous exposure of the vena not possible because of the friability of the thrombus and
cava on the right and the tumor on the left is not readily its adherence to the vena caval wall. In such cases, piece-
accomplished simply by reflecting the left colon medially. meal removal of the thrombus from above and below is
We have dealt with this by transposing the mobilized left necessary. Occasionally, a venous Fogarty catheter can be
kidney anteriorly through a window in the mesentery of inserted into the vena cava to assist in extraction of the
the left colon while leaving the renal vein attached. This thrombus. Under deep hypothermic circulatory arrest,
maneuver yields excellent exposure of the abdominal vena the entire interior lumen of the vena cava can be directly
cava with the attached left renal vein and kidney. Precise inspected to ensure that all fragments of thrombus are
retroperitoneal hemostasis is essential before proceeding completely removed. Hypothermic circulatory arrest can
with cardiopulmonary bypass due to the risk of bleeding be safely maintained for at least 40 minutes without
associated with systemic heparinization. incurring a cerebral ischemic event.12 In difficult cases,
The heart and great vessels are now exposed through this interval can be extended either by maintaining
the median sternotomy. The patient is heparinized, “trickle” blood flow at a rate of 5 to 10 ml/kg/minute13 or
ascending aortic and right atrial venous cannulae are by adjunctive retrograde cerebral perfusion.14
placed, and cardiopulmonary bypass is initiated (Figure Following complete removal of all tumor thrombus,
12-16). When the heart fibrillates, the aorta is clamped the vena cava is closed with a continuous 5-0 vascular
Chapter 12 Surgery of Renal Cell Carcinoma, Including Partial Nephrectomy 227

Figure 12-14 Technique of radical nephrectomy and vena cava thrombectomy with
infrahepatic tumor thrombus (see text for details).
228 Part III Kidney and Ureter

Figure 12-15 Surgical incision for performing radical


nephrectomy with removal of suprahepatic vena caval tumor Figure 12-16 Cannulae are placed in the ascending aorta
thrombus. and right atrium in preparation for cardiopulmonary bypass.

suture and the right atrium is closed. As soon as the vena the porta hepatis may also be occluded (Pringle maneuver).
cava and right atrium have been repaired, rewarming of After removal of the thrombus, repair of the vena cava is
the patient is initiated. If coronary artery bypass grafting performed as previously described. This technique is sim-
is necessary, this procedure is done during the rewarming pler than cardiopulmonary bypass with hypothermic circu-
period. Rewarming takes 20 to 45 minutes and is contin- latory arrest but may entail more operative bleeding.
ued until a core temperature of approximately 37 ˚C is
obtained. Cardiopulmonary bypass is then terminated.
PARTIAL NEPHRECTOMY
Decannulation takes place, and protamine sulfate is
administered to reverse the effects of the heparin. Recent interest in partial nephrectomy or nephron-
Platelets, fresh-frozen plasma, desmopressin acetate, or sparing surgery for RCC has been stimulated by advances
their combination may be provided when coagulopathy is in renal imaging, improved surgical techniques, the
suspected. Aprotinin has also proven effective in revers- increasing number of incidentally discovered low-stage
ing the coagulopathy associated with cardiopulmonary RCCs, and good long-term survival in patients undergo-
bypass but may induce thrombotic complications. ing this form of treatment. Partial nephrectomy entails
Mediastinal chest tubes are placed but the abdomen is complete local resection of a renal tumor while leaving
not routinely drained. the largest possible amount of normal functioning
In patients with supradiaphragmatic vena caval tumor parenchyma in the involved kidney.
thrombi that do not extend into the right atrium, veno- Accepted indications for partial nephrectomy include
venous bypass in the form of a caval-atrial shunt may be situations in which radical nephrectomy would render
employed.15 In this approach, the intrapericardiac vena the patient anephric with subsequent immediate need for
cava, infrarenal vena cava, and opposite renal vein are dialysis. This encompasses patients with bilateral RCC or
temporarily occluded. Cannulas are then inserted into RCC involving a solitary functioning kidney. The latter
the right atrium and infrarenal vena cava. These cannu- circumstance may be present due to unilateral renal age-
las are connected to a primed pump to maintain adequate nesis, prior removal of the contralateral kidney, or irre-
flow from the vena cava to the right heart (Figure 12-18). versible impairment of contralateral renal function
This avoids the obligatory hypotension associated with represented by patients with unilateral RCC, and a func-
temporary occlusion alone of the intrapericardiac and tioning opposite kidney, when the opposite kidney is
infrarenal vena cava. Following the initiation of ven- affected by a condition that might threaten its future
ovenous bypass, the abdominal vena cava is opened and function, such as calculus disease, chronic pyelonephritis,
the thrombus is removed. If bleeding from the hepatic renal artery stenosis, ureteral reflux, or systemic diseases,
veins is troublesome during extraction of the thrombus, such as diabetes and nephrosclerosis.15
Chapter 12 Surgery of Renal Cell Carcinoma, Including Partial Nephrectomy 229

Figure 12-17 The ostium of the renal vein is circumferentially Figure 12-18 Technique of venovenous bypass for removal
incised and the right atrium is opened. Following removal of of supradiaphragmatic vena caval tumor thrombus.
the tumor thrombus, the atriotomy and vena cavotomy
incisions are closed.

Recent studies have clarified the role of partial tomy for RCC is the risk of postoperative local tumor
nephrectomy in patients with localized unilateral RCC recurrence in the operated kidney, which has occurred in
and a normal contralateral kidney. The data indicate that 4% to 6% of patients. These local recurrences are most
radical nephrectomy and partial nephrectomy provide likely a manifestation of undetected microscopic multifo-
equally effective curative treatment for such patients who cal RCC in the remnant kidney. The risk of local tumor
present with a single, small (<4 cm), and clearly localized recurrence after radical nephrectomy has not been studied,
RCC.16 The results of partial nephrectomy are less satis- but it is presumably very low.
factory in patients with larger (>4 cm) or multiple local- We recently reviewed the results of partial nephrec-
ized RCCs, and radical nephrectomy remains the tomy for treatment of localized sporadic RCC in 485
treatment of choice in such cases when the opposite kid- patients managed at the Cleveland Clinic before
ney is normal. The long-term renal functional advantage December 1996.20 A technically successful operation with
of partial nephrectomy with a normal opposite kidney the preservation of function in the treated kidney was
requires further study. achieved in 476 patients (98%). The overall and cancer-
The technical success rate with partial nephrectomy is specific 5-year patient survival rate in the series were 81%
excellent, and long-term patient survival rates free of and 93%, respectively. Recurrent RCC developed postop-
cancer are comparable with those obtained after radical eratively in 44 of 485 patients (9%). Sixteen patients
nephrectomy, particularly for low-stage RCC (Table (3.2%) developed local recurrence in the remnant kidney,
12-1).17–20 The major disadvantage of partial nephrec- whereas 28 patients (5.8%) developed metastatic disease.
230 Part III Kidney and Ureter

More recently, we received the long-term (10-year) ventional arteriography and venography are significant.
results of partial nephrectomy in 107 patients with local- Furthermore, these studies yield limited anatomic spatial
ized sporadic RCC treated before 1988.21 All patients interrelationships between the tumor, normal
were followed up for a minimum of 10 years or until parenchyma, collecting system, and vascular supply.
death. Cancer-specific survival was 88.2% at 5 years and Advances in helical CT and computer technology now
73% at 10 years. Long-term preservation of renal func- allow the production of high quality, three-dimensional
tion was achieved in 100 patients (93%). These results images of the renal vasculature and soft tissue anatomy in
attest that partial nephrectomy is an effective therapy for any plane. New volume-rendering software allows real-
localized RCC that can provide both long-term tumor time interactive stereoscopic viewing of these images and
control and the preservation of renal function. provides a topographic road map of the renal surface and
Evaluation of patients with RCC prior to partial multiplanar views of the intrarenal anatomy. This per-
nephrectomy must include a detailed history and physi- mits the complex renal anatomy to be evaluated by using
cal examination, a laboratory evaluation including serum a single unified study in a format that is familiar to the
creatinine, liver function tests, and urinalysis or urine surgeon and consistent with intraoperative findings,
dipstick check to screen for preoperative proteinuria. thereby obviating mental reconstruction of several two-
Radiographic testing is used to rule out locally extensive dimensional imaging studies. A detailed prospective
or metastatic disease, including chest X-ray and abdomi- study at the Cleveland Clinic showed the utility of
nal CT, as well as possible bone scan and chest or head three-dimensional volume-rendering CT in accurately
CT depending on the clinical circumstances. showing the renal parenchyma and vascular anatomy
Partial nephrectomy is more technically challenging necessary for the performance of partial nephrectomy.22
than en bloc removal of the kidney by radical nephrectomy, The data from three-dimensional CT integrate essential
and it therefore requires a more detailed understanding information from angiography, venography, excretory
of renal anatomy. Knowledge of the relationship of the urography, and conventional two-dimensional CT into a
tumor and its vascular supply to the collecting system and single preoperative staging test that diminishes the need
adjacent normal renal parenchyma is essential for preop- for more invasive imaging. The use of a 3 to 5-minute
erative assessment, which must include a plan for com- videotape in the operating room provides concise, accu-
plete tumor removal and reconstruction of the renal rate, and immediate three-dimensional information to the
remnant. Therefore, more extensive and invasive preop- surgeon during the dissection, allowing him or her to anti-
erative imaging studies are often obtained before partial cipate the subtleties of the anatomy. Three-dimensional,
nephrectomy, compared with radical nephrectomy. In volume-rendered CT has become the imaging modality
some instances, this includes arteriography and occasion- of choice before partial nephrectomy, allowing hilar dis-
ally venography. Arteriography has been used to delin- section, tumor removal and reconstruction to proceed
eate the intrarenal vasculature and may aid in the excision quickly and confidently.
of the tumor while minimizing blood loss and injury to In patients with bilateral synchronous RCC, the kid-
normal adjacent parenchyma. It is most useful for non- ney most amenable to a partial nephrectomy is usually
peripheral tumors encompassing two or more renal arte- approached first by the author. Then, approximately
rial segments. Selective renal venography is performed in 1 month after a technically successful result has been
patients with large or centrally located tumors to evalu- documented, radical nephrectomy or a second partial
ate for intrarenal venous thrombosis and to assess the nephrectomy is performed on the opposite kidney.
adequacy of venous drainage of the planned renal rem- Staging surgery in this fashion obviates the need for tem-
nant. However, these radiographic studies provide only porary dialysis if ischemic renal failure occurs following
two-dimensional views, and the risks and costs of con- nephron-sparing excision of RCC.

Table 12-1 Results of Partial Nephrectomy for Renal Cell Carcinoma


Local Tumor 5-Year Cancer
Series No. Patients Recurrence (%) Specific Survival (%)

Steinbach et al.17 121 4.1 90

Lerner et al.18 185 5.9 89

Belldegrun et al.19 146 2.7 93

Cleveland Clinic20 485 3.2 92


Chapter 12 Surgery of Renal Cell Carcinoma, Including Partial Nephrectomy 231

It is usually possible to perform partial nephrectomy A variety of surgical techniques are available for per-
for malignancy in situ by using an operative approach forming partial nephrectomy in patients with malig-
that optimizes exposure of the kidney and by combining nancy.23 These include simple enucleation, polar
meticulous surgical technique with an understanding of segmental nephrectomy, wedge resection, transverse
the renal vascular anatomy in relation to the tumor. In resection, and extracorporeal partial nephrectomy with
the author’s experience, extracorporeal partial nephrec- renal autotransplantation. All of these techniques require
tomy is rare if ever indicated. We employ an extraperi- adherence to basic principles of early vascular control,
toneal flank incision through the bed of the 11th or 12th avoidance of ischemic renal damage, complete tumor
rib for almost all of these operations; we occasionally use excision with free margins, precise closure of the collect-
a thoracoabdominal incision for very large tumors ing system, careful hemostasis, and closure or coverage of
involving the upper portion of the kidney. These inci- the renal defect with adjacent fat, fascia, peritoneum, or
sions allow the surgeon to operate on the mobilized kid- Oxycel. Whichever technique is employed, the tumor is
ney almost at skin level and provide excellent exposure of removed with a surrounding margin of grossly normal
the peripheral renal vessels. With an anterior subcostal renal parenchyma. Recent studies suggest that the width
transperitoneal incision, the kidney is invariably located of the margin need not be more than 2 to 3 mm.
in the depth of the wound, and the surgical exposure is Intraoperative ultrasound is very helpful in achieving
simply not as good. accurate tumor localization, particularly for intrarenal
When performing in situ partial nephrectomy for lesions that are not visible or palpable from the external
malignancy, the kidney is mobilized within Gerota’s fas- surface of the kidney.24 The argon beam coagulator is a
cia while leaving intact the perirenal fat around the useful adjunct for achieving hemostasis on the transected
tumor. For small peripheral renal tumors, it may not be renal surface. If possible, the renal defect created by the
necessary to control the renal artery. In most cases, how- excision is closed as an additional hemostatic measure. A
ever, partial nephrectomy is most effectively performed retroperitoneal drain is always used as an additional
after temporary renal arterial occlusion. This measure hemostatic measure. A retroperitoneal drain is always left
not only limits intraoperative bleeding but, by reducing in place for at least 7 days. An intraoperative ureteral
renal tissue turgor, also improves access to intrarenal stent is placed only when major reconstruction of the
structures. In most cases, we believe that it is important intrarenal collecting system has been performed.
to leave the renal vein patent throughout the operation. In patients with RCC, partial nephrectomy is con-
This measure decreases intraoperative renal ischemia traindicated in the presence of lymph node metastasis,
and, by allowing venous backbleeding, facilitates hemo- because the prognosis for these patients is poor. Enlarged
stasis by enabling identification of small transected renal or suspicious looking lymph nodes should be biopsied
veins. In patients with centrally located tumors, it is helpful before initiating the renal resection. When partial
to occlude the renal vein temporarily to minimize intraop- nephrectomy is performed, after excision of all gross
erative bleeding from transected major venous branches. tumor, absence of malignancy in the remaining portion
When the renal circulation is temporarily interrupted, of the kidney should be verified intraoperatively by
in situ renal hypothermia is used to protect against postis- frozen-section examinations of biopsy specimens
chemic renal injury. Surface cooling of the kidney with ice obtained at random from the renal margin of excision. It
slush allows up to 3 hours of safe ischemia without per- is unusual for such biopsies to demonstrate residual
manent renal injury. An important caveat with this tumor but, if so, additional renal tissue must be excised.
method is to keep the entire kidney covered with ice slush
for 10 to 15 minutes immediately after occluding the
Segmental Polar Nephrectomy
renal artery and before commencing the partial nephrec-
tomy. This amount of time is needed to obtain core renal In a patient with malignancy confined to the upper or
cooling to a temperature (approximately 20 ˚ C ) that lower pole of the kidney, partial nephrectomy can be per-
optimizes in situ renal preservation. During excision of formed by isolating and ligating the segmental apical or
the tumor, invariably large portions of the kidney are no basilar arterial branch while allowing unimpaired perfu-
longer covered with ice slush and, in the absence of ade- sion to the remainder of the kidney from the main renal
quate prior renal cooling, rapid rewarming and ischemic artery. This procedure is illustrated in Figure 12-19 for a
renal injury can occur. Cooling by perfusion of the kidney tumor confined to the apical vascular segment. The api-
with a cold solution instilled via the renal artery is not rec- cal artery is dissected away from the adjacent structures,
ommended due to the theoretical risk of tumor dissemi- ligated, and divided. Often, a corresponding venous
nation. Mannitol is given intravenously 5 to 10 minutes branch is present, which is similarly ligated and divided.
before temporary renal arterial occlusion. Systemic or An ischemic line of demarcation will then generally
regional anticoagulation to prevent intrarenal vascular appear on the surface of the kidney and will outline the
thrombosis is not necessary. segment to be excised. If this area is not obvious, a few
232 Part III Kidney and Ureter

milliliters of methylene blue can be directly injected dis- the base of the defect. If this is done, there must be no
tally into the ligated apical artery to better outline the tension on the suture line and no significant angulation
limits of the involved renal segment. An incision is then of kinking of blood vessels supplying the kidney.
made in the renal cortex at the line of demarcation, Alternatively, a portion of perirenal fat may simply be
which should be several millimeters away from the visi- inserted into the base of the renal defect as a hemostatic
ble edge of the cancer. The parenchyma is divided by measure and sutured to the parenchymal margins with
sharp and blunt dissection, and the polar segment is interrupted 4-0 chromic. After closure or coverage of the
removed. In cases of malignancy, it is not possible to pre- renal defect, the renal artery is unclamped and circula-
serve a strip of capsule beyond the parenchymal line of tion to the kidney is restored. A Penrose drain is left in
resection for use in closing the renal defect. the perinephric space.
Often a portion of the collecting system will be
removed with the cancer during a segmental polar
Transverse Resection
nephrectomy. The collecting system is carefully closed
with interrupted or continuous 4-0 chromic sutures to A transverse resection is done to remove large tumors that
ensure a watertight repair. Small transected blood vessels extensively involve the upper or lower portion of the kid-
on the renal surface are identified and ligated with shal- ney. This technique is performed using surface hypother-
low figure-of-eight 4-0 chromic sutures. The edges of mia after temporary occlusion of the renal artery (Figure
the kidney are reapproximated as an additional hemosta- 12-21). Major branches of the renal artery and vein sup-
tic measure, using simple interrupted 3-0 chromic plying the tumor-bearing portion of the kidney are identi-
sutures inserted through the capsule and a small amount fied in the renal hilus, ligated, and divided. If possible, this
of parenchyma. Before these sutures are tied, perirenal should be done before temporarily occluding the renal
fat or Oxycel can be inserted into the defect for inclusion artery to minimize the overall period of renal ischemia.
in the renal closure. If the collecting system has been After occluding the renal artery, the parenchyma is
entered, a Penrose drain is left in the perinephric space. divided with blunt and sharp dissection, leaving a margin
of grossly normal tissue around the tumor. Transected
blood vessels on the renal surface are secured as previ-
Wedge Resection
ously described, and the hilus is inspected carefully for
Wedge resection is an appropriate technique for remov- remaining unligated segmental vessels. An internal
ing peripheral tumors on the surface of the kidney, par- ureteral stent may be inserted if extensive reconstruction
ticularly ones that are larger or not confined to either of the collecting system is necessary. If possible, the renal
renal pole. Because these lesions often encompass more defect is sutured together with one of the techniques pre-
than one renal segment, and because this technique is viously described. If this suture cannot be placed without
generally associated with heavier bleeding, it is best to tension or without distorting the renal vessels, a piece of
perform wedge resection with temporary renal arterial peritoneum or perirenal fat is sutured in place to cover
occlusion and surface hypothermia. the defect. Circulation to the kidney is restored, and a
In performing a wedge resection, the tumor is Penrose drain is left in the perirenal space.
removed with a surrounding margin of grossly normal
renal parenchyma (Figure 12-20). The parenchyma is
Simple Enucleation
divided by a combination of sharp and blunt dissection.
Invariably, the tumor extends deeply into the kidney, and Some RCCs are surrounded by a distinct pseudocapsule
the collecting system is entered. Often, prominent of fibrous tissue. The technique of simple enucleation
intrarenal vessels are identified as the parenchyma is implies circumferential incision of the renal parenchyma
being incised. These may be directly suture-ligated at around the tumors simply and rapidly at any location,
that time, while they are most visible. After excision of often with no vascular occlusion and with maximal
the tumor, the collecting system is closed with inter- preservation of normal parenchyma.
rupted or continuous 4-0 chromic sutures. Remaining Initial reports indicated satisfactory short-term clini-
transected blood vessels on the renal surface are secured cal results after enucleation with good patient survival
with figure-of-eight 4-0 chromic sutures. Bleeding at this and low rate of local tumor recurrence. However, most
point is usually minimal, and the operative field can be recent studies have suggested a higher risk of leaving
kept satisfactorily clear by gentle suction during place- residual malignancy in the kidney when enucleation is
ment of hemostatic sutures. performed.25
The renal defect can be closed in one of two ways. These latter reports include several carefully done
The kidney may be closed on itself by approximating the histopathologic studies that have demonstrated frequent
transected cortical margins with simple interrupted 3-0 microscopic tumor penetration of the pseudocapsule that
chromic sutures, after placing a small piece of Oxycel at surrounds the neoplasm. These data indicate that it is not
Chapter 12 Surgery of Renal Cell Carcinoma, Including Partial Nephrectomy 233

Figure 12-19 Technique of segmental (apical) polar nephrectomy with preliminary ligation
of apical arterial and venous branches.
234 Part III Kidney and Ureter

Figure 12-20 Technique of wedge resection for a peripheral tumor on the surface of the
kidney. The renal defect may be closed on itself or covered with perirenal fat.

Partial Nephrectomy for Renal Angiomyolipoma


always possible to be assured of complete tumor encap-
sulation prior to surgery. Local recurrence of tumor in Renal angiomyolipomas (AMLs) are benign hematomas
the treated kidney is a grave complication of partial whose course may be complicated by pain, hematuria,
nephrectomy for RCC, and every attempt should be hemorrhage, rupture, and even death. They may
made to prevent it. Therefore, it is the author’s view that develop spontaneously or be part of the tuberous scle-
a surrounding margin of normal parenchyma should be rosis complex, where they are often multiple and bilat-
removed with the tumor whenever possible. This pro- eral. These tumors have a propensity to grow, and
vides an added margin of safety against the development treatment has been recommended for asymptomatic
of local tumor recurrence and, in most cases, does not AMLs larger than 4 cm and symptomatic AMLs of any
appreciably increase the technical difficulty of the opera- size.27 Partial nephrectomy and selective angioem-
tion. The technique of enucleation is currently employed bolization are two renal-preserving treatment modali-
only in occasional patients with von Hippel-Lindau dis- ties available for patients with these benign neoplasms.
ease and multiple low-stage encapsulated tumors involv- Currently, there are few data reporting the efficacy and
ing both kidneys.26 ability to preserve renal function by using selective

Figure 12-21 Figure of transverse resection for a tumor involving the upper half of the
kidney.
Chapter 12 Surgery of Renal Cell Carcinoma, Including Partial Nephrectomy 235

embolization, and it is therefore best suited when a dis- 9. Giuliani L, Giberti C, Martorama G, Rovida S: Radical
tinct and accessible renal arterial branch supplies the extensive surgery for renal cell carcinoma. J Urol 1990;
tumor exclusively and not the adjacent normal 143:468.
parenchyma. Partial nephrectomy is considered the pre- 10. Novick AC: Surgery of the kidney. In Campbell’s
Urology, edited by Walsh PC, Retik AB, Stamey
ferred treatment in cases of bilateral tumors or tumors
TA, Vaughan ED. W.B. Saunders Co., Philadelphia,
in a solitary kidney. AMLs are well suited to a nephron-
2002.
sparing approach for several reasons. Because these 11. Welch M, Bazaral MG, Schmidt R, et al: Anesthetic
lesions are benign, the risk of residual microfocal dis- management for surgical removal of renal cell carcinoma
ease is of less long-term significance. Furthermore, with caval or atrial tumor thrombus using deep
most AMLs are exophytic and maintain a distinct hypothermic circulatory arrest. J Cardiothoracic
pseudocapsule that is readily identified and can be dis- Anesthesia 1980; 3:580.
sected to a narrow base. The amount of renal 12. Svenson L, Crawford E, Hess K, et al: Deep hypothermia
parenchyma that can be spared with an open procedure with circulatory arrest. J Thoracic Cardiovascu Surg 1993;
is usually much greater than one would predict radi- 106:19.
ographically. There are few published studies evaluating 13. Mault J, Ohtake S, Klingensmith M, et al: Cerebral
metabolism and circulatory arrest: effects of duration
the efficacy of partial nephrectomy for patients with
and strategies for protection. Ann Thoracic Surg 1993;
AMLs. Fazeli-Matin reported the largest series of 27
55:57.
patients undergoing partial nephrectomy for renal 14. Pagano D, Carey JA, Patel RL, et al: Retrograde
AMLs ranging in size up to 26 cm.28 All kidneys main- cerebral perfusion: clinical experience in emergency
tained good function postoperatively, no patient and elective aortic operations. Ann Thorac Surg 1995;
required dialysis, and there were no recurrent AMLs or 59:393.
related symptoms identified at a mean follow-up time of 15. Burt M: Inferior vena caval involvement by renal cell
39 months. When surgical treatment for renal AMLs is carcinoma: use of venovenous bypass as adjunct during
indicated, partial nephrectomy can be performed with a resection. Urol Clin N Am 1991; 18:437.
high-rate of success, even in patients with larger tumors 16. Butler BP, Novick AC, Miller DP, Campbell SA, Licht
involving a solitary kidney. MR: Management of small unilateral renal cell
carcinomas: radical versus nephron-sparing surgery.
Urology 1995; 45:34.
17. Steinbach F, Stockle M, Muller SC, et al: Conservative
surgery of renal cell tumors in 140 patients: 21 years of
REFERENCES experience. J Urol 1992; 148:24–29.
18. Lerner SE, Hawksin CA, Blute ML, et al: Disease
1. Robson CJ, Churchill BM, Anderson W: The results of outcome in patients with low-stage renal cell carcinoma
radical nephrectomy for renal cell carcinoma. J Urol 1969; treated with nephron-sparing or radical surgery. J Urol
101:297. 1996; 155:1868–1873.
2. Schefft P, Novick AC, Straffon RA, Stewart BH: Surgery 19. Belldegrun A, Tsui KH, deKernion JB, et al: Efficacy of
for renal cell carcinoma extending into the vena cava. nephron-sparing surgery for renal cell carcinoma: analysis
J Urol 1978; 120:28. based on the new 1997 tumor-node-metastasis staging
3. Libertino JA, Zinman L, Watkins E: Long-term results of system. J Clin Oncol 1999; 17:2868–2875.
resection of renal cell cancer with extension into inferior 20. Hafez KS, Fergany AF, Novick AC: Nephron-sparing
vena cava. J Urol 1987; 137:21. surgery for localized renal cell carcinoma: impact of
4. Novick AC, Kaye M, Cosgrove D, et al: Experience tumor size on patient survival, tumor recurrence and
with cardiopulmonary bypass and deep hypothermic TNM staging. J Urol 1999; 162:1930–1933.
circulatory arrest in the management of retroperitoneal 21. Fergany AF, Hafez KS, Novick AC: Long-term results of
tumors with large vena caval thrombi. Ann Surg 1990; nephron-sparing surgery for localized renal cell
212:472. carcinoma: 10-year follow-up. J Urol 2000; 163:442–445.
5. Neves RJ, Zincke H: Surgical treatment of renal cancer 22. Coll DM, Uzzo RG, Herts BR, et al: 3-dimensional
with vena cava extension. Br J Urol 1987; 59:390. volume-rendered computerized tomography for
6. Glazer AA, Novick AC: Long-term follow-up after preoperative evaluation and intraoperative treatment of
surgical treatment for renal cell carcinoma extending into patients undergoing nephron-sparing surgery. J Urol
the right atrium. J Urol 1996; 155:448. 1999; 161:1097–1102.
7. Goldfarb DA, Novick AC, Lorgi R, et al: Magnetic 23. Novick AC: Anatomic approaches in nephron-sparing
resonance imaging for assessment of vena caval tumor surgery for renal cell carcinoma. Atlas of Urol Clin N Am
thrombi: a comparative study with vena cavography and 1998; 6:39.
CT scanning. J Urol 1990; 144:1110. 24. Campbell SC, Fichtner J, Novick AC, et al: Intraoperative
8. Sagalowaky AI, Kadesky KT, Ewalt DM, Kennedy TJ: evaluation of renal cell carcinoma: a prospective study of
Factors influencing adrenal metastasis in renal cell the role of ultrasonography and histopathological frozen
carcinoma. J Urol 1994; 151:1181. sections. J Urol 1996; 155:1191–1195.
236 Part III Kidney and Ureter

25. Marshall FF, Taxy JB, Fishman EK, Chang R: The 27. Oesterling JE: The management of renal
feasibility of surgical enucleation for renal cell carcinoma. angiomyolipoma. J Urol 1986; 135:1121–1124.
J Urol 1986; 135:231. 28. Fazeli-Matin S, Novick AC: Nephron-sparing surgery for
26. Spencer WF, Novick AC, Montie JE, Streem SB, Levin renal angiomyolipoma. Urol 1998; 52:577–583.
HS: Surgical treatment of localized renal carcinoma in
von Hippel-Lindau disease. J Urol 1988; 139:507.
C H A P T E R

13 Laparoscopic Radical and Partial


Nephrectomy
Sidney C. Abreu, MD, and Inderbir S. Gill, MD, MCh

At specialized centers worldwide, laparoscopic radical to apply this minimally invasive approach to many
nephrectomy is now becoming established as routine patients with organ-confined kidney cancer who are can-
practice for management of indicated patients with local- didates for radical nephrectomy. Even circumstances ear-
ized renal cell carcinoma.1,2 Current data indicate that, lier considered relative contraindications, such as
compared to open radical nephrectomy, the laparoscopic previous abdominal surgery,7 morbid obesity,8 tumors
approach is associated with comparable operative time, with level I renal vein thrombus,9 and cytoreductive sur-
decreased blood loss, superior recovery, improved gery,10 are now amenable to this laparoscopic approach.
cosmesis, and equivalent cancer control over an interme- In 2004, laparoscopic radical nephrectomy could be
diate and long-term follow-up.3–5 performed for most patients with organ-confined
While laparoscopic radical nephrectomy, practiced for T1-T3aN0M0 renal tumors, who are not candidates for
over a decade now, has become an established procedure, nephron-sparing surgery.5,11 Specific contraindications
laparoscopic partial nephrectomy, a considerably more in 2004 include vena caval involvement, bulky lym-
technically challenging procedure, is a more recent inno- phadenopathy, and locally invasive tumors.5,11 Large
vation. However, ongoing advances in laparoscopic tech- tumor size was earlier considered a relative contraindica-
niques and operator skills have allowed the development tion. However, we believe this to be a decision based on
of a reliable technique of laparoscopic partial nephrec- the experience of the individual surgeon, and consider
tomy.6 Thus, laparoscopic partial nephrectomy is emerg- tumors as large as 15 cm to be amenable to laparoscopic
ing as an attractive minimally invasive nephron-sparing excision.12 General contraindications for major abdomi-
option at select institutions. nal laparoscopic surgery apply.
In this chapter, we present an update on laparoscopic
radical and partial nephrectomy for the management of
Approach Selection
renal tumors. For each procedure, we describe the current
indications and contraindications, laparoscopic operative From a technical standpoint, laparoscopic radical
technique, intraoperative and perioperative data, compli- nephrectomy can be performed safely and efficaciously
cations, oncologic outcome, and additional specific facets. by either the transperitoneal or the retroperitoneal
In the future, it appears likely that minimally invasive approach. The transperitoneal approach has been
techniques will become an established treatment option employed more commonly, primarily because of its
for treatment of a majority of kidney tumors, with open “familiar” anatomic landmarks and the larger working
surgery being reserved for specific important indications. space offered by this route. Nonetheless, since the kidney
is a retroperitoneal organ, a direct “retroperitoneo-
scopic” approach has considerable logical appeal, thus
LAPAROSCOPIC RADICAL NEPHRECTOMY
avoiding the peritoneal cavity completely. Critics of the
Current Indications and Contra-indications
retroperitoneal approach point to its somewhat smaller
In the decade since the first laparoscopic radical nephrec- initial working space, and a steeper learning curve. At the
tomy, standardization of technique has allowed surgeons Cleveland Clinic we are equally technically comfortable

237
238 Part III Kidney and Ureter

with either approach. It is obvious to us that balloon dila- reported1,18,19 Herein, we briefly describe both
tion affords an adequate working space, and a sound approaches.
knowledge of retroperitoneal surgical anatomy allows
rapid identification of anatomic landmarks.13
Transperitoneal Approach
Retroperitoneoscopy has the remarkable advantage of
direct access to, and rapid control of, the renal artery and Typically, a 3- or 4-port approach is employed (Figure
vein prior to any tumor manipulation.14,15 A prospective 13-1). The line of Toldt is incised to mobilize the colon,
randomized study comparing transperitoneal and and this incision is transversally carried medially along
retroperitoneal approaches to laparoscopic radical the undersurface of the liver or spleen. On the right side,
nephrectomy at the Cleveland Clinic has shown both sharp dissection is used to mobilize the duodenum medi-
approaches to be comparable as regards blood loss, hos- ally until the anterior aspect of the vena cava is exposed.
pital stay, analgesia requirements, and convalescence. The gonadal vein and ureter are identified and retracted
However, the retroperitoneal approach had the inherent laterally. Blunt dissection is performed between the infe-
advantages of a quicker renal hilar control and a shorter rior vena cava and the laterally retract gonadal vein/
operative time.16 We believe that equal facility with both ureter, to identify the psoas muscle. The ureter and
approaches will allow the experienced laparoscopic sur- gonadal vein are secured, divided, and tautly retracted
geon to select the appropriate technique for the particu- laterally. Progressive cephalad dissection is performed
lar case based on the individual patient’s characteristics. along the lateral border of the inferior vena cava with the
Two clinical situations where we prefer the retroperi- psoas muscle in clear view at all times until the renal vein,
toneal approach include morbid obesity and multiple which is mobilized circumferentially. Anterolateral rota-
prior abdominal surgeries.8,17 tion of the completely free lower pole brings the posteri-
orly located renal artery into view. A single hemlock clip
(Weck Closure Systems, Triangle Park, NC) is placed on
Surgical Technique
the renal artery in-continuity at this time to arrest renal
The techniques of transperitoneal and retroperitoneal arterial inflow. The renal vein is controlled and transected
laparoscopic radical nephrectomy have been previously with an Endo-GIA stapler (US Surgical, Norwalk, CT)

Figure 13-1 Port placement for transperitoneal left side radical nephrectomy.
Chapter 13 Laparoscopic Radical and Partial Nephrectomy 239

Retroperitoneal Approach
(Figure 13-2). The clip-occluded renal artery, now in
clear view, is circumferentially mobilized, three clips are Initially a horizontal 1.5-cm skin incision is made just
applied proximally towards the aorta and two towards below the tip of the 12th rib to gain entry into the
the kidneys, and the renal artery is transected. During retroperitoneal space. A trocar-mounted balloon dissec-
upper pole dissection, the adrenal gland is taken en tion device (Origin Medsystems, Menlo Park, CA) is
bloc, if necessary. The adrenal vein is dissected and con- inserted immediately anterior to the psoas muscle and
trolled with clips. The en bloc specimen is then com- posterior to the Gerota’s fascia. Typically 800 to 1000 ml
pletely freed and placed in an Endocatch bag for intact of air are instilled into the balloon for rapid and atrau-
extraction. matic creation of the working space in the retroperi-
For a left-side procedure, it is again important (as on toneum. The balloon dilatation outside and posterior to
the right side) to enter the correct avascular fascial plane Gerota’s fascia effectively displaces the Gerota’s fascia
between the anterior surface of Gerota’s fascia and the covered kidney anteromedially, allowing direct access to
posterior aspect of the descending mesocolon. It is also the posterior aspect of the renal hilum (Figure 13-3).
important to obtain complete mobilization of the spleen Laparoscopic examination from within the transparent
along its lateral border, which facilitates its medial dis- balloon confirms adequate expansion of the retroperi-
placement by gravity alone throughout the procedure. toneum. Secondary cephalad or caudad balloon dilata-
Occasionally, a distended descending colon and the tion, as required by the clinical situation, further enlarges
spleen must be retracted medially with a fan retractor in the retroperitoneal working space (Figure 13-4).
order to expose the renal hilum. For this purpose, an Following balloon dilatation and removal, a 10-mm
additional port can be placed in the lower abdomen Bluntip trocar (Origin Medsystems, Menlo Park, CA) is
through a Pfannenstiel incision, the site of subsequent placed as the primary port to prevent air leakage. Two
specimen extraction. Psoas muscle identification and secondary ports are placed under 30-degree laparoscopic
renal hilar control are performed as described for the visualization. A 10/12-mm port is placed three fingers
right-side procedure. breadths cephalad to the iliac crest, between the mid and

Figure 13-2 Renal artery and vein are sequentially controlled with clip-applier and Endo-GIA
vascular stapler.
240 Part III Kidney and Ureter

anterior axillary lines. For this purpose, the lateral peri-


toneal reflection must be clearly visualized laparoscopi-
cally and avoided. A second 10/12-mm port is placed at
the lateral border of the erector spinae muscle just
below the 12th rib (Figure 13-5). The kidney is
retracted anterolaterally, placing the renal hilum on
traction. Dissection is performed parallel to, and 1 to 2
cm anterior to, the psoas muscle, exposing the vertically
pulsating renal artery. Visualization of the vertically ori-
ented, distinct arterial pulsations indicates the location
of the renal artery, which is circumferentially mobilized,
clip-ligated, and divided. Next, the renal vein is con-
trolled with an Endo-GIA stapler (US Surgical,
Norwalk, CT). The specimen is then circumferentially
mobilized en bloc with or without the adrenal gland.
During mobilization of the specimen from the under-
side of the peritoneal envelope, use of electrocautery
must be minimized in order to avoid the possibility of
transmural thermal damage to the bowel. The entire
dissection is performed outside Gerota’s fascia, mirror-
ing established oncologic principles of open surgery.
Organ entrapment is rapidly performed by using an
Endocatch II bag (US Surgical, Norwalk, CT). The
intact specimen is extracted extraperitoneally through a
Figure 13-3 Balloon dissection of the retroperitoneum. Upon low, muscle-splitting Pfannenstiel incision.
inflating the balloon, the kidney is displaced anteromedially,
allowing direct access to the renal hilum.

Figure 13-4 Secondary cephalad balloon dilation is performed to further enlarge the initial
retroperitoneal working space.
Chapter 13 Laparoscopic Radical and Partial Nephrectomy 241

Figure 13-5 Port placement during retroperitoneoscopy. A, Primary 10 mm port is placed at


the tip of 12th rib. B, 10/12 mm port is placed at junction of lateral border of the erector
spinae muscle with underside of 12th rib. C, 10/12 mm port is placed three fingers breadths
cephalad to iliac crest, between mid and anterior axillary lines.

Results
cera are out of sight during retroperitoneoscopy, they
Worldwide intraoperative and perioperative results on must never be out-of-mind, since they are separated
laparoscopic radical nephrectomy are summarized in only by the peritoneal layer, and are therefore suscepti-
Table 13-1. ble to direct or transmural injury.

Complications Specimen Extraction (Morcellate or Leave Intact)


In the hands of urologists with reasonable experience in The author’s routine practice for extraction of the cancer-
minimally invasive techniques, laparoscopic radical ous specimen aims to provide an intact specimen for accu-
nephrectomy is a safe procedure with a low incidence of rate pathologic staging while achieving a superior
complications similar to open surgery. Both the transperi- cosmetic result. Valuable histopathologic information
toneal and retroperitoneal laparoscopic approaches are regarding the tumor stage, and completeness of tumor
associated with a low incidence of open conversion vary- resection with negative surgical margins, cannot be
ing from 0% to 8%.20–22 Worldwide intraoperative com- reliably obtained following specimen morcellation.
plications following laparoscopic radical nephrectomy are Although some argue that CT scanning can be used effec-
summarized in Tables 13-2 and 13-3. tively for clinical tumor staging, it is well known that CT
During laparoscopic radical nephrectomy, one must scan can overestimate tumor size and, more worrisomely,
always be mindful of aberrant major vessels arising from underestimate tumor stage when compared to final
the aorta.2,23 The superior mesenteric artery arises from pathology in the individual patient.5,11 Moreover, data
the aorta more medially and superiorly than the left from many institutions, including ours, confirm that over-
renal artery, and it is not accompanied by the respective all patient morbidity (analgesic requirement, hospital stay,
vein. If there is any concern whether or not the isolated convalescence) are similar between the intact extraction
vessel is indeed the renal artery, it should be dissected and morcellation techniques.25 Currently, we routinely
and traced distally up to its entrance in the renal hilum. perform intact specimen extraction through a low muscle-
Injuries to intraperitoneal organs (i.e., liver, spleen, splitting Pfannenstiel incision for male patients and
colon, duodenum) are rare. It is evident that lack of vio- through the vagina in the appropriate female patient. This
lation of the peritoneal cavity during retroperitoneal approach minimizes any practical cosmetic disadvantage
laparoscopy may minimize the chances of intraperi- of intact extraction, while maintaining oncologic sanctity
toneal organ injury. However, although peritoneal vis- of the intact pathologic specimen.11,25
242

Table 13-1 Intraoperative and Perioperative Data on Laparoscopic Radical Nephrectomy: Worldwide Experience
Author/Series Dunn et al.2 Gill et al.11 Ono et al.3 Chan et al.20 Barrett et al.21 Janetschek et al.22

Number of patients 60 100 103 67 72 73


Part III Kidney and Ureter

Age (years) 63.5 61.8 57.2 61 35–92 63.3

Male (%) 53 55 77 60 — 44

Right side (%) 47 54 53 45 50 53

Tumor size (cm) 5.3 5.1 (1.7–14) 3.1 (1.1–4.8) 5.1 (1–13) 4.5 (1–9)* 3.8 (3–6)

Transperitoneal/retroperitoneal 55/3† 27/73 85/18 66/1 72/0 73/0

OR time (hour) 5.5 2.8 4.7 4.4 2.5 2.3

EBL (cc) 172 212 254 289 — 170

Morcellate/intact 39/21 0/100 78/25 40/27 66/0 0/73

Specimen weight (g) 452 554 305 — 402

LOS (days) 3.4 1.6 — 3.8 4.4 7.2

Complications (%)

Intraoperative/postoperative — — 10/3 3/12 — —

Minor/major 34/3 11/3 — — 11/8 8/4

Open conversion 1 2 4 1 6 0

*Excluding 6 cases of open conversion.


†Intentionally combined approached was used in 3 patients.
Table 13-2 Complications Following Laparoscopic Radical Nephrectomy
Author/series Siqueira et al.23 Dunn et al.2 Barret et al.21 Ono et al.3 Gill et al.11 Chan et al.20

Number of patients 61 60 72 103 100 67

Approach Transperitoneal Transperitoneal Transperitoneal Transp(85)/ Transp(27)/ Transperitoneal


Retrop (18) Retrop(73)

Complications (%) 4 3 2 9 2 2

Vascular Superior mesenteric Superior mesenteric Vena cava injury (2)/


artery division/ artery ligation/ open surgical
open graft openreanastomosis suture repair
revascularization, to the aorta
death

Hemorrhage Aberrant right lower Bleeding from renal Bleeding in the Bleeding from renal Bleeding from renal
pole artery/open vein; Bleeding from hilar region artery (1), renal artery; Generalized
conversion the gonadal vein/ precluding vein(1)/open oozing in a patient
laparoscopic adequate conversion; Bleeding with ESRD/both
hemostasis without visualization/ from adrenal gland (1), cases* required open
blood transfusion open conversion periureteric artery (1)*/ conversion, an open
laparoscopic splenectomy was
control and blood also performed in
transfusion second case

Visceral Liver laceration Injury to spleen/ Injury to spleen/ Injury to spleen/


during port conservative conservative conservative
placement/oxidize treatment treatment treatment
cellulose packing

Bowel Partial ischemia Duodenal perfuration/ Bowel injury during


colitis/conservative reintervention morcellation/open
treatment for open conversion for
duodenojejunostomy; bowel resection and
Descending colon extensive irrigation
injury/open surgical
repair

*Retroperitoneal approach.
Chapter 13 Laparoscopic Radical and Partial Nephrectomy 243
244 Part III Kidney and Ureter

Table 13-3 Perioperative Complications Following A sponge-stick is externally inserted into the sterilely
Laparoscopic Radical Nephrectomy—Multicenter prepared vagina and tautly positioned in the posterior
Experience with 157 Cases fornix. Laparoscopically, a transverse posterior 3-cm
Ileus 4 (2.5%) colpotomy is created at the apex of the tented-up poste-
rior fornix, and the drawstring of the entrapped specimen
Urinary tract infection 2 (1.2%) is delivered into the vagina (Figure 13-6). After laparo-
scopic exit is completed, the patient is placed in a supine
Pulmonary embolus* 2
lithotomy position. The specimen is extracted intact per
Congestive heart failure† 1 (0.6%) vagina, and the posterior colpotomy incision repaired
transvaginally. This approach is contraindicated in
Transfusion 1 patients with pelvic adhesions from any etiology.27
Incisional hernia 1
Oncologic Outcome
Wound infection 1
To achieve optimal oncologic outcomes, established
Seroma 1 principles of renal cancer surgery must be respected. The
excised surgical specimen must be removed in a hermetic
Postoperative surgical exploration‡ 1
sac to avoid contact with the abdominal wall.28 Either
Duodenal perforation§ 1 intact specimen extraction or morcellation are safe
regarding oncologic outcomes. However, if morcellation
Splenic injury¶ 1 is employed, care should be taken to avoid rare compli-
cations, such as sack perforation, with potential for bowel
Total 16 (10%)
injury and tumor spillage. In the aggregate, laparoscopic
Adapted with permission from Cadeddu JA, Ono Y, Clayman, RV:
radical nephrectomy does not result in an increased risk
Urology 1998; 52:773. of port site seeding, local recurrence, or metastasis3,5,11,28
*Intraoperative mortality: postmortem examination indeterminate (Table 13-4).
(possible carbon dioxide embolus). Cancer-free patient survival is the ultimate criterion of
†One patient died of heart failure one month postoperatively.
‡Postoperative hemorrhage requiring exploratory laparotomy.
oncologic outcome. Portis et al.5 reported 5-year cancer
§Perforation discovery on postoperative day 1. specific survival for the laparoscopic radical nephrectomy
¶Minor splenic injury identified intraoperatively and managed similar to open surgery: 98% versus 92%. The authors
conservatively. also reported 5-year cancer specific survival of 97% fol-
lowing laparoscopic radical nephrectomy for T1Nx dis-
Specimen Extraction in Males ease. These data compare favorably with existing reports
During a transperitoneal approach, a Pfannenstiel skin of survival after conventional open total/radical nephrec-
incision is made at, or just below, the level of the pubic hair- tomy for T1N0: 91% to 95%. Cicco et al.29 have
line. Straightforward access is gained into the peritoneal reported a Kaplan–Meyer actuarial disease-free survival
cavity after the anterior rectus fascia is incised transversally rate of 91% at 54 months following retroperitoneal
and the rectus muscle is separated at the midline. During a laparoscopic radical nephrectomy.
retroperitoneal approach, a similar low Pfannenstiel skin Wound seeding after open radical nephrectomy,
incision is performed although slightly lateralized towards although extremely rare, has been reported in 2 of 518
the nephrectomy side.26 In order to avoid violation of the patients (0.4%). Similarly, a few port site recurrences have
peritoneal cavity, the anterior fascia is incised obliquely at been reported following laparoscopic radical nephrec-
the lateral border of the rectus abdominis. Subsequently, tomy.28 Fentie et al.30 observed one port site seeding 25
the rectus muscle fibers are retracted medially, the poste- months after radical nephrectomy for a grade 4 renal cell
rior rectus fascia is incised, and the peritoneal membrane is carcinoma. Castilho et al.31 reported multiple port site
reflected cephalad using finger dissection. Access is gained abdominal masses 5 months after the laparoscopic proce-
to the retroperitoneal space, and entrapped specimen is dure in a patient with ascites. Although the etiology of port
extracted. Alternatively, a muscle-splitting Gibson incision site recurrence remains unclear in these cases, in both pro-
can be performed for intact specimen extraction during cedures the specimens were extracted with morcellation.
either the transperitoneal or retroperitoneal approach. Local recurrence occurs occasionally following open
radical nephrectomy and appeared to be related to the
initial pathologic stage.30 As regards local recurrence fol-
Transvaginal Extraction in Females
lowing laparoscopic radical nephrectomy, Fentie et al.30
With the table set in a steep Trendelenburg position, have reported an incidence of 5% (3/57), including the
bowel loops are retracted cephalad out of the pelvis. case with port site recurrence. Ono et al.3 have reported
Chapter 13 Laparoscopic Radical and Partial Nephrectomy 245

Figure 13-6 The drawstring of the closed Endocach II bag, previously grasped by the
laparoscopic clamp, is delivered into the vagina.

1 case of local recurrence 43 months after the laparo- tution, 33 patients had larger, clinical stage T2 tumors,
scopic procedure. Although no concomitant port site 7 cm or greater in size. Mean tumor size was 9.8 cm, with
seeding was observed in this case, the intact specimen 17 tumors between 7 and 10 cm in size, and 16 tumors
could not be entrapped and was therefore removed with- >10 cm in size.12 This group was compared to a contem-
out a bag through an additional incision. porary group of 34 patients undergoing open radical
Distant metastases have been reported following nephrectomy for tumors >7 cm in size. Tumors >14 cm
laparoscopic radical nephrectomy. Cicco et al.29 reported or with IVC thrombus were excluded from this
1 patient with metastatic disease 9 months after the study. Estimated blood loss (294 cc—lap; 837 cc—open;
laparoscopic procedure. Hepatic tumor metastasis fol- p-value: <0.001) and hospital stay (1.8 days—lap; 6.1
lowed a local recurrence in a patient who had a pT2G3 days—open; p-value: <0.001) were reduced in the laparo-
renal cell carcinoma and tumor-free surgical margins. scopic group. Although there was a trend to less surgical
Gill et al.11 reported 2 patients with dialysis-dependent, complications in the laparoscopic group, this finding was
end-stage renal disease and pT1N0M0 renal cancer with not statistically significant (6.2% versus 23.5%; p-value:
negative surgical margins that had metastatic disease at 8 0.08) (Table 13-5).
and 12 months, respectively.11
Concomitant Adrenalectomy
Larger Renal Tumors
Performance of concomitant adrenalectomy depends
With increasing experience, laparoscopic radical on the site and size of the renal tumor and the pres-
nephrectomy for larger (≥7 cm) can be performed effica- ence of any abnormality in the adrenal gland on preop-
ciously.12 Of the senior author’s overall experience with erative CT. Generally, ipsilateral adrenalectomy is
over 300 laparoscopic radical nephrectomies at our insti- not necessary if the preoperative CT scan clearly
246 Part III Kidney and Ureter

Table 13-4 Laparoscopic Radical Nephrectomy: Intermediate and Long-term Follow-up


(Pathologically Confirmed RCC)
Author/series Cicco et al.29 Gill et al.11 Portis et al.5 Ono et al.3 Chan et al.20

Number of patients 41 80 64 103 67

Transperitoneal/retroperitoneal All retroperitoneal All retroperitoneal 52/12 85/18 All transperitoneal

Morcellate/intact 0/41 0/80 25/39 78/25 40/27

Pathologic tumor size (cm) 5.26 (2–9) 4.6 (1.7–14) 4.3 (2–10) 3.1 (1.1–4.8) 4.8 morcellate;
5.5 intact

Tumor grade 2.3±1.0 2±0.8 1.88 1.8±0.4 2.1±0.6

Pathologic tumor classification

pT1 25 61 102 46

pT2 2 6 — 8

pT3a 9 9 1 8

pT3b 3 3 — 5

pT4 — 1 — —

Port site recurrence 0 0 0 0 0

Local recurrence 1 0 1 1 0

Time after operation (months) 9.1 — 12 43 —

Metastasis 1 2 3 3 2

Cancer specific mortality 2* 1 1 0 2

Five-year disease-free (%) — — 98 89.7 95

Follow-up (months) 24.76 (2.4–52.7) 16.1 (1–36) 54 (0–94) 29 (3–95) 35.6 (12–111)

*One patient was M+ at the time of diagnosis.

demonstrates the adrenal gland to be normal in size, involvement was identified on preoperative CT scan.33 In
shape, contour, and location, thereby showing it to be the group of patients with gross involvement of the renal
uninvolved by the renal tumor.32 In our initial series, en vein, mean tumor size was 12.4 cm (6 to 20 cm). CT scan
bloc adrenalectomy was performed in 72% of the detected renal vein thrombus floating within the vein in
patients. Adrenal-sparing was performed in the setting 7 patients and invading the renal vein wall in 1 patient.
of bilateral radical nephrectomy, previous contralateral All procedures were successfully completed laparoscopi-
adrenalectomy, or elective preservation of the adrenal cally, except for one open conversion due to intraopera-
gland.11 tive hemorrhage (1000 ml) (Table 13-6). Either the
transperitoneal or retroperitoneal techniques were
Renal Vein Involvement employed to perform right or left laparoscopic radical
nephrectomy. After the renal artery was clipped and tran-
We recently reported a series of 16 patients who under- sected, the renal vein was dissected proximally towards
went laparoscopic radical nephrectomy for pathologically the inferior vena cava. While the renal vein segment con-
confirmed renal cell cancer with either microscopic taining the thrombus clearly appeared full and bulky, the
(8 patients) or gross (8 patients) tumor involvement of uninvolved, free-of-thrombus proximal renal vein seg-
the renal vein. Microscopic involvement was incidentally ment was flat on laparoscopic examination. This estab-
identified on postoperative histopathology. Gross lished a clear line of demarcation, where the articulating
Chapter 13 Laparoscopic Radical and Partial Nephrectomy 247

Table 13-5 The Large (>7 cm, pt2) Renal Tumor—Laparoscopic Versus Open Radical Nephrectomy
Laparoscopic Radical Open Radical
Nephrectomy Nephrectomy
(>7 cm) N=33 (>7 cm) N=34 p-value

Mean tumor size (cm) 9.8 10 0.52

Clinical status 0.66

Localized (%) 91.4 88.2

Metastatic (%) 8.6 11.8

Mean OR time (min) 199 200 0.35

Mean EBL (ml) 294 837 <0.001

Intraoperative complications (%) 6 18 0.12

Conversion to open surgery (%) 0 N/A N/A

Mean LOS (days) 1.8 6.1 <0.001

MSO4 equivalent 22 400 <0.001

Postoperative complications (%) 20 26 0.52

Convalescence (weeks) 4 8 0.018

Specimen weight (g) 890 719 0.17

RCC (%) 82 88 0.15

pT1 13 0 0.08

pT2 29 50

pT3 54 50

Positive surgical margins 1 0 0.32

Adapted with permission from Steinberg AP, Desai MM, Kaouk JH: J Endourol 2002; 16:A160 (abstract P28-21).

endoscopic gastrointestinal anastomosis (GIA) stapler involvement. Over a mean follow-up of 9.5 months (5 to
was safely fired (Figure 13-7). 16) no patient showed evidence of local recurrence, and
Intraoperative laparoscopic ultrasonography with 1 patient developed pulmonary metastasis.33
color Doppler is helpful to precisely delineate the proxi-
mal extent of the thrombus and should be used routinely.
Cytoreductive Nephrectomy
Placing the ultrasound probe over the thrombus-bearing
distal segment of the renal vein reveals the intraluminal Laparoscopic radical nephrectomy has been performed
mass without venous blood flow. This is in remarkable in the setting of cytoreductive surgery as preparation for
contrast with the sonographic findings of the proximal systemic immunotherapy. Walther et al.10 reported a
thrombus-free segment of the renal vein, which shows series of 11 patients undergoing laparoscopic cytoreduc-
evidence of turbulent flow from the vena cava without tive nephrectomy. Three procedures were converted to
any intraluminal mass. open surgery. The major complication rate in these
Histologic evaluation confirmed renal cell carcinoma in series was significantly greater than current series of
all 8 cases, in which gross tumor was found within the laparoscopic radical nephrectomy for organ-confined
renal vein. Inked surgical renal vein vascular margins were disease. The authors concluded that laparoscopy afforded
negative for cancer in all these 8 patients with gross venous quicker recovery and more rapid implementation of
248 Part III Kidney and Ureter

Table 13-6 Pre and Perioperative Data on Laparoscopic Radical Nephrectomy for Renal Tumor
with Renal Vein Involvement
Microscopic Renal Vein Level I Gross Renal Vein
Thrombus±SD (Range) Thrombus±SD (Range) p-value

Number of points 8 8 N/A

Mean age (years) 62.8±9.7 (48–72) 55.9±6.7 (44–64) 0.15

Mean BMI 29.4±4.6 (25–37) 25.9±5.5 (21–36) 0.17

Number of men/women 6/2 5/3 N/A

Number of left/right 3/5 4/4 N/A

Mean centimeter tumor size 7.8±2.9 (4.7–13) 12.4±5.4 (6–20) 0.09

Number of tumor thrombus visible on


preoperative imaging 1 8 N/A

Mean centimeter renal vein diameter on CT N/A 3.3±6.5 (3–4) N/A

Number of renal vein thrombus type

Floating intraluminal 0 7 N/A

Invasive into vessel wall 1* 1

Number of renal vein thrombus enhancement 0 2 N/A

Approach transperitoneal/retroperitoneal 2/6 7/1 N/A

Mean minutes operative time 188.8±87.3 (75–330) 195.7±39.1 (150–270) 0.68

Mean milliliter estimated blood loss 381.9±393.7 (30–1125) 353.6±334.3 (75–1000) 0.91

Number of conversion to open surgery 0 1 >0.99

Mean days hospital stay 1.9±1.1 (1–4) 2.3±0.8 (1–3)† 0.33

Postoperative complications (Number) Wound infection (1) 0 0.78

Adapted with permission from Desai MM, Gill IS, Ramani AP: J Urol 2003; 169:487.
*Questionable appearance on preoperative CT.
†Excludes patient treated with open conversion.

treatment with interleukin-2 (37 days—laparoscopy with laparoscopic radical nephrectomy. After overcoming the
morcellation versus 67 days—open surgery; p-value: learning curve, laparoscopic radical nephrectomy was
0.006). Moreover, since accurate staging of the primary shown to be 12% less expensive than open radical
tumor is not a primary concern in the setting of metasta- nephrectomy. Nonetheless, the intraoperative costs,
tic disease, the authors suggested that tumor morcella- mainly related to disposable instrumentation and a
tion should be performed to further reduce surgical slightly longer operative time, were 33% greater in
trauma.10 comparison to the open radical nephrectomy. Post-
operatively, the reduced nursing care and decreased hos-
pitalization time resulted in a 68% lower postoperative
Financial Analysis
costs.34 Furthermore, when the intangible nonhospital
Financial implications of laparoscopic radical nephrec- economic factors are considered, such as earlier patient
tomy are important factors in determining its wide dissem- return to the work-force, and increased productivity
ination within the urologic community. At our institution, laparoscopic approach becomes even more financially
we recently addressed this issue of cost-effectiveness of advantageous and attractive.
Chapter 13 Laparoscopic Radical and Partial Nephrectomy 249

Figure 13-7 Thrombus bearing distal renal vein is clearly demarcated from uninvolved,
proximal, flattened vein, and transected with a GIA stapler adjacent to the Vena Cava.

LAPAROSCOPIC PARTIAL NEPHRECTOMY


and adequate preoperative correction. Morbid obesity
Current Indications and Contraindications
and a history of prior renal surgery may prohibitively
Initially, laparoscopic partial nephrectomy was reserved increase the technical complexity of the procedure, and
for only the select patient with the favorably located, should be considered a relative contraindication for laparo-
small, peripheral, superficial, exophytic tumor.35–38 After scopic partial nephrectomy at this time.
gaining experience, we have carefully expanded our indi- Three-dimensional computed tomography, with vol-
cations to include select patients with more complex ume-rendered video reconstruction, is an important tool
tumors; those invading deeply into the parenchyma up to to determine candidacy for laparoscopic partial nephrec-
the collecting system or renal sinus, completely tomy and to aid in preoperative planning.40 Finally, a
intrarenal tumor, tumor abutting the renal hilum, tumor substantive laparoscopic partial nephrectomy involves
in a solitary kidney, or a tumor substantial enough to renal hilar control, transection of major intrarenal ves-
require hemi-nephrectomy.6,39 Laparoscopic partial sels, entry into and repair of the collecting system (usu-
nephrectomy for these complex tumors is performed ally), suture-control of parenchymal blood vessels, renal
only in the setting of compromised or threatened global parenchymal reconstruction, all under ischemia condi-
nephron mass wherein nephron preservation is an impor- tions. Therefore, significant laparoscopic experience,
tant consideration. In 2004, our absolute contraindica- including expertise with time-sensitive intracorporeal
tions for laparoscopic partial nephrectomy include renal suturing is essential.
vein thrombus, multiple (>3) renal tumors, and 2 mid-
pole, central, completely intrarenal tumor.40 Patients
Technique
with a bleeding diathesis or renal failure-induced platelet
dysfunction, and those on anticoagulant therapy, are at Laparoscopic partial nephrectomy (Table 13-7) can be
significantly increased risk for postoperative hemor- performed either transperitoneally or retroperitoneally.
rhage, and should be approached only with great caution Our technique has been described elsewhere.6,39
250 Part III Kidney and Ureter

Table 13-7 Technical Steps of Laparoscopic Figure-of-8 sutures are specifically placed at the site of
Partial Nehprectomy visible transected intrarenal vessels. Finally, the renal
Cystoscopy, ureteral catheterization parenchyma is reconstructed using 0 Vicryl suture over
prefashioned surgicel bolsters to complete a hemostatic
Port placement renorrhaphy (Figure 13-11). A Jackson–Pratt drain is
placed in all patients undergoing pelvicaliceal repair or if
Hilar dissection/preparation for cross-clamping there is any concern about hemostasis.
Mobilize kidney
Laparoscopic Renal Hypothermia
Identify tumor
At the Cleveland Clinic, we have recently developed a
Laparoscopic ultrasonography and circumferential scoring laparoscopic technique of renal hypothermia with ice-
Renal hypothermia (if needed) slush by laparoscopic techniques and evaluated its effi-
cacy in 10 initial patients.41 An Endocatch-II bag is
Hilar cross-clamping (preceded by IV Mannitol) placed around the completely mobilized and defatted
kidney, and its mouth is closed around the intact renal
Tumor excision hilum. The renal hilum is occluded en bloc with a
Pelvicaliceal repair (if necessary) Satinsky clamp. The bottom of the bag is retrieved
through a 12-mm port site and cut open. Finely crushed
Specific figure-of-8 sutures to control transected ice-slush is rapidly introduced into the bag to completely
parenchymal vessels surround the kidney, thereby achieving renal hypother-
mia (Figure 13-12). Pneumoperitoneum is reestablished,
Hemostatic renorrhaphy over bolsters
and laparoscopic partial nephrectomy is performed after
Unclamp hilum, confirm hemostasis opening the bag and removing the ice from the vicinity
of the tumor. Mean time required to introduce 600 to
Jackson–Pratt drain 750 cc of ice-slush around the kidney was 4 minutes.
Using a needle thermocouple probe core renal tempera-
Adapted with permission from Gill IS, Desai MM, Kaouk JH, et al: ture was documented to drop to the 5 to 19 ˚C range.
J Urol 2002; 167:469.
Our laparoscopic technique of renal surface contact
hypothermia with ice-slush mirrors the method routinely
Selection of laparoscopic approach depends on location used during open partial nephrectomy.41 Further refine-
of the tumor. The transperitoneal approach is preferred ments in the laparoscopic ice delivery system are neces-
for anterior, lateral, and upper-pole tumors. Posterior sary to increase the efficiency of introduction of ice
or posterolaterally located tumors may be better around the kidney.
approached retroperitoneoscopically. A ureteral catheter
is inserted cystoscopically up to the renal pelvis.
Results
Essentially, our operative strategy involves preparation
of the renal hilum for cross clamping, followed by In our initial 50 patients, mean tumor size was 3 cm
mobilization of the kidney and isolation of the tumor. (range 1.4–7), and partial nephrectomy was performed
Transperitoneally, a Satinsky clamp is employed to for an imperative indication in 48% of patients. Warm
occlude the renal hilum en bloc (Figure 13-8). ischemia time was 23 minutes, mean operative time was
Conversely, during retroperitoneoscopy, the renal artery 3 hours, and mean hospital stay was 2.2 days. On pathol-
and vein are dissected separately for individual placement ogy, renal cell carcinoma was confirmed in 68% of
of Bulldog clamps (Figure 13-9). Laparoscopic ultra- patients, all with a negative surgical margin.6
sonography precisely delineates tumor size, intra- More recently, we have retrospectively compared 200
parenchymal extension, distance from renal sinus, and patients undergoing partial nephrectomy at The
rules out any preoperatively unsuspected secondary renal Cleveland Clinic either by the laparoscopic (n = 100) or
masses. An adequate margin of normal renal parenchyma open (n = 100) approach.42 Median tumor size was 2.8 cm
is scored circumferentially around the tumor under sono- in the laparoscopic group and 3.3 cm in the open group
graphic guidance. The hilum is clamped, and the tumor ( p = 0.005), and a solitary kidney was present in 7 and 28
is excised and entrapped within an Endocatch bag patients, respectively ( p = 0.001). The tumor was located
(USSC, Norwalk, CT). Injection of dilute indigo centrally in 35% and 33% of cases ( p = 0.83) and the indi-
carmine through the ureteral catheter identifies any cation for a partial nephrectomy was imperative in 41%
pelvicaliceal entry (Figure 13-10), which is suture and 54% of cases, respectively ( p = 0.001). Comparing
repaired by intracorporeal laparoscopic techniques. the laparoscopic versus open groups, median surgical
Figure 13-8 Transperitoneal partial nephrectomy. A Satinsky clamp is used to obtain en
bloc control the renal hilum.

Figure 13-9 Retroperitoneal partial nephrectomy. Bulldog clamps are used for individual
control of mobilized renal artery and vein.
252 Part III Kidney and Ureter

Figure 13-10 Retrograde injection of diluted methylene blue via retrograde ureteral catheter
clearly identifies the site of leakage.

time was 3 hours versus 3.9 hours ( p < 0.001), blood loss laparoscopic cases and no open cases ( p = 0.11); median
was 125 cc versus 250 cc ( p < 0.001), and warm ischemia width of margin was 4 mm in each group ( p = 0.11).42 No
time was 28 minutes versus 18 minutes ( p < 0.001). In the patient in the laparoscopic group developed a local or
laparoscopic and open groups, analgesic requirement was port site recurrence. No kidney was lost due to warm
20.2 mg morphine sulfate equivalent versus 252.5 mg ( p ischemic injury in this study of 200 patients. The world-
< 0.001), hospital stay was 2 days versus 5 days ( p < wide single institutional experiences with laparoscopic
0.001), and convalescence averaged 4 weeks versus 6 partial nephrectomy are detailed in Table 13-8.
weeks ( p < 0.001). No laparoscopic patient was converted
to open surgery. The laparoscopic group had a higher
Complications
incidence of intraoperative complications (5% versus
0%; p = 0.02) including hemorrhage (n = 3), ureteral In the first 100 consecutive patients undergoing laparo-
injury (n = 1), and bowel serosal injury (n = 1). scopic partial nephrectomy at the Cleveland Clinic, com-
Postoperative complications (9% versus 14%; p = 0.27). plications occurred in 29% of patients: intraoperative
Renal/urologic complications occurred in 7 patients in 5%, postoperative 9%, and late 15%.42 Intraoperative
the laparoscopic group and 2 patients in the open group. complications consisted of parenchymal hemorrhage in 3
Median preoperative serum creatinine (1.0 mg/dl versus patients, which were controlled laparoscopically without
1.0 mg/dl) and postoperative serum creatinine (1.0 mg/dl open conversion in each instance. Likely causes for the
versus 1.1 mg/dl) were similar ( p = 0.99). Pathology con- hemorrhage included an unoccluded renal artery, which
firmed renal cell carcinoma in 75% of patients in the was unsuspected in the setting of multiple vessels, and
laparoscopic and 85% in the open group ( p = 0.003). inadequate occlusion by the laparoscopic bulldog clamp.
Parenchymal margin of resection was positive in two Additional intraoperative complications included a
Chapter 13 Laparoscopic Radical and Partial Nephrectomy 253

Figure 13-11 Renal parenchymal repair over bolsters.

ureteral injury repaired laparoscopically by ileal replace- plication rate of 10%.6 In a multicenter European experi-
ment (1), and a small, superficial, serosal bowel abrasion ence in 53 patients, 4 cases (8%) were converted to open
repaired laparoscopically without sequelae (1). All 100 surgery, and urine leak was noted in 5 patients (10%).44
partial nephrectomies were completed laparoscopically
without open conversion.42 Postoperatively, urologic
complications occurred in 7 patients: urine leak (3) of Financial Analysis
which 1 subsided spontaneously within 7–10 days, and 2 Recently, we reviewed the financial data of 30 patients
required restenting and percutaneous CT-guided needle undergoing partial nephrectomy: laparoscopic (n = 15)
aspiration; delayed nephrectomy for secondary hemor- and open (n = 15).45 All 30 patients in this retrospective
rhage 5 days postoperatively (1); an enlarging perineal study had a normal contralateral kidney and an uncom-
hematoma successfully managed by percutaneous plicated perioperative course. Cost data, collected by the
embolization (1); and hematuria which subsided sponta- financial department of our institution, were divided into
neously with bed rest (1). Late complications after dis- intraoperative and postoperative categories. Laparoscopic
charge from the hospital occurred in 15% of patients: and open groups were comparable as regards tumor size
incisional hernia (3), wound infection (2), percutaneous (2.4 cm versus 2.5 cm; p = 0.50). Intraoperative costs were
embolization for perineal hematoma (1), congestive heart 20.1% greater for the laparoscopic group ( p < 0.001) and
failure (1), hematuria (1), urine leak (1), wound dehis- postoperative costs were 55% lesser for the laparoscopic
cence (1), pneumonia (1), and pulmonary embolism (1).42 group ( p < 0.001). Overall, hospital costs for laparoscopic
A worldwide literature review of 97 patients undergoing partial nephrectomy were 15.6% less compared to open
laparoscopic partial nephrectomy revealed a major com- surgery ( p = 0.002).45
254 Part III Kidney and Ureter

Figure 13-12 The kidney is enclosed within the deployed Endocatch II bag. Finely
crushed ice slush is being inserted into the mouth of the bag with a 30 cc syringe.
The nozzle of the syringe has been cut off to facilitate rapid ice injection.
Table 13-8 Worldwide Single Institutional Experience with Laparoscopic Partial Nephrectomy for Tumor
Postoperative Complications (Number) Wound Infection (1) 0 0.78

Mean Mean
References Mean Hilar Number Estimated Operating Mean Number Number Follow
(Number Tumor Control of Calyceal Blood Room Time Hospital Urine Complication up
of Points) Size (cm) (Number) Repair (%) Hemostasis Loss (cc) (hour) Days Leak (%) (%) (months)

McDougall37 (2) 2.5 No 0 Argon beam coagulator, 225 7.1 7 1 (50) — 17


snare

Hoznek38 (5) 2.7 Yes (4) 0 Harmonic, bipolar, glue 225 2.6 8 1 (20) 1 (20) 22
No (1)

Janetschek22 (25) 1.9 No 0 Bipolar, argon beam 287 2.7 5.8 0 3 (12) 22.2
coagulator, glue

Harmon43,* (15) 2.3 No 3 Argon beam coagulator, 368 2.8 2.6 0 0 8


bolster

Gill6 (50) 3.0 Yes 18 Suture over bolsters 270 3.0 2.2 1 (2) 6 (12) 7.2

Overall (97) — No (43) 21 (22) — 275 — — 3 (3) 10 (10) 15.3


Yes (54)

Adapted with permission from Gill IS, Desai MM, Kaouk JH, et al: J Urol 2002; 167:469.
*Two institutional experience.
256 Part III Kidney and Ureter

REFERENCES 19. Gill IS: Laparoscopic radical nephrectomy for cancer.


Urol Clin North Am 2000; 27:707.
1. Clayman RV, Kavoussi R, Soper NJ, et al: Laparascopic 20. Chan DY, Cadeddu JA, Jarrett TW, et al: Laparoscopic
nephrectomy: initial case report. J Urol 1991; 146:278-282. radical nephrectomy: cancer control for renal cell
2. Dunn MD, Portis AH, Shalav AL, et al: Laparoscopic carcinoma. J Urol 2001; 166(6):2095–2099.
versus open radical nephrectomy: a 9-year experience. 21. Barrett PH, Fentie DD, Taranger LA: Laparoscopic
J Urol 2000; 164:1153. radical nephrectomy with morcellation for renal cell
3. Ono Y, Kinukawa T, Hattori R, et al: The long-term carcinoma: the Saskatoon experience. Urology 1998;
outcome of laparoscopic radical nephrectomy for small 52:23.
renal cell carcinoma. J Urol 2001; 165:1867. 22. Janetschek G, Jeschke K, Peschel R, et al: Laparoscopic
4. Cadeddu JA, Ono Y, Clayman RV, et al: Laparoscopic surgery for stage T1 renal cell carcinoma: radical and
nephrectomy for renal cell cancer evaluation of efficacy wedge resection. Eur Urol 2000; 38:131.
and safety: a multicenter experience. Urology 1998; 23. Siqueira TM, Kuo RL, Gardner TA, et al: Major
52:773. complications in 213 laparoscopic nephrectomy cases: the
5. Portis AJ, Yan Y, Landman J, et al: Long-term follow-up Indianapolis experience. J Urol 2002; 168:1361.
after laparoscopic radical nephrectomy. J Urol 2002; 24. Cadeddu JA, Ono Y, Clayman RV: Laparoscopic
167:1257. nephrectomy for renal cell cancer: evaluation of efficacy
6. Gill IS, Desai MM, Kaouk JH, et al: Laparoscopic partial and safety: a multicenter experience. Urology 1998; 52:773.
nephrectomy for renal tumor: duplicating open surgical 25. Kaouk JH, Gill IS: Laparoscopic radical nephrectomy:
techniques. J. Urol 2002; 167:469. morcellate or leave intact? Rev Urol 2002; 4:38.
7. Parsons JK, Jarret TJ, Chow GK, et al: The effect of 26. Matin S, Gill IS: Modified Pfannensteil incision for intact
previous abdominal surgery on urological laparoscopy. specimen extraction following retroperitoneoscopic renal
J Urol 2002; 168:2387. surgery. Urology 61(4):830–832.
8. Matin SF, Gill IS, Hsu TH, et al: Laparoscopic renal and 27. Gill IS, Cherullo EE, Meraney AM, et al: Vaginal
adrenal surgery in obese patients: comparison to open extraction of the intact specimen following laparoscopic
surgery. J Urol 1999; 162:665. radical nephrectomy. J Urol 2002; 167:238.
9. Savage SJ, Gill IS: Laparoscopic radical nephrectomy for 28. Tsivian A, Sidi AA: Port site metastases in urological
renal cell carcinoma in a patient with level I renal vein laparoscopic surgery. J Urol 2002; 169:1213.
tumor thrombus. J Urol 2000; 163:1243. 29. Cicco A, Salomon L, Hoznek H: Carcinological
10. Walther MM, Lyne C, Libutti SK, et al: Laparoscopic risks and retroperitoneal laparoscopy. Eur Urol
cytoreductive nephrectomy as preparation for 2000; 38:606.
administration of systemic interleukin-2 in the treatment 30. Fentie DD, Barrett PH, Taranger LA: Metastatic
of metastatic renal cell carcinoma: a pilot study. Urology renal cell cancer after laparoscopic radical nephrectomy:
1999; 53:496. long-term follow-up. J Endourol 2000; 14:407.
11. Gill IS, Meraney AM, Schweizer D, et al: Laparoscopic 31. Castilho LN, Fugita OE, Mitre AI: Port site recurrences
radical nephrectomy in a 100 patients: a single-center of renal cell carcinoma after videolaparoscopic radical
experience from the United States. Cancer 2001; nephrectomy. J Urol 2001; 165:519.
92:1843. 32. Gill IS, McClellan BL, Kerbl K, et al: Adrenal
12. Steinberg AP, Desai MM, Kaouk JH, et al: The large involvement from renal cancer: predictive value of
(>7 cm; pT2) renal tumor: laparoscopic versus open computed tomography. J Urol 1994; 152:1082.
radical nephrectomy. J Endourol 2002; 16:A160. 33. Desai MM, Gill IS, Ramani AP: Laparoscopic radical
(Abstract P28-21.) nephrectomy for cancer with level I renal vein
13. Sung GT, Gill IS: Anatomic landmarks and time involvement. J Urol 2003; 169:487.
management during retroperitoneal laparoscopic radical 34. Meraney AM, Gill IS: Financial analysis of laparoscopic
nephrectomy. J Endourol 2002; 16:165. versus open radical nephrectomy and
14. Gill IS, Rassweiler JJ: Retroperitoneoscopic renal surgery: nephroureterectomy. J Urol 2002; 167:1757.
our approach. Urology 1999; 54:734. 35. Winfield HN, Donovan JF, Godet AS, et al: Laparoscopic
15. Gill IS, Schweizer D, Hobart MG, et al: Retroperitoneal partial nephrectomy: initial case report for benign disease.
laparoscopic radical nephrectomy: the Cleveland Clinic J Endourol 1993; 7:521.
experience. J Urol 2000; 163:1665. 36. Gill IS, Delworth MG, Munch LC: Laparoscopic
16. Gill IS, Strzempkowski B, Kaouk J, et al: Prospective retroperitoneal partial nephrectomy. J Urol 1994;
randomized comparison: transperitoneal versus 152:1539.
retroperitoneal laparoscopic radical nephrectomy. J Urol 37. McDougall EM, Elbahnasy AM, Clayman RV:
2002; 167:19. (Abstract 78.) Laparoscopic wedge resection and partial
17. Steinberg AP, Ramani AP, Abreu SC, et al: Reoperative nephrectomy—the Washington University experience
laparoscopy: Cleveland Clinic experience. J Endourol and review of literature. J Soc Laparoendosc Surg 1998;
2002; 16:A97. (Abstract P18-24.) 2:15.
18. Gill IS: Retroperitoneal laparoscopic nephrectomy. Urol 38. Hoznek A, Salomon L, Antiphon P, et al: Partial
Clin North Am 1998; 25:343. nephrectomy with retroperitoneal laparoscopy. J Urol
1999; 162:1922.
Chapter 13 Laparoscopic Radical and Partial Nephrectomy 257

39. Desai MM, Gill IS, Kaouk JH, et al: Laparoscopic partial 43. Harmon WJ, Kavoussi LR, Bishoff JT, et al: Laparoscopic
nephrectomy with suture-repair of the collecting system. nephron-sparing surgery for solid renal masses using the
Urology 2003; 61:99. ultrasonic shears. Urology 2000; 56:754.
40. Gill IS: Minimally invasive nephron-sparing surgery. Urol 44. Rassweiler JJ, Abbou C, Janetschek G, et al: Laparoscopic
Clin North Am 30(3):551–579. partial nephrectomy. The European experience. Urol Clin
41. Gill IS, Abreu SC, Desai MM, et al: Laparoscopic ice North Am 2000; 27:721.
slush renal hypothermia for partial nephrectomy: the 45. Steinberg AP, Desai MM, Gill IS, et al: Financial analysis
initial experience. J Urol 170(1):52–56. of laparoscopic versus open partial nephrectomy.
42. Gill IS, Matin SF, Desai MM, et al: Comparative analysis J Endourol (in press).
of laparoscopic vs. open partial nephrectomy for renal
tumors in 200 patients. J Urol 170(1):64–68.
C H A P T E R

14Treatment of Advanced
Renal Cell Carcinoma
Tracey Krupski, MD, Hyung Kim, MD,
Robert A. Figlin, MD,
and Arie Belldegrun, MD

Advanced renal cell carcinoma (RCC) continues to pres- can now be identified. Treatments associated with signif-
ent a major challenge for physicians. Approximately 20% icant toxicities can be reserved for patients most likely to
to 30% of patients have metastatic RCC at the time of benefit from the therapy. An accurate stratification of
diagnosis, and metastatic RCC remains resistant to con- patients based on risk factors will allow physicians to bet-
ventional oncologic therapies.1 Immunotherapy with ter interpret the results of clinical trials. As in the treat-
interleukin-2 (IL-2) and interferon-α (IFN-α) remains ment of any malignancy, the ultimate goal is complete
the standard of care for metastatic RCC, however, cure, and the remarkable examples of spontaneous
response rates remain modest at 10% to 30%, and the regression of advanced RCC hold out the hope that with
median survival for patients presenting with metastatic the right medical intervention, this is possible.
RCC is 12 to 18 months. Despite the poor prognosis
generally associated with advanced RCC, a unique char-
EPIDEMIOLOGY OF RENAL CELL CARCINOMA
acteristic of RCC is that in a small subset of patients
durable remissions and long-term survival is possible. RCC is the third most common genitourinary malig-
Spontaneous regression of primary and metastatic nancy. It is estimated that 31,500 new cases of RCC, rep-
lesions in RCC is second only to melanoma in terms of resenting 3% of all cancers, will be diagnosed in the
regression rates. The most common pattern of disease United States in 2003.7 In the United States since 1950,
regression is resolution of pulmonary metastasis after there has been a 126% increase in the incidence of RCC
nephrectomy.2 Freed et al.3 found 51 cases of regression accompanied by a 36.5% increase in annual mortality.8
of RCC. Of these, 3 had no treatment of the primary This phenomenon may partially be explained by the
lesion and 18 had histologic confirmation of regression. increased number of incidental tumors detected by wide-
A review of the available literature by Snow and spread use of radiologic imaging. However, other
Schellhammer4 in 1982 found regression of metastatic unidentified factors are important, as the incidences of
renal tumors in fewer than 1% of cases. However, regres- both localized and advanced RCC have increased. Not
sion prior to primary therapy has also been documented. until the year 2002 did this trend stabilize. The rise in
Disease stabilization, defined as a failure of sustained incidence for all stages from 1973 to 1997 was three
growth, has been documented in both primary RCC and times greater than rise in the rate of mortality.
metastatic foci.5 Oliver et al.6 observed 73 patients with Consequently, the 5-year survival for all cases has nearly
metastatic RCC who received no treatment. In this doubled from 34% in 1954 to 62% in 1996.8,9 Recently,
series, 3 patients had complete regression and 2 patients obesity has been found to be a risk factor for death in sev-
had partial regression of disease. In addition, 5% of eral cancers, including kidney cancer. Calle et al.10 exam-
patients had stable disease. ined how obesity influenced the death rate from kidney
To treat RCC better, efforts have been directed at cancer. A patient’s degree of obesity was determined by
delineating the variables important in determining prog- their body-mass index and categorized according to the
nosis. Patients most likely to progress and die from RCC World Health Organization’s classification system of

258
Chapter 14 Treatment of Advanced Renal Cell Carcinoma 259

normal range, grades 1, 2, and 3 overweight. As the Survival Using 4.5 cm Cut-off
degree of obesity increased, so did the relative risk of
death from cancer with patients in the highest group hav- 100
ing a 1.7 relative risk. ⱕ4.5 cm
75
P = 0.0001

Survival (%)
Prognosis
P = NS 4.5 –7.0 cm
60
The natural history of RCC is complex and many factors
impact the biologic behavior of these tumors. Various >7.0 cm
prognostic models that integrate clinically relevant vari- 25
ables have been proposed. The evaluation of biomolecu-
lar markers for staging and determining prognosis is an 0
active area of investigation. However, until these biomol- 0 12 24 36 48 60 72
ecular markers have been rigorously tested and validated, Time from Nephrectomy (months)
clinical variables, such as stage, grade and performance
status, will continue to be the most important variables Figure 14-1 The 4.5-cm cutoff provides optimal stratification
for assessing prognosis. of early-stage kidney cancer. This modification has been
proposed to the TMN system.

Tumor Stage
Clinic’s experience with collecting system invasion is sim-
Stage categorizes the anatomic spread of RCC and is rec- ilar. Clear cell carcinoma most commonly invades, and
ognized as the most important prognosticator for RCC. when the stage is high, urothelial involvement has little
Currently, the two most commonly used staging systems impact on prognosis. However, low stage T2 lesions have
are the stages described by Flocks and Kadesky11 and a worse prognosis when urothelial invasion is involved.22
modified by Robson12, and the TNM system proposed
by the Union Internationale Contre le Cancer (UICC).13
Grade and Histology
The TNM system has been modified twice since its
introduction. The change in 1997 entailed expanding the The initial report correlating grade and patient outcome
T1 stage from <2.5 to <7 cm. Tumor thrombus above the was published in 1932.23 In 1971, Skinner and colleagues
diaphragm became T3c instead of T4 and thrombus noted the correlation between nuclear features and sur-
below the diaphragm was changed to T3b.14 Several vival, and Fuhrman et al.24 subsequently developed the
studies have validated the latest modification of the findings into a 4-tier grading system. Grade 1 has no
TNM system.15–17 However, others have suggested that observable nucleoli, grade 2 has small nucleoli, grade 3
further changes to the 1997 modification will increase has prominent nucleoli, and grade 4 has large pleomor-
the discriminatory power of the staging system.18,19 phic atypical cells with prominent nucleoli. Analysis of
The 1997 TNM staging system uses a 7-cm cutoff patients from UCLA found the 5-year cancer-specific
between T1 and T2 tumors. The T1 tumors are further survival based on tumor grade is 89% for grade 1, 65%
stratified into those <4 cm, which are T1a, and those for grade 2, and 46% for grades 3 and 417 (Figure 14-4).
greater than 4 cm, which are T1b. Using the UCLA Inspection of the Mayo clinic data revealed similar 5-year
database, various cutoffs for separating T1 and T2 pri- crude survival rates based on histologic grade (Zincke,
mary tumors were evaluated. Patients were optimally personal communication).
stratified when a 4.5-cm cutoff was used (Figure 14-1). In While nuclear grading clearly impacts outcome, the
addition, the difference in survival was not statistically effect of histologic subtype on outcome is more variable.
different between patients with 4.5 and 7-cm tumors and There are five main histologic subtypes of RCC recog-
patients with tumors >7 cm.18 In a similar analysis of nized as a result of a collaborative effort between the
patients who underwent a partial nephrectomy, Hafez et UICC and the American Joint Committee on Cancer
al.20 suggested that a 4-cm cutoff should be used in the (AJCC) in 1997.14 Clear cell carcinomas account for 70%
TNM staging system. Recently, collecting system inva- to 80% of RCC tumors while papillary tumors account
sion has been analyzed with respect to survival rates. for 10% to 15% and both arise from the proximal
Analysis of 895 patients found 124 patients with collect- tubules. Chromophobe tumors arise from the interca-
ing system invasion. Multivariate analysis revealed a 1.4 lated cells of the collecting ducts and account for 5% of
times increased risk of death after controlling for tumor kidney cancers. Collecting duct or medullary carcinomas
grade and stage. The difference in 3-year survival is most of the kidney are a rare and very aggressive form of kid-
apparent in stage 1 tumors, where noninvaders had an ney cancer. Chao et al.25 reported on 6 patients with col-
81% survival versus 67% for invaders.21 The Cleveland lecting duct tumors. Tumors affected patients at a
260 Part III Kidney and Ureter

younger age, presented with stage IV disease, and had an action between multiple prognostic factors. Using the
average survival of 11.5 months. Renal medullary carci- UCLA kidney cancer database, 14 clinical variables were
noma occurs almost exclusively in African-Americans assessed for inclusion in a prognostic model. The 1997
with sickle cell trait or disease and has a very poor prog- TNM stage, tumor grade, and ECOG-PS were identi-
nosis. Sarcomatoid RCC is no longer considered a dis- fied as strong predictors of prognosis and were combined
tinct histologic subtype but rather a high-grade form of to develop the 5-tiered UCLA integrated staging system
RCC that portends poor prognosis. Sarcomatoid features (UISS) (Figure 14-2). Using the UISS the projected 2-
are present in <5% of RCC and are typified by a spindle and 5-year survivals for patients in groups are: I, 96% and
cell growth pattern.26 Ro et al.27 noted that in low stage 94%; II, 89% and 67%; III, 66% and 39%; IV, 42% and
disease, 2 factors independently predictive of a poorer 23%; V, 9% and 0% (Figure 14-3). This novel system for
prognosis were the amount of tumor necrosis and pro- staging and predicting survival for patients with RCC is
portion of sarcomatoid tumor. Renal tumors that do not simple to use and superior to using stage alone in pre-
fit into one of the five histologic categories are desig- dicting survival.36
nated as unclassified renal tumors.28 As molecular classi- More recently, a simplified UISS algorithm has been
fication becomes available, it is expected that these developed that assigns patients with localized and
tumors will either be categorized as one of the estab- metastatic RCC into either low, intermediate or high-
lished subtypes or a new subtype will be defined. risk groups.33 For each of the risk groups, a relevant set
Each subtype has distinctive characteristics and biol-
ogy that predict tumor behavior. Amin et al.29 reviewed ULCA Integrated Staging System (UISS)
405 cases of renal tumors and assessed the role of histol-
ogy in predicting outcome. They reported 5-year dis- TNM ECOG Overall
UISS Grade
ease-specific survivals of 100%, 86%, 76%, and 24% for Stage PS 5 Yr Survival (%)
chromophobe, papillary, clear cell, and unclassified 0 1– 2
I I 94
tumors, respectively. However, the limited number of
I 0 3–4 67
histologic cases in each group precluded controlling for II
II any any
stage. Motzer et al.30 recently analyzed 64 patients with
III 0 any
metastatic non-clear cell RCC and reported that patients
III ≥1 1
who had chromophobe tumors also fared better than
III III ≥1 2–4 39
patients who originally had collecting duct or papillary
IV 0 1–2
histology. In 12 patients with chromophobe histology, 4
IV 0 3–4
patients had survivals of over 3 years. IV 23
IV ≥1 1–3
V IV ≥1 4 0
Patient Performance Status
Figure 14-2 From 14 possible variables, TMN stage, tumor
The Karnovsky performance scale and the Eastern grade and ECOG-PS proved to optimally stratify patient
Cooperative Oncology Group performance status prognosis.
(ECOG-PS) scale measure the impact of multiple objec-
tive and subjective symptoms on the patient’s overall
UCLA Integrated Staging System
functional status. The ECOG-PS scale at the time of (UISS)
presentation is an important prognostic factor in patients 100
I
with metastatic RCC and it is useful for selecting patients
for treatments.31,32 ECOG-PS is an independent predic-
75
tor for survival in patients with both metastatic and local-
ized disease. Patients with RCC and an ECOG-PS score II
of 0 had a 81% 5-year survival while patients with an 50
III
ECOG-PS score 1 to 4 had a 51% 5-year survival.33
Performance status was found by Elson34 to be one of the
25
primary determinants of prognosis. Further, a decreased IV
performance status was associated with reduced survival
P = 0.001 V
in a Memorial Sloan Kettering study by Motzer et al.35 0
0 12 24 36 48 60 72 84 96
The UCLA Integrated Staging System Months

Recent studies suggest that the ultimate clinical behavior Figure 14-3 The UISS stratifies patients beyond stage alone.
of RCC can be predicted by considering a complex inter- The difference in survival curves is clearly shown.
Chapter 14 Treatment of Advanced Renal Cell Carcinoma 261

of clinical outcome data was generated, including overall patient with metastatic RCC to the chest or abdomen
survival, disease-specific survival, freedom from recur- should have an MRI or CT of the brain. A bone scan
rence in nonmetastatic (NM) patients, outcome after should be obtained in any patient with bone pain, ele-
recurrence in NM patients, and freedom from progres- vated serum calcium or metastasis noted elsewhere.
sion in metastatic (M) patients undergoing cytoreductive Paraneoplastic findings are common in RCC, and the
nephrectomy and immunotherapy. This outcome data history and laboratory evaluation should be directed to
can now be used to predict prognosis of other patients. assess for cachexia, fever, night sweats, hypercalcemia,
The Mayo Clinic also has developed an outcome predic- polycythemia, anemia, and hepatic dysfunction.
tion model called the SSIGN score. SSIGN represents A brain metastasis has the potential for producing
stage, size, grade, and necrosis. Stage relates to the 1997 severely debilitating and crippling effects. Any brain
TMN system, while size refers to tumors >5 cm. This metastasis, regardless of the total extent of the disease,
scoring system is designed to predict outcomes of patients should be treated with radiation, surgery or both, prior to
with clear cell carcinoma.37 A postoperative nomogram administering any further treatment. Regardless of
also exists for RCC. Kattan’s nomogram predicts the 5- organ, solitary metastatic lesions amenable to surgery
year freedom from failure and utilizes the factors symp- should be resected. Although randomized studies are not
toms, histology, pathologic stage, and tumor size.38 available, surgical extirpation provides palliative benefits
Other stratification systems that address prognosis and may extend survival.40–42 One study found 66% of
have been described. In 1998 Elson et al.34 developed a patients treated with immunotherapy and surgical resec-
scoring system to determine the prognosis of advanced tion of lung metastasis were alive with a median follow-
RCC by stratifying patients into 5 groups based on up of 48 months.43 Slaton et al.44 reported on 15 patients
ECOG-PS performance status, time from diagnosis to who underwent resection of the primary tumor with con-
metastasis, weight loss, previous chemotherapy, and current resection of a metastatic site (8 lung, 3 bone, 2
number of metastatic sites.34 Most recently, Motzer et nodal, 1 liver, and 1 contralateral adrenal). Nine received
al.35 developed a model based on a study of 670 patients adjuvant immunotherapy. Seven had no evidence of dis-
with advanced RCC treated at Memorial Sloan Kettering ease, and 4 had stable metastasis at 17 months.
Cancer Center by defining the relationship of pretreat-
ment clinical features and survival. Features associated
Inferior Vena Cava Involvement
with shorter survival on multivariate analysis included
decreased performance scale, increased serum lactate Extension of tumor thrombus into the inferior vena cava
dehydrogenase level, hypercalcemia, and having the pri- (IVC) often poses a challenge to surgeons. However, sev-
mary tumor in place. eral studies show it can be safely done.44,45 Zisman et al.45
Paraneoplastic signs and symptoms occur in 20% of compared surgical complications in patients with
patients diagnosed with RCC. Polycythemia, hypercal- metastatic disease undergoing resection of the kidney
cemia, and hepatic dysfunction are the results of proteins and caval thrombus with patients undergoing a radical
elaborated by the tumor and constitute a paraneoplastic nephrectomy alone. There was no increase in surgical
syndrome. Anemia of hypoalbuminemia is thought to be complications. Zisman et al.45 further found that in
related to tumor volume. While in univariate analysis patients who had isolated caval involvement but no
each of these impacts disease-specific survival signifi- metastasis, the survival was not statistically different from
cantly, multivariate analysis found only hypoalbumine- those without the thrombus. In another retrospective
mia, weight loss, anorexia, and malaise correlated with analysis, patients with IVC thrombus and metastatic
disease-specific survival. This analysis controlled for RCC who underwent both a nephrectomy and received
TMN stage, ECOG-PS, and Fuhrman grade. The pres- immunotherapy had better survival than patients treated
ence of any one of these findings constitutes cachexia and with either therapy alone.46
impacts survival. Patients with localized RCC had a sig-
nificantly worse disease-specific survival if they had a
Lymph Node Involvement
cachexia-related finding, as did those patients with
metastatic RCC.39 The older literature regarding lymphadenopathy reported
conflicting results.47–49 The only randomized phase III
trial to address the benefits of lymph node dissection
Evaluation of Patient
during radical nephrectomy for patients with resectable,
All patients diagnosed with RCC require a thorough nonmetastatic RCC was performed by the European
workup for metastatic disease. In addition to a meticu- Organization for the Research and Treatment of Cancer
lous history and physical exam, the abdomen and pelvis Genitourinary Group in study # 30881. In this study, the
should be imaged with a CT scan. A CXR or chest CT dissection extended from the crus of the diaphragm infe-
should be obtained to rule out pulmonary metastasis. Any riorly to the bifurcation of the aorta. The study random-
262 Part III Kidney and Ureter

ized 772 patients to undergo standard nephrectomy or for patients with metastatic RCC became a source of
nephrectomy with node dissection. The preliminary debate. In the 1980s, IFN-α was established, an effective
results have been reported. After preoperative staging, the therapy for metastatic RCC.54 In 1992, the Food and
incidence of unsuspected lymph node metastasis was Drug Administration approved recombinant human IL-2
3.3%. The complication rates were not increased in the monotherapy for RCC.
group undergoing lymph node dissection.50 Several authors have noted a response to IL-2
Retrospective studies evaluating the role of lym- immunotherapy with the primary tumor in place.55
phadenectomy in patients with clinically bulky lymph However, advocates of neoadjuvant nephrectomy argued
nodes suggest that lymph node dissection will improve that the primary tumor may produce an immunosuppres-
survival. Vasselli et al.51 examined survival in patients sive effect and that neoadjuvant nephrectomy will further
with metastatic RCC. The patients without clinical evi- improve the response to immunotherapy. Another argu-
dence of lymphadenopathy had a 14.5-months survival as ment for neoadjuvant nephrectomy was that the primary
compared to an 8.5 median survival of those with positive tumor rarely responses to immunotherapy. Wagner
nodes. Furthermore, survival was 3 months longer in et al.56 at the National Cancer Institute reported a 6%
those who underwent complete, or even incomplete, sur- extrarenal response to immunotherapy; however, there
gical extirpation of nodes as opposed to those deemed were no responses of the primary tumor.
unresectable. The UCLA experience confirms the find- One of the primary concerns with neoadjuvant
ing that clinically positive nodes in the setting of metasta- nephrectomy was that surgery would delay or even pre-
tic disease are associated with poor prognosis. In those clude administration of immunotherapy.57 However,
patients, lymphadenectomy improves survival when per- several studies have examined this issue and found that
formed on carefully selected patients undergoing cytore- immunotherapy can be given in a timely manner fol-
ductive nephrectomy prior to immunotherapy. It was lowing surgery. The median time to delivery was
noted that bulky lymph nodes almost never respond to 40 days.58 Laparoscopic nephrectomy has also been
immunotherapy and should be resected when possi- advocated as a modality that would decrease time to
ble52,53 (Figure 14-4). immunotherapy.59
Recently, the results of two multi-institutional ran-
domized studies have been published and they establish
METASTATIC RENAL CELL CARCINOMA
cytoreductive nephrectomy, and immunotherapy
Role of Nephrectomy
improves survival in advanced RCC. Southwest
Before the era of immunotherapy, the natural history of Oncology Group (SWOG) trial 8949 began in 1989 and
metastatic RCC was not improved by debulking randomized 246 patients with metastatic RCC into 2
nephrectomy. deKernion et al.2 published the survival arms. Patients in first arm were treated with radical
rates in patients with metastatic disease treated with nephrectomy and IFN-α, and patients in the second arm
nephrectomy alone. The 2- and 5-year survival rates were treated with IFN-α alone. Although the response
were 25% and 13%, respectively. Nephrectomies were rates were similar, the median survival was 8 months in
reserved for palliation of local symptoms, such as pain the interferon-only arm compared to 12 months in the
and hematuria. However, with the advent of modern surgery in first arm (p = 0.02). The trend for increased
immunotherapy, the role of cytoreductive nephrectomies survival was maintained for all stratification factors,
including measurable disease, performance status, and
Metastatic Patients and LND site of metastasis.60
1.00 A multi-institutional, phase III study from Europe
reached a similar conclusion. The EORTC-GU trial
0.75 30947 applied the same protocol as the SWOG study.
The two arms included a combination arm consisting of
Survival

0.50 Treated with IMT nephrectomy followed by immunotherapy and a control


LND LND 63% arm that received immunotherapy alone. Both time to
No LND 71% progression (p = 0.04) and survival (p = 0.3) were signifi-
0.25 P = 0.0002
n = 107 cantly improved in the combination arm over the control
0.00 No LND n = 22 arm. Median survival was only 7 months in the interferon
0 12 24 36 48 60 72 84 96 only arm and it was 17 months in the surgery plus inter-
feron arm.61 These two trials are the first prospective
Months
studies demonstrating the benefit of nephrectomy in the
Figure 14-4 Among patients with metastatic RCC who are modern immunotherapy era.
treated with immunotherapy, lymphadenectomy appears to Following the publication of the SWOG study,
confer a survival advantage. Pantuck et al.62 studied patients treated with nephrectomy
Chapter 14 Treatment of Advanced Renal Cell Carcinoma 263

and IL-2-based immunotherapy at UCLA. They retro- antidepressants are often used prophylactically as well. In
spectively identified 89 patients who met the preopera- an effort to minimize toxicity, various combinations of
tive eligibility criteria used in the SWOG study. The agents at various dosing schedules have been proposed.
median survival in these patients was 16.7 months with a In the only randomized study to demonstrate a sur-
5-year survival of 19.6%. Figure 14-5 shows the vival advantage for IFN-α, patients were randomized to
Kaplan–Meier survival curve for this group superim- receive vinblastine alone versus IFN-α and vinblastine.
posed on survival curves for the two groups in the The median survival for the vinblastine only arm was
SWOG study. An important caveat is that these studies 37.8 weeks and for the combination arm was 67.6 weeks
enrolled and evaluated patients with favorable clinical (p = 0.0049).65 In an early report of a randomized trial
features, such as good performance status and minimal comparing IFN-α versus medroxyprogesterone acetate
number of metastatic sites. Therefore, the results of (MPA), the 1-year survival in the IFN-α group was 43%
these studies may not apply to patients with higher-risk compared to 31% in the MPA group.66
features. In other phase III trials, the addition of 5-fluorouracil
(5-FU) or 13-cis-retinoic to IFN-α-based regimens did
not significantly impact survival.64,67 In a randomized
SYSTEMIC THERAPY
trial, 284 patients were treated with IFN-α alone or IFN-α
RCC is refractory to chemotherapy and radiation plus 13-cis-retinoic acid. While progression free rates and
therapy. However, it is responsive to systemic cytokine overall survival were not statistically different, the
therapy. The most commonly used agents include IFN-α median duration of response was higher in the group that
and IL-2. received both IFN-α and 13-cis-retinoic acid, suggesting
and 13-cis-retinoic acid may enhance the durability of the
response to IFN-α68 An ongoing phase III ECOG trial is
Interferon-α
evaluating IFN-α alone versus IFN-α and thalidomide.
IFN-α, originally described as an antiviral agent, has The role of IFN-α as adjuvant therapy for high risk,
broad immunoregulatory properties and has been shown localized RCC has also been evaluated in a randomized
to be effective in metastatic RCC. In one study, 1042 trial. An Italian study randomized 264 patients to IFN-α
patients treated with IFN-α demonstrated an overall three times a week for 6 months or to observation fol-
response rate of 12% with an average response time of lowing nephrectomy. After 62 months, the overall sur-
4 months.63 The dosing regimen of 5 to 20 million U/d of vival was 66% for both groups but disease-free survival
IFN-α appears to be effective with minimal toxicity.64 The was slightly lower in the IFN-α group.69 Therefore,
toxicity associated with INF-α is usually seen several there is currently no role for adjuvant immunotherapy in
weeks after initiating treatment and includes malaise, patients without documented metastatic disease.
myalgia, fever, chills, anorexia, weight loss, depression, and
decreased cognitive function. Prophylactic use of aceta-
Interleukin-2
minophen, NSAIDs, and antihistamines is routine, and
IL-2 was approved in 1992 by the Food and Drug
SWOG & UCLA Administration (FDA) for the treatment of mRCC based
on a 14.5% response rate in 255 patients treated with
100 Retrospective high-dose IL-2. A follow-up study of the 255 patients
originally presented to the FDA was published 8 years
P < 0.05
80
later. The median survival for all 255 patients was 16.3
months and 10% of all patients remained alive and dis-
ease free.70 IL-2 does not have any direct cytotoxic
60
Survival

effects; however, it induces the immune system to pro-


duce a cytotoxic response against RCC.
40 IL-2 is associated with significant toxicity due to vas-
cular permeability.71 Major toxicities include vascular
Nx + IL-2
20
collapse, vascular leak syndrome, cardiac arrhythmias,
Nx + IFN
fever, chills, diarrhea, hepatic dysfunction, and renal fail-
IFN
ure. IL-2 is administered either as low- or high-dose reg-
0
imen. The high-dose regimen needs to be administered
0 24 48 72 96
in a closely monitored inpatient setting with ready access
Figure 14-5 Kaplan–Meier survival curves for patients to vasopressors. Patients should have normal or mini-
undergoing cytoreductive nephrectomy followed by either mally impaired cardiac, renal, and pulmonary function.
interferon or interleukin immunotherapy. Approximately 50% of patients required vasopressor sup-
264 Part III Kidney and Ureter

port in one study.72 The high-dose regimen should TIL cells due to processing failure. At the present time,
preferably be used at referral centers with experience in it may not be technically feasible to consistently deliver
administering high-dose IL-2 and managing related TIL-based treatment in a multi-institutional fashion.
complications. The low-dose IL-2 regimen is associated
with significantly less toxicity and can be administered on
Active Immunotherapy
an outpatient basis.
A review of 10 trials with over 500 patients involving Active immunotherapies sensitize effector cells (T lym-
high-dose intravenous IL-2 revealed a response rate of phocytes) to target tumor antigens. A major obstacle in
19% with a median duration of 22 months.73 The the search for an effective agent is the lack of tumor-spe-
response rates in studies using low-dose IL-2 with or cific antigen.81 However, carbonic anhydrase IX (CA-9)
without IFN-α were 11% to 17%74,75 These results are has recently been identified as a potential target. CA-9 is
comparable to those seen with high-dose IL-2; however, the only characterized protein that is widely expressed in
high-dose IL-2 was more likely to produce complete RCC with 95% to 100% of the clear cell variant demon-
responses that resulted in longer disease-free intervals. In strating expression. Further, this expression is specific to
a randomized 3 arm comparison of high-dose intra- the primary tumor and metastatic deposits.82,83 The rea-
venous, low-dose intravenous and intermediate-dose son for this overexpression is a direct consequence of a
subcutaneous IL-2 regimens, the response rates were mutation in the von Hippel-Lindau gene, which nor-
16%, 4%, and 11%, respectively.57 However, long-term mally suppresses CA-9.
survival results, which may confirm the improved dura- A better understanding of the immune system and the
bility of responses seen with higher-dose regimens, are molecular changes involved in tumor formation provide
not yet available. Recent work by the Cytokine Working insight into better treatment strategies. An effective
Group examined the role of high-dose IL-2 in patients tumor vaccine needs to recruit cytotoxic T cells specific
with locally advanced as well as M1 disease who were for RCC and antigen presentation is the first step.
completely resected. They found no improvement in Dendritic cells (DC) are the primary antigen presenting
survival with the addition of IL-2 in these high-risk pop- cells for stimulating T cell mediated immune responses,
ulations (ASCO 2003). and thus, manipulation of DC ex vivo to express tumor-
specific antigen may yield a potent vaccine.84,85 In animal
studies, DC-based tumor vaccines were more effective
NOVEL THERAPIES
when combined with systemic cytokines. An elegant
Passive Immunotherapy
solution to avoid the toxicity of systemic therapy yet
Immunotherapy is characterized as active when the agent retain the synergy with a vaccine would be to engineer
induces a state of immune responsiveness against the the DC ex vivo to express tumor antigen, as well as an
tumor in the host or as passive when the immunologic immunomodulatory cytokine, such as GM-CSF. This
agents are directly active against the tumor. Examples of allows for lower systemic levels of cytokine yet keeps
passive immunotherapy include monoclonal antibody high levels in the tumor itself. At UCLA, preclinical
therapy and adoptive immunotherapy. The transfer of studies designed to assess the effectiveness of a fusion
cells, which can mount an antitumor response, is coined protein consisting of CA-9 and GM-CSF are currently
adoptive immunotherapy. An example of adoptive ther- underway.
apy utilizes tumor-infiltrating lymphocytes (TIL), which Heat shock protein represents another potential tar-
are derived from the tumor and adoptively transferred to get for treating RCC. Heat shock protein normally
the tumor-bearing host. In 1980, the NCI isolated infil- functions to allow proper folding of intracellular
trating lymphoid cells from solid tumors.76 These cells polypeptides and aids with intracellular transport. They
were expanded ex vivo using IL-2 and shown to have have been associated with degraded proteins from
direct antitumor activity.77 In a murine model, TIL were tumor. Heat shock protein vaccines have been shown to
effective in eliminating micrometastases.78 be effective against RCC in a clinical trial.86 A pivotal,
The initial single-institution studies showed great 500-subject phase III trial of adjuvant HSP vaccine
promise. Figlin et al.79 reported a 34.6% overall response (Oncophage-Antigenic) in high-risk RCC is underway.
rate to treatment with TIL. This lead to a multicenter Future studies will likely combine the heat shock pro-
randomized trial comparing CD8+TIL combined with tein with IL-2 or GMCSF.
IL-2 versus IL-2 alone. The response rate in the combi-
nation arm was 9.9% compared to 11.4% in the IL-2
Antiangiogenic
alone arm. This lead the investigators to conclude that
the combination of TIL and IL-2 was no better than IL-2 In RCC, increase in vascular endothelial growth factor
alone.80 However, it should be noted that 41% of the (VEGF) and angiogenesis is a direct consequence of the
subjects randomized to the TIL arm did not receive the VHL mutation commonly found in clear cell RCC. An
Chapter 14 Treatment of Advanced Renal Cell Carcinoma 265

understanding of this mechanism provides a straightfor- complete responders remained in remission for an aver-
ward rationale for targeting angiogenesis in RCC. age of 22 months.92
Bevacizumab is a monoclonal antibody designed to neu-
tralize VEGF. A randomized placebo-controlled study is
SUMMARY
ongoing to evaluate the effectiveness of high- and low-
dose bevacizumab.87 In an early report of 110 patients Advanced RCC remains as a difficult problem for physi-
randomized on the study, there was a highly significant cians. For metastatic RCC, immunotherapy with IFN-α
prolongation in time to progression of high-dose beva- or IL-2 remains as the standard of care, with typical
cizumab compared to placebo. Three partial responses response rates of about 20%. While nephrectomy fol-
were seen in the high-dose group and none in the low- lowed by immunotherapy appears to be the most effica-
dose or placebo groups. cious treatment, careful patient selection is imperative.
Improved ability to define patient prognosis allow us to
better select patients for various treatments. In an effort
Chemotherapy
to improve response rates and minimize toxicity, novel
Gemcitabine is the latest chemotherapeutic agent to be approaches continue to be explored. Active areas of
used in clinical trials. A phase I trial initially evaluated investigation include vaccines therapies, bone marrow
multiple dosing regimens of 5-FU with weekly gemc- transplantation and targeted pharmaceuticals.
itabine. The results of this work suggested antitumor
activity against RCC, in particular. The most efficacious
dosing regimen was defined by this study as well.88 A REFERENCES
multiinstitutional phase II study used gemcitabine 600
mg/m(2) on days 1, 8, and 15 with continuous infusion of 1. Kirkali Z, Tuzel E, Mungan MU: Recent advances in
5-FU 150 mg/m(2)/day for days 1 to 21. While no kidney cancer and metastatic disease. BJU Int 2001;
patient had a complete response, 7 of 39 had a partial 88:818–824.
response (17%).89 To increase this objective response 2. deKernion JB, Ramming KP, Smith RB: The natural
rate, additional agents have been added to the regimen in history of metastatic renal cell carcinoma: a computer
analysis. J Urol 1978; 120:148–152.
a search for synergy.
3. Freed SZ, Halperin JP, Gordon M: Idiopathic regression
Neri et al.90 combined 1000 mg/m gemcitabine intra- of metastases from renal cell carcinoma. J Urol 1977;
venously on days 1, 8, 15, and 28 with intramuscular 118:538–542.
IFN-α (1 MU) and 4.5 million IU of IL-2 subcuta- 4. Snow RM, Schellhammer PF: Spontaneous regression of
neously for 6 months. The overall response rate was 28% metastatic renal cell carcinoma. Urology 1982;
and median survival was 20 months. No toxicity greater 20:177–181.
than grade 2 was noted and therefore this appears to be a 5. Tandon PL, Kumar M, Hafeez MA: Metastasis from renal
well-tolerated regimen. Daily thalidomide was also cell carcinoma twenty years after nephrectomy. A case
added to the gemcitabine and 5-FU regimen with no report. Brit J Urol 1963; 35:30.
improvement in response rate. Further, the addition of 6. Oliver RT, Miller RM, Mehta A, et al: A phase 2 study of
thalidomide was associated with an unacceptable rate of surveillance in patients with metastatic renal cell
carcinoma and assessment of response of such patients to
deep venous thrombosis.91 However, none of these trials
therapy on progression. Mol Biother 1988; 1:14–20.
was prospective nor have they employed a control arm or 7. Jemal A, Murray T, Samuels A, et al: Cancer statistics,
immunotherapy alone arm. 2003. CA Cancer J Clin 2003; 53:5–26.
8. Ries LAEM, Kosary CL, et al: SEER Cancer Statistics
Bone Marrow Transplantation Review, 1973–1997. Bethesda, Maryland, National Cancer
Institute, 2000.
Researchers at the National Heart, Lung and Blood insti- 9. Pantuck AJ, Zisman A, Belldegrun AS: The changing
tute expanded their research on graft-versus-leukemia natural history of renal cell carcinoma. J Urol 2001;
reactions to graft-versus-tumor effects. Because mRCC 166:1611–1623.
is partially responsive to immunotherapy, the authors 10. Calle EE, Rodriguez C, Walker-Thurmond K, et al:
postulated that allogeneic lymphocytes from healthy Overweight, obesity, and mortality from cancer in a
prospectively studied cohort of U.S. adults. N Engl J Med
donors would attack metastatic RCC. Nineteen patients
2003; 348:1625–1638.
underwent immunosuppression with cyclophosphamide 11. Flocks R, Kadesky M: Malignant Neoplasms of the
and fludarabine followed by infusion of peripheral blood kidney: analysis of 353 patients followed 5 years or more.
stem cell allograft from an HLA identical or single mis- Trans Am Assoc Genitourin Surg 1957; 49:105-110.
match sibling. Transplantation-related deaths occurred 12. Robson CJ, Churchill BM, Anderson W: The results of
in 2 patients, and 8 patients died of disease progression. radical nephrectomy for renal cell carcinoma. J Urol 1969;
However, 53% experienced disease regression. The three 101:297–301.
266 Part III Kidney and Ureter

13. Beahrs OH: Staging of cancer. CA Cancer J Clin 1991; 30. Motzer RJ, Bacik J, Mariani T, et al: Treatment outcome
41:121–125. and survival associated with metastatic renal cell
14. Guinan P, Sobin LH, Algaba F, et al: TNM staging of carcinoma of non-clear-cell histology. J Clin Oncol 2002;
renal cell carcinoma: workgroup no. 3. Union 20:2376–2381.
International Contre le Cancer (UICC) and the American 31. Fallick ML, McDermott DF, LaRock D, et al:
Joint Committee on Cancer (AJCC). Cancer 1997; Nephrectomy before interleukin-2 therapy for patients
80:992–993. with metastatic renal cell carcinoma. J Urol 1997;
15. Minervini R, Minervini A, Fontana N, et al: Evaluation of 158:1691–1695.
the 1997 tumour, nodes and metastases classification of 32. Citterio G, Bertuzzi A, Tresoldi M, et al: Prognostic
renal cell carcinoma: experience in 172 patients. BJU Int factors for survival in metastatic renal cell carcinoma:
2000; 86:199–202. retrospective analysis from 109 consecutive patients. Eur
16. Gettman MT, Blute ML, Spotts B, et al: Pathologic Urol 1997; 31:286–291.
staging of renal cell carcinoma: significance of tumor 33. Zisman A, Pantuck AJ, Wieder J, et al: Risk group
classification with the 1997 TNM staging system. Cancer assessment and clinical outcome algorithm to predict the
2001; 91:354–361. natural history of patients with surgically resected renal
17. Tsui KH, Shvarts O, Smith RB, et al: Prognostic cell carcinoma. J Clin Oncol 2002; 20:4559–4566.
indicators for renal cell carcinoma: a multivariate analysis 34. Elson PJ, Witte RS, Trump DL: Prognostic factors for
of 643 patients using the revised 1997 TNM staging survival in patients with recurrent or metastatic renal cell
criteria. J Urol 2000; 163:1090–1095. (Quiz 1295.) carcinoma. Cancer Res 1988; 48:7310–7313.
18. Zisman A, Pantuck AJ, Chao D, et al: Reevaluation of the 35. Motzer RJ, Mazumdar M, Bacik J, et al: Survival
1997 TNM classification for renal cell carcinoma: T1 and and prognostic stratification of 670 patients with
T2 cutoff point at 4.5 rather than 7 cm better correlates advanced renal cell carcinoma. J Clin Oncol 1999;
with clinical outcome. J Urol 2001; 166:54–58. 17:2530–2540.
19. Gelb AB, Shibuya RB, Weiss LM, et al: Stage I renal cell 36. Zisman A, Pantuck AJ, Dorey F, et al: Improved
carcinoma. A clinicopathologic study of 82 cases. Am J prognostication of renal cell carcinoma using an
Surg Pathol 1993; 17:275–286. integrated staging system. J Clin Oncol
20. Hafez KS, Fergany AF, Novick AC: Nephron sparing 2001;19:1649–1657.
surgery for localized renal cell carcinoma: impact of 37. Frank I, Blute ML, Cheville JC, et al: An outcome
tumor size on patient survival, tumor recurrence and prediction model for patients with clear cell renal cell
TNM staging. J Urol 1999; 162:1930–1933. carcinoma treated with radical nephrectomy based on
21. Palapattu GSPA, Belldegrun AS, et al: Collecting system tumor stage, size, grade and necrosis: the SSIGN score.
invasion in renal cell carcinoma: impact of prognosis and J Urol 2002; 168:2395–2400.
future staging strategies (in press). 38. Kattan MW, Reuter V, Motzer RJ, et al: A postoperative
22. Uzzo RG, Cherullo EE, Myles J, et al: Renal prognostic nomogram for renal cell carcinoma. J Urol
cell carcinoma invading the urinary collecting 2001; 166:63–67.
system: implications for staging. J Urol 2002; 39. Kim HL, Belldegrun AS, Freitas DG, et al: Paraneoplastic
167:2392–2396. signs and symptoms of renal cell carcinoma: implications
23. Hand JR, Broders AC: Carcinoma of the kidney: the for prognosis. J Urol 2003; 170(6):2221-2224.
degree of malignancy in relation to factors bearing on 40. von der Maase H, Geertsen P, Thatcher N, et al:
prognosis. J Urol 1932; 28:199. Recombinant interleukin-2 in metastatic renal cell
24. Fuhrman SA, Lasky LC, Limas C: Prognostic significance carcinoma—a European multicentre phase II study. Eur J
of morphologic parameters in renal cell carcinoma. Am J Cancer 1991; 27:1583–1589.
Surg Pathol 1982; 6:655–663. 41. Koretz MJ, Lawson DH, York RM, et al: Randomized
25. Chao D, Zisman A, Pantuck AJ, et al: Collecting duct study of interleukin 2 (IL-2) alone vs IL-2 plus
renal cell carcinoma: clinical study of a rare tumor. J Urol lymphokine-activated killer cells for treatment of
2002; 167:71–74. melanoma and renal cell cancer. Arch Surg 1991;
26. Goldstein NS: The current state of renal cell carcinoma 126:898–903.
grading. Union Internationale Contre le Cancer (UICC) 42. Kollender Y, Bickels J, Price WM, et al: Metastatic renal
and the American Joint Committee on Cancer (AJCC). cell carcinoma of bone: indications and technique of
Cancer 1997; 80:977–980. surgical intervention. J Urol 2000; 164:1505–1508.
27. Ro JY, Ayala AG, Sella A, et al: Sarcomatoid renal cell 43. Tanguay S, Swanson DA, Putnam JB Jr: Renal cell
carcinoma: clinicopathologic. A study of 42 cases. Cancer carcinoma metastatic to the lung: potential benefit in the
1987; 59:516–526. combination of biological therapy and surgery. J Urol
28. Zisman A, Chao DH, Pantuck AJ, et al: Unclassified renal 1996; 156:1586–1589.
cell carcinoma: clinical features and prognostic impact of a 44. Slaton JW, Balbay MD, Levy DA, et al: Nephrectomy and
new histological subtype. J Urol 2002; 168:950–955. vena caval thrombectomy in patients with metastatic renal
29. Amin MB, Tamboli P, Javidan J, et al: Prognostic impact cell carcinoma. Urology 1997; 50:673–677.
of histologic subtyping of adult renal epithelial neoplasms: 45. Zisman A, Pantuck AJ, Chao DH, et al: Renal cell
an experience of 405 cases. Am J Surg Pathol 2002; carcinoma with tumor thrombus: is cytoreductive
26:281–291. nephrectomy for advanced disease associated
Chapter 14 Treatment of Advanced Renal Cell Carcinoma 267

with an increased complication rate? J Urol 2002; cell carcinoma: a randomised trial. Lancet 2001;
168:962–967. 358:966–970.
46. Zisman A, Wieder JA, Pantuck AJ, et al: Renal cell 62. Pantuck AJ, Belldegrun AS, Figlin RA: Nephrectomy and
carcinoma with tumor thrombus extension: biology, role interleukin-2 for metastatic renal cell carcinoma. N Engl J
of nephrectomy and response to immunotherapy. J Urol Med 2001; 345:1711–1712.
2003; 169:909–916. 63. Wirth MP: Immunotherapy for metastatic renal cell
47. Skinner DG, Vermillion CD, Colvin RB: The surgical carcinoma. Urol Clin North Am 1993; 20:283–295.
management of renal cell carcinoma. J Urol 192; 64. Motzer RJ, Russo P: Systemic therapy for renal cell
107:705–710. carcinoma. J Urol 2000; 163:408–417.
48. Rafla S: Renal cell carcinoma. Natural history and results 65. Pyrhonen S, Salminen E, Ruutu M, et al: Prospective
of treatment. Cancer 1970; 25:26–40. randomized trial of interferon alfa-2a plus vinblastine
49. Giuliani L: Lymphadenectomy and renal cell versus vinblastine alone in patients with advanced renal
carcinoma: why is there so much controversy? Eur Urol cell cancer. J Clin Oncol 1999; 17:2859–2867.
1983; 9:374. 66. Medical Research Council Renal Cancer Collaborators:
50. Blom JH, van Poppel H, Marechal JM, et al: Radical Interferon-alpha and survival in metastatic renal
nephrectomy with and without lymph node dissection: carcinoma: early results of a randomised controlled trial.
preliminary results of the EORTC randomized phase III Lancet 1999; 353:14–17.
protocol 30881. EORTC Genitourinary Group. Eur Urol 67. Negrier S, Escudier B, Lasset C, et al: Recombinant
1999; 36:570–575. human interleukin-2, recombinant human interferon alfa-
51. Vasselli JR, Yang JC, Linehan WM, et al: Lack of 2a, or both in metastatic renal-cell carcinoma. Groupe
retroperitoneal lymphadenopathy predicts survival of Francais d’Immunotherapie. N Engl J Med 1998;
patients with metastatic renal cell carcinoma. J Urol 2001; 338:1272–1278.
166:68–72. 68. Motzer RJ, Murphy BA, Bacik J, et al: Phase III trial of
52. Pantuck AJ, Zisma A, Dorey F, et al: Renal cell carcinoma interferon alfa-2a with or without 13-cis-retinoic acid for
with retroperitoneal lymph nodes: role of lymph node patients with advanced renal cell carcinoma. J Clin Oncol
dissection. J Urol 2003; 169. 2000; 18:2972—2980.
53. Pantuck AJ, Zismon A, Dorey F: Renal cell carcinoma 69. Giorgio PPL, Boracchi P: Adjuvant interferon to radical
with retroperitoneal lymph nodes: impact on survival nephrectomy in Robson’s stage II and III renal cell
and benefits of immunotherapy. Cancer 2003; 97(12): carcinoma: contradictory results in pN0 and pN1 patients.
2995-3002. Proc ASCO, 1999.
54. deKernion JB, Sarna G, Figlin R, et al: The treatment of 70. Fisher RI, Rosenberg SA, Fyfe G: Long-term survival
renal cell carcinoma with human leukocyte alpha- update for high-dose recombinant interleukin-2 in
interferon. J Urol 1983; 130:1063–1066. patients with renal cell carcinoma. Cancer J Sci Am 2000;
55. Haas GP, Hillman GG, Redman BG, et al: 6(Suppl 1):S55–S57).
Immunotherapy of renal cell carcinoma. CA Cancer 71. Belldegrun A, Webb DE, Austin HA 3rd, et al: Effects of
J Clin 1993; 43:177–187. interleukin-2 on renal function in patients receiving
56. Wagner JR, Walther MM, Linehan WM, et al: immunotherapy for advanced cancer. Ann Intern Med
Interleukin-2 based immunotherapy for metastatic renal 1987; 106:817–822.
cell carcinoma with the kidney in place. J Urol 1999; 72. Yang JC, Rosenberg SA: An ongoing prospective
162:43–45. randomized comparison of interleukin-2 regimens for the
57. Walther MM, Yang JC, Pass HI, et al: Cytoreductive treatment of metastatic renal cell cancer. Cancer J Sci Am
surgery before high dose interleukin-2 based therapy in 1997; 3(Suppl 1):S79–S84.
patients with metastatic renal cell carcinoma. J Urol 1997; 73. Milowsky MI, Nanus DM: Advanced renal cell carcinoma.
158:1675–1678. Curr Treat Options Oncol 2001; 2:437–445.
58. Levy DA, Swanson DA, Slaton JW, et al: Timely delivery 74. Yang JC, Topalian SL, Parkinson D, et al: Randomized
of biological therapy after cytoreductive nephrectomy in comparison of high-dose and low-dose intravenous
carefully selected patients with metastatic renal cell interleukin-2 for the therapy of metastatic renal cell
carcinoma. J Urol 1998; 159:1168–1173. carcinoma: an interim report. J Clin Oncol 1994;
59. Walther MM, Lyne JC, Libutti SK, et al: Laparoscopic 12:1572–1576.
cytoreductive nephrectomy as preparation for 75. Dutcher JP, Fisher RI, Weiss G, et al: Outpatient
administration of systemic interleukin-2 in the treatment subcutaneous interleukin-2 and interferon-alpha for
of metastatic renal cell carcinoma: a pilot study. Urology metastatic renal cell cancer: five-year follow-up of the
1999; 53:496–501. Cytokine working group study. Cancer J Sci Am 1997;
60. Flanigan RC, Salmon SE, Blumenstein BA, et al: 3:157–162.
Nephrectomy followed by interferon alfa-2b compared 76. Yron I, Wood TA Jr, Spiess PJ, et al: In vitro growth of
with interferon alfa-2b alone for metastatic renal-cell murine T cells. V. The isolation and growth of lymphoid
cancer. N Engl J Med 2001; 345:1655–1659. cells infiltrating syngeneic solid tumors. J Immunol 1980;
61. Mickisch GH, Garin A, van Poppel H, et al: Radical 125:238–245.
nephrectomy plus interferon-alfa-based immunotherapy 77. Belldegrun A, Muul LM, Rosenberg SA: Interleukin 2
compared with interferon alfa alone in metastatic renal expanded tumor-infiltrating lymphocytes in human renal
268 Part III Kidney and Ureter

cell cancer: isolation, characterization, and antitumor lysate-pulsed dendritic cells can expand specific T cells
activity. Cancer Res 1988; 48:206–214. and mediate tumor regression. Cancer Res 2001;
78. Lafreniere R, Rosenberg SA: Adoptive immunotherapy of 61:8513–8519.
murine hepatic metastases with lymphokine activated 86. Amato RJML, Wood L, et al: Active specific
killer (LAK) cells and recombinant interleukin 2 (RIL 2) immunotherapy in patients with renal cell carcinoma
can mediate the regression of both immunogenic and using autologous tumor derived heat shock protein-
nonimmunogenic sarcomas and an adenocarcinoma. peptide complex-96 vaccine, Proc ASCO, 1999.
J Immunol 1985; 135:4273–4280. 87. Yang JCHL, Steinberg SM, Rosenberg SA, Novotny WA:
79. Figlin RA, Pierce WC, Kaboo R, et al: Treatment of A randomized double-blind placebo-controlled trial of
metastatic renal cell carcinoma with nephrectomy, bevacizumab (anti-VEGF antibody) demonstrating a
interleukin-2 and cytokine-primed or CD8(+) selected prolongation in time to progression in patient with
tumor infiltrating lymphocytes from primary tumor. metastatic renal cancer. Proc Am Soc Clin Oncol 2002;
J Urol 1997; 158:740–745. Abstract 15.
80. Figlin RA, Thompson JA, Bukowski RM, et al: 88. Mani S, Vogelzang NJ, Bertucci D, et al: Phase I study to
Multicenter, randomized, phase III trial of CD8(+) tumor- evaluate multiple regimens of intravenous 5-fluorouracil
infiltrating lymphocytes in combination with recombinant administered in combination with weekly gemcitabine in
interleukin-2 in metastatic renal cell carcinoma. J Clin patients with advanced solid tumors: a potential broadly
Oncol 1999; 17:2521–2529. active regimen for advanced solid tumor malignancies.
81. Neumann E, Engelsberg A, Decker J, et al: Cancer 2001; 92:1567–1576.
Heterogeneous expression of the tumor-associated 89. Rini BI, Vogelzang NJ, Dumas MC, et al: Phase II trial
antigens RAGE-1, PRAME, and glycoprotein 75 in of weekly intravenous gemcitabine with continuous
human renal cell carcinoma: candidates for T-cell-based infusion fluorouracil in patients with metastatic renal
immunotherapies? Cancer Res 1998; 58:4090–4095. cell cancer. J Clin Oncol 2000; 18:2419–2426.
82. Bui MH, Seligson D, Han KR, et al: Carbonic anhydrase 90. Neri B, Doni L, Gemelli MT, et al: Phase II trial
IX is an independent predictor of survival in advanced of weekly intravenous gemcitabine administration
renal clear cell carcinoma: implications for prognosis and with interferon and interleukin-2 immunotherapy
therapy. Clin Cancer Res 2003; 9:802–811. for metastatic renal cell cancer. J Urol 2002;
83. Oosterwijk E, Ruiter DJ, Hoedemaeker PJ, et al: 168:956–958.
Monoclonal antibody G 250 recognizes a determinant 91. Desai AA, Vogelzang NJ, Rini BI, et al: A high rate of
present in renal cell carcinoma and absent from normal venous thromboembolism in a multi-institutional phase II
kidney. Int J Cancer 1986; 38:489–494. trial of weekly intravenous gemcitabine with continuous
84. Mulders P, Tso CL, Gitlitz B, et al: Presentation of renal infusion fluorouracil and daily thalidomide in patients
tumor antigens by human dendritic cells activates tumor- with metastatic renal cell carcinoma. Cancer 2002;
infiltrating lymphocytes against autologous tumor: 95:1629–1636.
implications for live kidney cancer vaccines. Clin Cancer 92. Childs R, Chernoff A, Contentin N, et al: Regression of
Res 1999; 5:445–454. metastatic renal cell carcinoma after nonmyeloablative
85. Geiger JD, Hutchinson RJ, Hohenkirk LF, et al: allogeneic peripheral-blood stem-cell transplantation.
Vaccination of pediatric solid tumor patients with tumor N Engl J Med 2000; 343:750–758
C H A P T E R

15 Transitional Cell Carcinoma of the


Renal Pelvis and Ureter: Evaluation
and Treatment
Badrinath R. Konety, MD, MBA,
and Richard D. Williams, MD

Transitional cell carcinoma (TCC) of the upper urinary 17 to 170 months.17,18 Based on karyotypic similarities
tract occurs less frequently than bladder cancer. The pre- between bladder and upper tract TCC, it is likely that
cise incidence of renal pelvic and ureteral cancers is diffi- similar risk factors affect both upper and lower tract
cult to discern since they are grouped with other kidney tumors.19 The risk factors associated with increased inci-
cancers. Ureteral cancers are less common than renal dence of upper tract TCC include recurrent bladder car-
pelvic cancers. Renal pelvic tumors comprise 5% to 7% cinoma in situ (CIS) after intravesical BCG therapy,20,21
of all renal tumors.1–5 Worldwide statistics vary and are and multifocal CIS in the bladder at the time of cystec-
not accurate, since renal pelvis tumors are not reported tomy.22–24 Up to 19% of patients with upper urinary tract
separately. The highest incidence is found in Balkan TCC have been reported to initially present with
countries (Bulgaria, Greece, Romania, and Yugoslavia), metastatic disease.25
where urothelial cancers account for 40% of all renal
cancers. The majority of renal pelvic and ureteral cancers
ETIOLOGY
are transitional cell in origin with squamous cell carci-
noma (SCC) and adenocarcinoma forming a small Workers in chemical, petrochemical, and plastic indus-
minority.2,6,7 Ureteral tumors most frequently involve tries, aniline dye workers, and those exposed to coal,
the lower-third of the ureter.7–10 Bilateral involvement or coke, tar, and asphalt are at increased risk for renal pelvis
metachronous renal pelvic tumor development in the and ureteral TCC (relative risk = 4 to 5.5). Aniline dyes,
contralateral kidney occurs in about 2% of patients.11,12 β-naphthylamine and benzidine compounds have all
The estimated incidence of TCC arising in the ureter been implicated as causative agents for upper tract
and other portions of the urinary tract other than renal TCC.26,27 TCC in various locations in the urinary tract
pelvis or bladder in the United States is 2450 cases for can develop up to 18 years after exposure.27 However,
the year 2004, with 690 deaths occurring as a result of most of these chemicals are more commonly associated
these tumors.3 TCC of any origin is uniformly fatal with lower urinary tract tumors because it is believed that
unless treated since it is almost never discovered inciden- prolonged contact with parent compounds or metabo-
tally at autopsy.13 Patients with Balkan nephropathy have lites in urine is required for induction of carcinogenesis.
a higher incidence of upper tract TCC and there is also Cigarette smoking is a major risk factor for upper tract
an increased incidence of bilaterality in 10% of these TCC. Cessation of smoking somewhat decreases the risk
patients but the tumors tend to be low grade.2,8,11,12,14 but does not eliminate it completely. Duration of smok-
Patients with upper tract TCC are at risk of developing ing is the strongest predictor of risk.28 Balkan endemic
bladder TCC, with an estimated incidence of 15% to nephropathy is a chronic tubulointerstitial disorder con-
60% within 5 years following the upper tract TCC.15,16 fined to the Balkan countries, which ultimately results in
Patients with primary bladder TCC will develop upper renal failure. Cancer of the renal pelvis is 100 to 200
tract TCC in 2% to 4% of cases, with a mean interval of times more frequent in these countries than in control

269
270 Part III Kidney and Ureter

regions.29 Analgesic abuse is another predisposing factor yield more precise anatomic information regarding the
for the development of upper tract TCC. One study upper tract urothelium, it is laborious to perform and not
found the relative risk for abusers of phenacetin contain- all centers have the same level of expertise in performing
ing analgesics was 2.4 for men and 4.2 for women.29 The and interpreting these studies. A well-performed IVP
dose–effect relationship was noted for both phenacetin will reveal an abnormality in the majority of cases (Figure
and aspirin. The latency period was 24 to 26 years. While 15-1A,B). The finding of a non-functional kidney on
phenacetin is no longer available in the United States, IVP, which occurs in up to 50% of cases,8,37 can portend
acetaminophen has similar metabolites to phenacetin and a worse prognosis.39 Classic signs for upper tract urothe-
is widely used as an antipyretic. Case control studies have lial carcinoma on IVP include the “goblet” or
not revealed any significant association between routine “Bergmann’s sign,” which is a meniscus-shaped filling
acetaminophen use and bladder TCC or upper tract defect in the ureter. A “stipple” sign may be observed
TCC.30 However, abuse of the drug has not been stud- with contrast being trapped in the fronds of a papillary
ied. Both Balkan nephropathy and phenacetin abuse are tumor, either in the renal pelvis or ureter. Retrograde
associated only with renal pelvis TCC. Renal papillary pyelograms will demonstrate evidence of tumor in the
necrosis itself is not a necessary factor for the develop- form of filling defects or an intrinsic narrowing or stric-
ment of renal pelvis TCC, but the combination of ture of the ureter in almost all cases (Figure 15-2).39
phenacetin use and papillary necrosis causes a 20-fold Stricture or stenosis more commonly occurs at the
increased risk for renal urothelial tumors, indicating that ureteropelvic junction or ureter and can indicate infiltra-
both are probably independent risk factors. Chronic bac- tive disease.8 In patients who have undergone a cystec-
terial infection with urinary calculi and obstruction may tomy for prior bladder TCC, a loopogram or
predispose to the development of urothelial cancer. SCC pouchogram obtained by instilling contrast into the ileal
is the most common entity in these cases. Schistosomiasis conduit or neobladder may be required to adequately
may also predispose to SCC. Cyclophosphamide has also visualize the upper tracts. Conventional upper tract
been implicated in the development of renal pelvis TCC. imaging can yield false positive results in 36% to 60% of
About 6 cases have been reported occurring after an cases.40,41 An alternative to the IVP is a contrast-
average of 5 years following chemotherapy.31 Thorium enhanced CT scan, which is often required to optimally
dioxide (Thorotrast), which was used as a contrast agent evaluate the renal parenchyma. Presence of a mass lesion
for retrograde pyelograms about 70 years ago, is also protruding into the contrast-filled collecting system can
known to be a causative factor since it supposedly causes usually be identified on the CT scan especially in cases of
vascular insufficiency and a foreign body response. papillary tumors >1 cm in size (Figure 15-3). CT scan-
Cancers tend to occur after a latency period of 20 years.32 ning is easier to perform and less labor-intensive, with
greater standardization of technique, and has the specific
advantage of distinguishing intrarenal stones from
DIAGNOSIS
tumors if films are obtained before and after intravenous
Upper tract TCC can present with ureteral obstruction contrast administration. The curvilinear pattern of calci-
or hematuria (gross or microscopic). It can be asympto- fication occasionally observed in upper tract TCC is usu-
matic and discovered during upper tract surveillance ally distinguishable by CT scan. A single shot (IVP) in
after development of bladder TCC. An increased index the form of a plain abdominal X-ray immediately follow-
of suspicion, especially in patients who have undergone ing the CT scan can capture an image of the contrast
BCG or other intravesical therapy for bladder TCC, is filled renal pelvis and ureters. More recently, CT uro-
prudent since the incidence of upper tract cancer in these grams have been performed in order to obtain a three-
patients can be as high as 38%.33 These tumors can occur dimensional view of the calyces and ureters. They appear
up to 15 years following successful treatment of bladder to be equivalent to an IVP in imaging the ureters.42
TCC, which mandates at least yearly surveillance of the Thinly collimated axial images obtained during a single
upper tracts in these patients.34 Hematuria is the most breath hold using a multirow detector CT scan are then
common presenting symptom, occurring in 56% to 98% reformatted to obtain a coronal image of the collecting
of patients.8,35,36 Flank pain is usually a sign of upper tract system. The images are of higher resolution than those
obstruction. Systemic symptoms, such as weight loss, obtained using standard single detector helical CT scan-
anorexia, and lethargy, are late findings that can occur in ning. CT urography can detect the majority of ureteric
10% to 15% of patients.36–38 tumors with a sensitivity as high as 94%.43 CT urogra-
phy, however, does expose the patient to a greater dose of
radiation. Sensitivity for detecting upper tract malig-
Radiographic Evaluation
nancy can approach 100% with a specificity of 60%. The
The traditional evaluation of the upper tract has included negative predictive value was 100% in one reported
an intravenous pyelogram (IVP). While the IVP may study.43 However, more experience with this technique is
Chapter 15 Transitional Cell Carcinoma of the Renal Pelvis and Ureter 271

Figure 15-1 IVP with tomographic and excretory phase images of a patient with invasive
TCC of the right upper pole who presented with gross hematuria and pyelonephritis.
272 Part III Kidney and Ureter

needed before it can be widely employed. MR urography


has also been used in a similar manner with slightly lower
reported sensitivity of 80%.44 CT scanning or MRI can
also be used to assess local extent of tumor and presence
of metastatic disease.

Cytology and Other Markers


Urine cytology is a very specific method that can be used
to diagnose TCC of the upper and lower urinary tracts
(Figure 15-4A,B). It is very sensitive for high-grade papil-
lary tumors and CIS but less so for low-grade disease7,45
and is also operator dependent. Positive upper tract cytolo-
gies in the absence of bladder disease identify the presence
of upper tract TCC. Often retrograde pyelography and
ureteroscopy fail to reveal a lesion in the upper tract even
in the presence of frankly positive urine cytology. Upper
tract or urine cytologies that do not revert to negative
6 months following a completely negative urologic workup
may indicate indolent or latent disease that will be discov-
ered subsequently.46 The value of obtaining upper tract
washing for cytology in the presence of bladder TCC is
questionable. Contamination from tumor cells in the blad-
der make it very difficult to accurately interpret these
results, especially in the absence of a radiologic abnormal-
ity in the upper tracts.47 However, properly performed
studies with simultaneous specimens being obtained from Figure 15-2 Retrograde pyelogram obtained in the patient
from Figure 15-1 demonstrating a filling defect in the right
the bladder and upper tracts with the use of normal saline
upper pole due to an infiltrative TCC.
to keep the ureteral catheter clear of bladder urine until it
has been placed in the ureter can be successful.
Several urine-based markers have been studied to
increase the accuracy of diagnosing upper tract cancer.
Urinary levels of NMP22, a nuclear matrix protein-based
Direct Endoscopic Evaluation
marker, are found to be elevated in patients with upper
tract cancer.48 However, the numbers of patients in these Endoscopic evaluation, including retrograde pyelograms
studies are small and the specificity is low, yet the sensi- and brush biopsies, are usually performed to obtain a
tivity is higher than cytology for low-grade disease. The more definitive diagnosis following the detection of a
BTA stat test (Mentor Corp., Santa Barbara, CA) has a positive upper tract cytology. Brush biopsy through an
sensitivity of 82% and specificity of 89% compared to open-ended ureteral catheter was originally described by
voided urine cytology (11% and 54%, respectively) or Gill et al.52 Brush biopsy has a sensitivity of up to 90%
ureteral wash cytology (48% and 33%, respectively) for and a specificity of up to 88%.7,53,54 Ureteroscopic visu-
upper tract tumors.49 In one study, telomerase was alization using a flexible or semirigid ureteroscope and
detected using cells obtained from upper tract wash- direct biopsy is also effective in diagnosing these tumors,
ings and measured using the telomeric repeat amplifica- with a sensitivity of 86% and specificity of 90%.53
tion protocol (TRAP) assay in 14 of 17 patients.50 In Complications of brush biopsy or ureteroscopy include
comparison, voided urine cytology detected only 2 of the risk of infection, ureteral or renal pelvic perforation,
13 cases and ureteral washing cytology detected only 8 of bleeding, mucosal flap injury, and mucosal edema with
15 cases.50 Studies conducted thus far have included small resultant ureteral obstruction.
numbers of patients and more data are required from
larger studies before any of these tests can be routinely
HISTOPATHOLOGY
recommended. Other markers, such as p27 (cyclin-
dependent kinase inhibitor) expression in tumor tissue, TCC is the most common histologic type of upper uri-
can also help to predict prognosis of upper tract tumors. nary tract cancer. Cancers may be papillary or sessile and
Low levels of p27 staining in tumors can be indicative of multifocality is common. CIS can be particularly difficult
worse disease-free survival in these patients.51 to identify grossly and may vary from a whitish-appearing
Chapter 15 Transitional Cell Carcinoma of the Renal Pelvis and Ureter 273

Figure 15-3 Contrast enhanced CT scan of a patient with a large papillary Ta TCC (arrow)
arising from the left renal pelvis, which led to spontaneous bleeding and a perinephric
hematoma (arrow).

Figure 15-4 A, Urine cytology, low-grade papillary TCC. (Modified from Kleer E, Osterling
JE: Prob Urol 1992; 6(3):531, with permission.) B, Urine cytology, high-grade papillary TCC.
(Modified from Kleer E, Osterling JE: Prob Urol 1992; 6(3):531, with permission.)
274 Part III Kidney and Ureter

plaque, due to epithelial hyperplasia, to a velvety red Table 15-1 Staging Systems for Upper Tract
patch from increased submucosal vascularity.2 Progression Urothelial Tumors
to invasive TCC can occur from a papillary, low-grade TNM staging system
lesion or by transformation of CIS.2,55 Multiple tumors Primary tumor (T)
can occur by implantation of tumor cells onto mechani- Tx Primary tumor cannot be assessed
cally disrupted mucosa or due to the “field effect” on the T0 No evidence of primary tumor
entire urothelium, which is believed to contribute to the Ta Papillary noninvasive carcinoma
multifocal nature of TCC. CIS or atypia of the lower Tis Carcinoma in situ
ureters is found in 7% to 25% of patients undergoing cys- T1 Tumor invades subepithelial connective tissue
tectomy for bladder cancer.56 Presence of upper tract CIS T2 Tumor invades the muscularis
is also more common in heavily pretreated patients who T3 (For renal pelvis only) Tumor invades beyond
muscularis into peripelvic fat or the renal
have failed multiple courses of intravesical agents, such as
parenchyma
BCG.57 These tumors can occur long after treatment of T4 Tumor invades adjacent organs, or through
bladder disease, which suggests that these patients will the kidney into the perinephric fat
benefit from long-term follow-up. Many of these instances
of upper tract CIS may be asymptomatic and diagnosed on Lymph nodes (N)
work-up of positive urine or selective ureteral cytology. Nx Regional lymph nodes cannot be assessed
N0 No regional lymph node metastasis
N1 Metastasis in a single lymph node, 2 cm or
STAGING AND GRADING less in greatest dimension
The most commonly used staging system for upper tract N2 Metastasis in a single lymph node, more than
cancer is the TNM staging system (Table 15-1). Upper 2 cm but not more than 5 cm in greatest
dimension; or multiple lymph nodes, none
tract TCC spreads by direct invasion, mucosal seeding,
more than 5 cm in greatest dimension
hematologic and lymphatic routes. The most common N3 Metastasis in a lymph node, more than 5 cm
sites of metastases are lung, bone, and liver. The likeli- in greatest dimension
hood of nodal metastases increases with increasing stage,
with 48% of T3 tumors and 78% of T4 tumors present- Distant metastasis (M)
ing with nodal metastases.58 Mx Distant metastasis cannot be assessed
The traditional grading system for bladder TCC is M0 No distant metastasis
also applicable to TCC of the upper urinary tract. M1 Distant metastasis
Broder’s original grading system, as modified by Ash,
grades tumors on a scale of 1 to 4, with grade 1 tumors AJCC staging system in comparison to TNM
being mainly papillomas and grade 4 tumors being AJCC stage TNM staging
Stage 0 T0
highly anaplastic and poorly differentiated.2,59 The
Stage I Ta, Tis, T1, N0, M0
World Health Organization’s grading system, proposed Stage II T2, N0, M0
by Mostofi, eliminates papillomas and grades tumors Stage III T3, N0, M0
from grade 1 to grade 3. The most recent grading sys- Stage IV T4 or any T, N+, M+
tem now divides tumors into low grade or high grade.60
Papillomas and papillary urothelial neoplasms of low
malignant potential (PUNLMP) are separated out as
CONSERVATIVE SURGICAL MANAGEMENT
well. Low-grade tumors include those previously con-
Endoscopic Management
sidered grade 1 and grade 1-2, while high-grade tumors
are those considered grade 2, grade 2-3, and grade 3. The established standard of care for the management of
Prognosis of upper tract tumors is clearly dependent upper tract TCC, regardless of stage or grade, is a radical
on tumor stage. There is a progressive decrease in sur- nephroureterectomy with resection of a generous cuff of
vival with increasing stage with the most significant drop bladder. The first radical nephroureterectomy was
being observed in stage T3 tumors, which involve perire- reported by LeDantu and Albarran.64 This approach is
nal or periureteral fat.61 When matched stage for stage, based on the characteristic biologic behavior of these
there does not appear to be a significant difference in sur- tumors. They tend to be multifocal, contralateral tumors
vival between renal pelvis and ureteral tumors.62 Other are uncommon (<5%), and there is a high incidence of
factors correlated with survival in these patients include ipsilateral tumor recurrence after partial resection.65
tumor grade, multifocality, concomitant bladder carci- Furthermore, prior to the advent of the smaller caliber and
noma, and ploidy.56 Tumor ploidy may be predictive of flexible endoscopes, conservative endoscopic management
survival with aneuploid tumors, demonstrating 5- and of these tumors was extremely difficult and fraught with
10-year survivals of 25% and 0%, respectively.63 errors in diagnosis and complications, rendering it obvi-
Chapter 15 Transitional Cell Carcinoma of the Renal Pelvis and Ureter 275

ously unpopular. Vest first reported the successful man- invasive (T2-T3). Correlation between grade and stage
agement of papillary ureteric tumors with local resection determined on ureteroscopic biopsy samples and final
alone in 1945.66 Over the past two decades a number of pathologic grade/stage determined from the
authors have reported successful long-term endoscopic nephroureterectomy specimen tend to be more accurate
management of upper tract TCC. There are certain basic in the case of proximal ureteral tumors than distal
tenets that must be observed while adopting this approach. ureteral tumors.69 Following adequate biopsy sampling,
The patient must agree to submit to long-term close sur- the tumors are ablated using either a Nd:Yag laser or
veillance with repeated upper tract endoscopy, tumors Holmium laser delivered through a 200 or 365 μm fiber.
should be of low grade and stage and the patient must The holmium laser is used at a setting of 1 J and 10 Hz
understand that nephroureterectomy may still become while the Nd:Yag is used at a 30 W setting.72 The holmium
necessary if there is evidence of stage or grade progression. laser has a penetration depth of 0.5 mm while the Nd:Yag
Upper tract tumors can be visualized using ureteral can penetrate 5 to 6 mm below the surface, which can
contrast studies and ureteroscopy. Biopsies can be result in ureteral wall damage and may go unnoticed until
obtained using a 2.4Fr flat wire basket or a 3Fr flexible later.73 Some experienced endoscopists advocate using a
cup biopsy forceps, which can be placed through a flexi- combination of the two types of laser to achieve a more
ble 7Fr ureteroscope or a 6.9Fr semirigid ureteroscope. complete ablation of all of the tumors. A ureteral stent is
This approach can successfully confirm the presence of left in place for 1 to 2 weeks following the procedure. In
upper tract TCC in up to 98% of cases.67 In their series situations where postablation instillation of a chemother-
of 51 patients, Keeley et al.68 found that ureteroscopic apeutic agent, such as mitomycin is anticipated, an open-
biopsy yielded a diagnosis of cancer in 94.1% of cases and ended ureteral catheter can be left in place attached to a
histologic grading was possible in 82.4%. The 11.5Fr Foley catheter to facilitate upper tract access.
resection loop can also be used through the ureteroscope, Complications reported with this approach have been
particularly in sampling and/or treating distal ureteric minimal. Ureteral perforation, ureteral strictures and
tumors.69 Upper tract tumors are akin to bladder tumors incomplete tumor resection have all been reported. The
in terms of the correlation between pathologic grade and incidences of ureteral perforation in the two largest
stage. Low-grade tumors predominantly tend to be low recent series of endoscopic management of upper tract
stage and very few high-grade tumors are of low stage tumors have been 0%68 and 5.4%.74 Ureteral strictures
(Ta-T1).38,70 Previous studies indicate that almost all tend to occur more frequently in patients who suffer a
grade 1 tumors are Ta.70 Keeley et al.71 found that even ureteral perforation, with an overall incidence of 8.6%
low- to moderate-grade tumors tend to be of lower stage (Table 15-2). This is higher than that observed with
(Ta-T1) while 67% of high-grade tumors were muscle nononcologic ureteroscopy and laser use. One potential

Table 15-2 Results of Endoscopic Ablation of Upper Urinary Tract Tumors


Subsequent
No. of Complication Recurrence Nephrouret- Follow-Up
Author Patients Rate (Site) erectomy (Months)

Englemeyer and Belig75 10 2/10 (20%) 2 (P), 5 (U) — 24–66

Elliott et al.74 37 8/37 (22%) 8 (P), 9 (U), 19 (B) 6 3–132

Martinez-Pinero et al.76 28 12/28 (43%) 2 (P), 6 (U) 3 2–119

Keeley et al.68 41 2/41 (5%) 3 (P), 5 (U), 15 (B) 8 3–116

Chen et al.67 23 2/23 (9%) 5 (P), 8 (U), 7 (B) 4 8–103

Blute et al.53 13 (ureteral) 0% 2 (U); 4 (B) 0 6–50

8 (renal pelvis) 0% 1 (P) 0 12–48

Johnson77 3 33% 1 (U), 2 (B) 0 5–22

Schilling et al.78 10 0% 1 (B) 0 3–31

Modified from Chen GL, Bagley DH: J Endourol 2001; 15:399.


P, renal pelvis; U, ureter; B, bladder.
276 Part III Kidney and Ureter

reason for this may be that in the case of tumor ablation performing adjuvant radiation after nephroureterectomy
the laser energy is being directed against the urothelium, in this situation are also viable options.87 The percuta-
directly permitting a greater amount of damage to occur, neous nephrostomy can also be utilized for instillation of
while in the case of laser lithotripsy, the beam is directed chemotherapeutic/immunotherapeutic agents following
away from the urothelium. TCC recurrence rates after the resection.88,89 Percutaneous resection is effective for
endoscopic ablation have been comparable to those low-grade tumors but the outcome of local resection for
obtained with open resection of renal pelvis TCC (50% high-grade disease is dismal with 80% of patients dying of
versus 45% to 65%, respectively).8 Overall, the recur- their disease in spite of subsequent nephroureterectomy in
rence rate of tumors in the renal pelvis is 37% and of one series.90 Multiple resections may be required to clear
those in the ureter is 43% after endoscopic management the renal pelvis of disease. Seeding of the nephrostomy
(see Table 15-2). Recurrences appear to be grade related, tract does not appear to be a significant occurrence as evi-
with 76% of kidneys with grade 1 tumors being cleared denced by absence of tumor in the tract resected at subse-
of all disease at one point, 64% of kidneys with grade 2 quent nephroureterectomy.90 Renal salvage is possible in
tumors being cleared of disease and only 40% of kidneys 60% of patients with low-grade disease at a follow-up of
bearing grade 3 tumors being cleared of disease.68 64 months.90
However, in this same report, local recurrences occurred In patients who have biopsy-proven CIS or positive
in 25% of those with grade 1 tumors and 44% of those upper tract cytologies, instillation of BCG into the upper
with grade 2 tumors requiring repeat endoscopic treat- tract can be an effective therapy. This approach could also
ment at mean follow-up times of 40.3 months and 27.6 be used, in solitary kidneys or in poor surgical candidates for
months, respectively.68 Tumors smaller than 1.5 cm can nephroureterectomy, as adjuvant therapy following either
also be cleared more effectively compared to larger percutaneous or retrograde resection of renal pelvis
tumors as evidenced by the difference in recurrence tumors. Instillation can be performed either antegrade via a
rates—25% versus 50%.68 percutaneous nephrostomy tube or retrograde through a
An alternative endoscopic approach in patients with 4Fr or 5Fr ureteral catheter inserted temporarily. The BCG
bulky renal pelvis tumors is percutaneous resection or is instilled as a continuous infusion over 30 to 60 minutes (1
ablation. While this approach has demonstrated equiva- ml/minute) with the bag hanging 20 cm above the
lence with more traditional surgical approaches in terms of patient.88,89 The dose of BCG used is usually up to three
cancer control, case selection is critical.79 Percutaneous times the intravesical dose91 and instillation is avoided in the
access is obtained through the upper or midpole calyx to presence of gross hematuria. Patients void following instil-
gain easy access to the collecting system as well. lation and the treatment is repeated weekly for 6 weeks. In
Alternatively, the involved calyx can also be accessed and an analysis of data from at least eight previous studies, it
the tract dilated and a working sheath placed. The tumor appears that an initial response rate of 79% can be achieved
is debulked using cold cup forceps and the base is resected with a follow-up ranging from 4 to 82 months. The
using a standard resection loop. The base can also be ful- response can be durable in several of these patients as
gurated with electrocautery or a laser. A second look observed by Okubo et al.,89 who obtained a long-term
nephroscopy is typically performed 3 to 7 days later and response over a median follow-up of 49 months in 6 of 9
the resection base biopsied again to rule out the presence (66%) of their initial responders. Other series have found a
of residual tumor.80 Disease-specific survival at a mean fol- long-term response rate with renal preservation of around
low-up of 51 months using this approach is reported to be 50% for patients with CIS.91 Fever appears to be a common
100% for grade 1, 94% for grade 2, and 62% for grade 3 side effect in these patients and septicemia due to bacterial
TCC.80 The procedure is invasive with increased risk of dissemination or systemic BCGosis can occur in these
hemorrhage, extravasation, and possible tumor dissemina- patients and could be lethal. In one series, the incidence of
tion along the nephrostomy tract or into the retroperi- sepsis was 2 of 37 patients (5%).91 This mandates emergent
toneum. The incidence of percutaneous tract seeding after evaluation with resuscitation, starting of triple antitubercu-
nephrostomy and upper tract TCC resection is low. Most lous drug therapy with isoniazid (plus pyridoxine), rifampin,
reports involve recurrences that have occurred following and ethambutol. Steroids may need to be used in addition if
percutaneous access after nephroscopy81 or prolonged the patient is not responding to this regimen in the acute
nephrostomy tube drainage.82–84 Very few reported cases setting. Mitomycin instillation can also be used as initial
in the literature have occurred after percutaneous resec- treatment in place of BCG with similar results.92
tion of an upper tract TCC.85,86 Suggested methods to
prevent nephrostomy tract or local recurrence after percu-
Surgical Approaches
taneous access include maintaining low intrarenal pres-
Partial Ureterectomy
sures, using a 30Fr working sheath, use of sterile water,
and keeping the irrigating solution at a height <40 cm Segmental ureteral resection is a viable curative option
above the patient.86 Resecting the nephrostomy tract and for tumors of the ureter. Judicious use of this option is
Chapter 15 Transitional Cell Carcinoma of the Renal Pelvis and Ureter 277

important, keeping in context the stage and grade of Recurrent TCC are also more likely to be of higher
tumor. It is most applicable to low-grade, distal ureteral grade and stage when they are located in the renal
tumors. The 5-year survival for patients with grade pelvis.8,104 Renal pelvis TCC also appear to have a higher
1 tumors, who have undergone partial ureterectomy, is likelihood of developing systemic disease subsequently
100%.93 The ipsilateral recurrence rate following seg- (0%) as compared to ureteral TCC (19%). Hence, the
mental resection is low and the recurrent tumors also more traditional nephroureterectomy is a better
tend to be low grade94 (Table 15-3). Recurrent tumors approach for renal pelvis or proximal ureteral TCC espe-
usually tend to occur distal to the original tumor, though cially if they are of high grade or stage.
occasional proximal recurrences have been noted.95,99
Potential explanations for this include the presence of
Open Nephroureterectomy
adjacent CIS that is undetected at the time of primary
resection, seeding of the upper tract due to vesico- One of the major drawbacks of the open nephroureterec-
ureteric reflux, or de novo occurrence of new tumors. tomy is the need for two separate incisions to enable
Patients with grade 2, stage Ta or T1 tumors can also be complete excision of the distal ureter with a cuff of blad-
managed by segmental resection of the ureter containing der mucosa. Typically, this is achieved by using a flank
the tumor. Anderstrom et al.39 found a 100% survival incision for the nephrectomy and an additional Gibson
rate in 16 patients managed in this manner at 83 months incision to access the distal ureter and bladder. It should
follow-up. The success of conservative resection for also be noted that prior to proceeding with an open
muscle invasive or high-grade disease is not as clear-cut. nephroureterectomy, it is essential to ensure that the
Leitenberger et al.100 found that 4 of 13 patients man- bladder is tumor-free by means of a cystoscopy and pos-
aged with partial ureterectomy or nephrectomy alone for sible biopsy. Several urologists have advocated an endo-
high stage (≥pT2) disease developed ipsilateral recur- scopic approach to excising the distal ureter and bladder
rences. Other studies have found a low rate of ipsilateral cuff transvesically, thereby avoiding the need for a second
recurrence in patients managed with conservative resec- incision.105 The two principal endoscopic approaches
tion of high-grade tumors.38 However, it is possible that have been a “stripping” technique wherein a ureteral
the lower duration of subsequent survival in these catheter is placed and secured with a tie around the dis-
patients is too short to permit appearance of locally tal ureter. Following resection of the kidney and proxi-
recurrent tumors. mal two-thirds of the ureter, a resectoscope is used to
Removal of the kidney and only a portion of the ureter circumscribe the mucosa around the ureteral orifice
for a renal pelvic or proximal ureteric TCC has a high while applying upward traction on the catheter through
rate of recurrence in the remaining ureteric stump.8,37,101 the flank incision. This allows the intramural ureter to be
The incidence of tumor recurrence is proportional to the detached from the bladder along with a rim of surround-
residual length of ureter left behind, suggesting that it is ing bladder mucosa.106,107 There are multiple variations
a function of potentially affected urothelium left in to this approach with the orifice being cauterized to
situ.57,102,103 This observation has provided the impetus encourage closure or simply ligated. Another commonly
for complete ureterectomy in these patients. Zincke used technique is the “pluck” approach. The ureteral ori-
et al.93 found that recurrence rates following segmental fice is circumscribed endoscopically using a resectoscope
resection for distal ureteral TCC was much lower than loop or a hook electrode until perivesical fat is visi-
that for renal pelvis TCC following nephrectomy with ble.108,109 This denotes that the distal ureter is free from
partial ureterectomy (15% versus 62%, respectively). surrounding attachments and can then be extracted in

Table 15-3 Results of Segmental Ureteral Resection and Reimplantation


for Localized Ureteric Tumors
No. of Local Ureteral Duration of
Study Patients (n) Recurrences (%) Follow-Up (Months)

Johnson and Babaian95 6 1/6 (16.6) 44.4

Zungri et al.96 35 3/35 (8.5) 86

Maier et al.97 17 3/17 (17.6) 41.4

Wallace et al.98 7 1/7 (14.3) 93.6

Anderstrom et al39 21 1/21 (4.7) 83


278 Part III Kidney and Ureter

continuity with the renal specimen through the flank is now considered the standard of care by many. Cancer
incision. The mucosa around the opening in the bladder control and complication rates are reported to be compa-
wall is fulgurated to cause scarring and sealing of this rable to the open approach. The morbidity, as with other
area. A drain is left in place postoperatively. Mean blad- laparoscopic procedures, is significantly lower with
der recurrence rates using the two different approaches shorter hospital stays and recovery times (Table 15-4).
are comparable (19.3% versus 24%, respectively).105 The efficacy of both transperitoneal and retroperitoneal
However, a total of 9 patients (7%) have developed local laparoscopic nephroureterectomy appear to have similar
recurrence at the site of the ureteral orifice with the advantages over the traditional open approach.111,119 A
pluck technique, which is of significant concern. hand-assisted approach has been advocated by some as
Complication rates are higher with the stripping tech- easier for traditional surgeons to master and as a means
nique (10% versus 2.7%) and it may not be applicable in to allow better control and manipulation during the
all cases.105 The main complications with the pluck tech- removal of large tumors or more complex cases. Using
nique are the risk of intraperitoneal extravasation, while this approach Keeley et al.120 observed better retraction,
ureteral catheter-related problems, such as breakage/dis- dissection, and tactile feedback, which translated to
lodgement or retention of the catheter, are the main shorter operative times than standard “pure” laparoscopic
problems following the stripping approach. Prognosis nephroureterectomy. The hand-assisted laparoscopic nep-
and survival following traditional management of TCC hroureterectomy still requires a longer operative time
of the renal pelvis and ureter are very dependent on stage compared to the traditional open nephroureterectomy
and grade. The presence of renal parenchyma around the but hospital stay, time to oral intake, time to resuming
renal pelvis acts as an anatomic barrier and this impacts normal activity, and quality of life are better with the
prognosis. Stage T3 renal pelvis TCC is associated with hand-assisted approach as determined by prospective
a 54% 5-year survival rate, while that of ureteral T3 studies.121 In a prospective comparison of the standard
tumors is 24%.8,36 Metastatic upper tract TCC has a dis- laparoscopic nephroureterectomy to the hand-assisted
mal prognosis, with the majority of patients dying by 3 approach, Landman et al.111 found that complication
years and 0% of patients survive for 5 years, even with rates were slightly lower with the hand-assisted approach
administration of adjuvant therapy.37,62 Overall reported (45% versus 31%). Operative time was shorter with the
stage-specific 5-year survival rates for upper tract TCC hand-assisted approach and there were no differences in
are 100% for Ta/CIS tumors, 92% for T1, 73% for T3, hospital stay or postoperative pain medication require-
and 41% for T4 tumors.38 Median survival for those with ments. Return to work times were 1 to 3 weeks longer in
stage T4 disease can be as short as 6 months. Overall the hand-assisted group though this was not statistically
median survival does not appear to differ significantly significant. All three approaches appear to be oncologi-
between patients who undergo segmental ureterectomy cally equivalent as assessed by the incidence of recurrent
for a ureteral tumor compared to those who undergo a tumors. Based on these data the laparoscopic or hand-
nephroureterectomy, and is reported to be as high as 58 assisted approaches are considered by many to be the
months.38 Previous studies indicate a clear effect of both standard of care.
stage and grade on 5-year survival, with rates ranging There is still some controversy about the best method
from 54% to 75% for stage I/A/Tis, 43% to 87% for of excising the lower ureter with a cuff of bladder mucosa
stage II/T1 or T1, 0% to 54% for stage III/C/T3, and while performing a laparoscopic nephroureterectomy.
0% to 19% for stage IV/D/T4/N+ or M+.36 Most large- The methods described in the open nephroureterectomy
scale studies for treatment outcomes for upper tract section for endoscopic resection of the distal ureter
TCC using reported epidemiologic data from the transvesical to avoid a second incision are applicable to,
American College of Surgeons or the Surveillance or have been adapted for, use in conjunction with laparo-
Epidemiology and End Results (SEER) program con- scopic nephroureterectomy. Gill et al.122 have used an
sider mainly renal pelvis TCC, which is grouped with intravesical approach with needlescopic (2 mm) instru-
kidney cancers and which renders the data hard to inter- ments. Small-sized laparoscopic instruments are placed
pret. Most recent data from the American College of transvesically and the bladder cuff around the ureteric
Surgeons National Cancer Database indicate that overall orifice is circumscribed with electrocautery and dissected
survival for cancers of the kidney and renal pelvis is 76% around a previously placed ureteral balloon catheter,
(Figure 15-5). which occludes the ureter and prevents tumor spillage.
Dissection is carried out until the intramural portion of
the ureter is free of all attachments and the lower ureter
Laparoscopic Nephroureterectomy
and bladder cuff can be extracted in continuity with the
Since the first report of a laparoscopic nephroureterec- rest of the specimen. Shalhav et al.123 have proposed a
tomy in a child by the Washington University group in combined laparoscopic and endoscopic approach. An
1991,110 this approach has rapidly gained acceptance and occluding balloon catheter is placed in the ureter. The
Chapter 15 Transitional Cell Carcinoma of the Renal Pelvis and Ureter 279

100
90
80
70
60
Percent

50
40
30
20
10
0
At diag After 1 year. After 2 year. After 3 year. After 4 year. After 5 year.
SURVIVAL
When diagnosed on stage: I II III
IV Overall
Source: NCDB, Commission on Cancer, ACoS. Survial Reports, v2.0 −− March 9, 2004

Five Year Survival Table fo Kidney and Renal Pelvis Cancer Cases Diagnosed in 1995 &
1996
All States/Data Reported from 1653 Hospitals
Hospitals of type: All

Stage Cases At dx 1 year 2 year 3 year 4 year 5 year 95% Confidence Interval

I 5147 100 93.10 87.96 82.87 77.66 73.91 72.53 - 75.29

II 15158 100 92.94 87.10 82.35 77.12 72.31 71.47 - 73.15

III 6334 100 82.76 70.78 62.79 55.96 50.59 49.17 - 52.01

IV 9423 100 33.75 17.66 11.22 8.17 6.49 5.91 - 7.07

Overall 39944 100 75.00 65.22 59.30 54.28 50.35 49.79 - 50.91

Source: NCDB, Commission on Cancer, ACoS/ACS. Survival Reports, v2.0—March 9, 2004

Figure 15-5 Stage specific 5-year survival rates for kidney and renal pelvic tumors
diagnosed 1995–1996. (Data from the National Cancer Database maintained by the
American College of Surgeons, with permission.)

intramural ureter is unroofed using a hot Collins knife excise it along with a cuff of mucosa. All of the minimally
and a 1-cm cuff of bladder mucosa is circumscribed invasive approaches run the risk of leaving a portion of
around the ureteric orifice using a resectoscope until the intramural ureter intact.
perivesical fat is clearly visible. A rollerball electrode is
used to fulgurate the edges of the ureter. A balloon
CHEMOTHERAPY FOR ADVANCED DISEASE
catheter is replaced in the renal pelvis to prevent leakage
of tumor into the retroperitoneum. The lower ureter is Upper tract urothelial cancers are fairly chemosensitive.
thus freed of all attachments and can then be extracted in Given the relatively low frequency of upper tract TCC
continuity with the rest of the specimen. Other authors compared to bladder TCC, very few trials have focused
have reported simply resecting the ureteral orifice trans- on developing chemotherapeutic regimens specific for
vesically using a resectoscope.108 The availability of upper tract disease. Single-agent and combination regi-
hand-assisted laparoscopy has also permitted a more mens that have shown activity in bladder TCC have been
standard approach using hand traction on the lower generally used to treat upper tract disease with similar
ureter to dissect it down to the bladder extravesically and efficacy. Most of the data cited below pertain to bladder
280

Table 15-4 Comparison of Outcomes from Laparoscopic Nephroureterectomy with Open Nephroureterectomy
Part III Kidney and Ureter

Number of Mean Mean Local


Cases/Open Method of Distal Mean LOS Follow-up Recurrence
Study Approach Conversions Ureter Resection Pathology EBL (cc) (days) (Months) Rate

Chung et al.112 Retroperitoneal 6/14 Open bladder cuff NS NS 9 12.6 15% (B)

Salomon et al.113 Retroperitoneal 4/0 Open bladder cuff PT2-T3, grade 2-3 220 5.7 18 25% (local)

Keeley and Tolley114 Transperitoneal 18/3 Pluck NS NS NS NS 11%

Hobart et al.115 Retroperitoneal 17/2 Transvesical PTa-T4, grade 1-3 278 2.2 6.4 NS

Shalhav et al.116 Transperitoneal 25/0 Stapled PTa-T3, grade 1-4 199 3.6 39 40% (28% B,
12% local)

McNeill et al.117 Transperitoneal 25/3 Open bladder cuff PT1-T3, grade 1-3 5 35 16%*

Yoshino et al.118 Retroperitoneal 23/1 Stapled 2.5 cm cuff PTa-T4, Grade 1-3 304 NS 19* 17.4% (B)

Landman et al.111 Transperitoneal 11/0 Cystoscopic unroofing, PTa-T1, Grade 1-4 190 3.3 27.4 30% (B)
(1 retroperitoneal) cauterization

NS, Not significant; B, bladder.


*Median follow-up.
Chapter 15 Transitional Cell Carcinoma of the Renal Pelvis and Ureter 281

TCC but has served as a basis for the chemotherapeutic that the use of adjuvant radiation may decrease the like-
approaches adopted for upper tract TCC. lihood of local recurrence after nephroureterectomy in
Single-agent chemotherapy with cisplatin, methotrex- patients with locally advanced T3/T4 disease.138–140
ate, cyclophosphamide, or gemcitabine has yielded Brookland and Richter139 treated 23 patients determined
responses ranging from 25% to 35%.124,125 The methotrex- to have poor risk disease with surgery alone or surgery
ate, vinblastine, adriamycin, cyclophosphamide (MVAC) followed by adjuvant radiation. They found that 1 of 9
regimen, which has a reported response rate as high as (11%) evaluable patients treated with adjuvant radiation
70% in bladder TCC, can also be used for upper tract developed local and distant recurrence, while 5 of 11 (42%)
TCC.126,127 In direct comparisons, it is clear that combi- evaluable patients who were treated with surgery alone
nation therapy is more effective than single-agent ther- developed local or local and distant recurrence. Other
apy such as cisplatin.128 In one report, 20 patients with authors report similar low rates of local recurrence with
upper tract TCC were included in a randomized trial the use of adjuvant radiation therapy.138,141 The reported
comparing MVAC with the cisplatin, cyclophosphamide, doses of radiation used have ranged from 40 to 50 Gy.
adriamycin (CISCA) regimen.129 There was no differ- Adequate and complete local control of tumor is impor-
ence in response rates between these patients and those tant considering the fact that it can be an independent
with primary bladder TCC. In spite of observed com- predictor of survival.142 Maulard-Durdux et al.143
plete responses, there is a tendency for patients to relapse. reported the largest retrospective series, with 26 patients
Duration of response varies from 8.5 to 39 months.128 treated with adjuvant radiation of 45 Gy following com-
Alternative newer regimens, such as gemcitabine with plete surgical resection of primary tumor in patients with
cisplatin or paclitaxel with carboplatin have yielded locally advanced disease, of whom 42% had regional
response rates of 50% to 60%.130 A randomized trial nodal metastases. They noted a low local recurrence rate
comparing MVAC to gemcitabine with cisplatin obtained of 15% but the overall 5-year survival rate was not dif-
similar response rates, median overall survival, and dura- ferent than that obtained in historic series using surgery
tion of response.131 The gemcitabine/cisplatin regimen alone. These results highlight the fact that while local
was much less toxic and better tolerated. It is now con- failure is important, most patients who die succumb to
sidered the de facto standard for the treatment of most metastases, which is not in areas addressed by radiation.
urothelial cancers. A sequential approach has also been Hence, it is difficult to recommend routine adjuvant
used to treat urothelial cancers. Adriamycin/gemcitabine radiation even in patients with evidence of residual local
followed by ifosfamide/paclitaxel/cisplatin has been used disease. These individuals may benefit from adjuvant
in an effort to maximize response while limiting toxic- combination chemotherapy or a combination of radiosen-
ity.132 Results of randomized trials are awaited. sitizing single-agent chemotherapy with cisplatin fol-
Neoadjuvant chemotherapy has been used in upper lowed by radiation to optimize local control.
tract disease and more so in bladder cancer. Large ran-
domized trials have failed to show a clear survival benefit
OUTCOME
to this approach. One study using the cisplatin,
methotrexate, vinblastine (CMV) regimen demonstrated Overall risk of recurrence after initial standard treatment
a 5.5% higher survival in the neoadjuvant chemotherapy of upper tract TCC, which has traditionally been open
group at 3 years of follow-up, which was not statistically nephroureterectomy, has been related to stage and grade
significant.133 The theoretical advantage of neoadjuvant of tumor. Five-year survival can range from 100% for
chemotherapy in downstaging the tumor and allowing low-stage disease to 0% for T4 disease.70 More recent
organ preservation by conservative surgery does not series have indicated a local recurrence rate of 23% to
really exist for upper tract disease since the only candi- 27%.38,141 Almost 50% of these recurrences are in the
dates for such surgery are those with superficial disease. bladder, with 9% occurring in the retroperitoneum, 18%
Data supporting the use of adjuvant chemotherapy in in the remaining upper tract, and 22% being distant.38 In
urothelial cancers are also weak and this applies to upper multivariate analysis, tumor stage and surgical procedure
tract disease as well. Most trials have not shown a clear were related to the risk of recurrence.38 Only patient age
benefit and the advantage of routine adjuvant and stage were indicative of survival in multivariate
chemotherapy over salvage therapy at the time of recur- analysis. Local disease relapse tends to occur early and
rence is not well established.134–137 the median time to such recurrence can be 12 months.38
Follow-up of patients with upper tract TCC involves
obtaining periodic IVP and/or CT scans to image the
RADIATION THERAPY FOR UPPER TRACT TCC
upper tracts and rule out local disease recurrence. While
The use of radiation therapy as an adjuvant to surgery in there is no established follow-up schedule that is known
patients with locally advanced upper tract TCC has been to be optimal, it is reasonable to base it on stage/grade of
reported with variable results. Original studies suggested primary tumor and type of surgical procedure performed.
282 Part III Kidney and Ureter

We typically recommend obtaining an IVP 3 months in the Balkan nephropathy area. J Cancer Res Clin
postoperatively and a CT scan 6 months following sur- Oncol 1985; 110:181.
gery if the renal unit is left intact. Subsequent follow-up 15. Kakizoe T, Fujita T, Murase T, et al: Transitional cell
will include annual IVP and/or CT scan. In those who carcinoma of the bladder in patients with renal pelvic
and ureteral cancer. J Urol 1980; 124:17.
undergo nephroureterectomy, CT scans are obtained at
16. Kimball FN, Ferris HW: Papillomatous tumor of the
yearly intervals for stage T2 or higher and biannual stud-
renal pelvis associated with similar tumors of the ureter
ies for lower stage disease. We also obtain a voided urine and bladder. J Urol 1934; 31;257.
cytology and perform office cystoscopy to rule out the 17. Oldbring J, Cliffberg I, Mikulowski P, et al: Carcinoma
presence of bladder TCC in patients at 3-month intervals of the renal pelvis and ureter following bladder
for the first 2 years, every 6 months for the following carcinoma: frequency, risk factors and clinicopathologic
2 years, and yearly thereafter following treatment of findings. J Urol 1989; 141:1311.
upper tract TCC. 18. Rabbani F, Perrotti M, Russo P, et al: Upper tract tumors
after an initial diagnosis of bladder cancer: argument for
long term surveillance. J Clin Oncol 2001; 19:94.
19. Fadl-Elmula I, Gorunova L, Mandahl N, et al:
REFERENCES Cytogenetic analysis of upper urinary tract transitional
cell carcinomas. Cancer Genet Cytogenet 1999; 115:123.
1. Fraley EE: Cancer of the renal pelvis. In Skinner DG, 20. Hudson M, Herr HW: Carcinoma in situ of the bladder.
deKernion JB (eds): Genitourinary Cancer, p 134. J Urol 1995; 153:564.
Philadelphia, WB Saunders, 1978. 21. Herr HW, Wartinger DD, Oettgen HF: Bacillus
2. Melamed MR, Reuter VE: Pathology and staging of Calmette Guerin therapy for superficial bladder cancer:
urothelial tumors of the kidney and ureter. Urol Clin a 10-year follow-up. J Urol 1992; 147:1020.
North Am 1993; 20:333. 22. Zincke H, Garbeff PJ, Beahrs RJ, et al: Upper urinary
3. Jemal A, Tiwari RC, Murray T, et al: Cancer Statistics tract transitional cell cancer after radical cystectomy for
2004. CA Cancer J Clin 2004; 54:8. bladder cancer. J Urol 1994; 131:50.
4. McLaughlin JK, Silverman DT, Fraumeni JF, et al: 23. Malkowicz SB, Skinner DG: Development of upper tract
Cigarette smoking and cancers of the renal pelvis and carcinoma after cystectomy for bladder cancer. Urology
ureter. Cancer Res 1992; 52:254. 1990; 36:20.
5. Rubenstein MA, Walz BJ, Bucy JG: Transitional cell 24. Sharma TC, Melamaed MR, Whitmore WF: Carcinoma
carcinoma of the kidney: 25-year experience. J Urol in situ of the ureter in patients with bladder carcinoma
1978; 119:594. treated by cystectomy. Cancer 1970; 26:583.
6. Bennington JL, Beckwith JB: Tumors of the kidney, 25. Akaza H, Koiso K, Niijima T: Clinical evaluation of
renal pelvis and ureter. In Atlas of Tumor Pathology, 2nd urothelial tumors of the renal pelvis and ureter based on
series, Fascicle 12. Washington DC, Armed Forces a new classification system. Cancer 1987; 59:1369.
Institute of Pathology, 1975. 26. Wallace D: Cancer of the bladder. Am J Roentgenol
7. Vicente J, Pilar L, Sole-Balcells FJ, et al: Transitional cell 1968; 102:581.
carcinoma in the upper urinary tract: diagnosis and 27. Davies JM: Bladder tumors in electric cable industry.
management. Urol Int 1995; 2:7. Lancet 1965; 2:143.
8. Mazeman E: Tumors if the upper urinary tract calyces, 28. Ross RK, Paganini-Hill A, Henderson BE: Analgesics,
renal pelvis and ureter. Eur Urol 1976; 2:120. cigarette smoking and other risk factors for cancer of the
9. Petkovic SD: Conservation of the kidney in operations renal pelvis and ureter. Cancer Res 1989; 49:1045.
for tumors of the renal pelvis and calyces. Br J Urol 29. Petkovic SD: Epidemiology and treatment of renal pelvic
1972; 44:1. and ureteral tumor. J Urol 1975; 14:858.
10. Murphy DM, Zincke H, Furlow WL: Management of 30. Rosenberg L, Rao RS, Palmer JR, et al: Transitional cell
high-grade transitional cell cancer of the upper tract. cancer of the urinary tract and renal cell cancer in
J Urol 1980; 125:25. relation to acetaminophen use (United States). Cancer
11. Huben RP, Mounzer AM, Murphy GP: Tumor grade Causes Control 1998; 9:83.
and prognostic variables in upper tract urothelial tumors. 31. Levine E: Transitional cell carcinoma of the renal pelvis
Cancer 1988; 62:2016. associated with cyclophosphamide therapy. Am J
12. Krogh J, Kvist E, Rye B: Transitional cell carcinoma of Roentgenol 1992; 159:1027.
the upper urinary tract: prognostic variables and post 32. Verhaak RL, Harmsen AE, Van Urink AJM, et al: On
operative recurrences. Br J Urol 1991; 67:32. the frequency of tumor induction in a Thorotrast kidney.
13. Ressequie LT, Nobrega FT, Farrow GM, et al: Cancer 1974; 34:2061.
Epidemiology of renal and ureteral cancer in Rochester, 33. Schwalb DM, Herr HW, Fair WR: The management of
Minnesota, 1950-1974 with special reference to clinical clinically unconfirmed positive urinary cytology. J Urol
and pathological features. Mayo Clin Proc 1978; 1993; 150:1751.
53:503. 34. Herr HW, Cookson MS, Soloway SM: Upper tract
14. Radovanovic Z, Krajinovic S, Jankovic S, et al: Family tumors in patients with primary bladder cancer followed
history of cancer among cases of upper urothelial tumors for 15 years. J Urol 1996; 156:1286.
Chapter 15 Transitional Cell Carcinoma of the Renal Pelvis and Ureter 283

35. Raabe NK, Fossa SD, Bjerkehagen B: Carcinoma of the 54. Sheline AM, Amendola MA, Pollack HM, et al:
renal pelvis. Scand J Urol Nephrol 1992; 26:357. Fluoroscopically guided retrograde brush biopsy in the
36. Guinan P, Vogelzang NJ, Sylvester J, et al: Renal pelvic diagnosis of transitional cell carcinoma of the upper
cancer: a review of 611 patients treated in Illinois urinary tract: results in 45 patients. Am J Roentgenol
1975–1985. Urology 1992; 40:393. 1989; 153:313.
37. Kleer E, Oesterling JE: Transitional cell carcinoma of 55. Holtz F: Papillomas and primary carcinoma of the
the upper tracts. Prob Urol 1992; 6:531. ureter: report of 20 cases. J Urol 1962; 88:380.
38. Hall MC, Womack S, Sagalowsky AI, et al: Prognostic 56. Donat SM, Herr HW: Transitional cell carcinoma of the
factors, recurrence and survival in transitional cell renal pelvis and ureter: diagnosis, staging, management
carcinoma of the upper urinary tract: a 30-year and prognosis. In Richie J, Oesterling JE (eds):
experience in 252 patients. Urology 1998; 52:594. Urologic Oncology, p 215. Philadelphia, WB Saunders,
39. Anderstrom C, Johansson SL, Pettersson S, et al: 1997.
Carcinoma of the ureter: a clinicopathological study of 57. Herr HW, Whitmore WF: Ureteral carcinoma in situ
49 cases. J Urol 1989; 142:280. after successful intravesical therapy for superficial
40. Gaboardi F, Bozzola A, Dotti E, et al: Conservative bladder tumors: incidence, pathogenesis and
treatment of upper urinary tract tumors with Nd:Yag management. J Urol 1987; 138:292.
laser. J Endourol 1994; 8:37. 58. Freiha FS: Renal, renal pelvis and ureteral tumors:
41. Igawa M, Urakami S, Shiina H, et al: Limitations of should retroperitoneal nodes be treated? Front Radiat
ureteroscopy in diagnosis of invasive upper tract Ther Oncol 1994; 28:155.
urothelial cancer. Urol Int 1996; 56:13. 59. Bergkvist A, Ljungqvist A, Moberger G: Classification of
42. McTavish JD, Jinzaki M, Zou KH, et al: Multi-detector bladder tumors based on the cellular pattern.
row CT urography: comparison of strategies for Preliminary report of a clinical-pathological study of 300
depicting the normal urinary collecting system. cases with a minimum follow-up of eight years. Acta
Radiology 2002; 225:783. Chir Scand 1965; 130:371.
43. Caoili EM, Cohan RH, Korobkin M, et al: Urinary tract 60. Epstein JE, Amin MB, Reuter VE, et al: The World
abnormalities: initial experience with multi-detector row Health Organization/International Society of Urologic
CT urography. Radiology 2002; 222:353. Pathology consensus classification of urothelial
44. Zielonko J, Studniarek M, Markuszewski M: MR (transitional cell) neoplasms of the urinary bladder.
urography of obstructive uropathy: diagnostic value of Bladder Consensus Conference Committee. Am J Surg
the method in selected clinical groups. Eur Radiol 2003; Pathol 1998; 22:1435.
13:802. 61. Grabstald J, Whitmore WF, Melamed MR: Renal pelvic
45. Konety BR, Getzenberg RH: Urine based markers of tumors. JAMA 1971; 218:845.
urologic malignancy. J Urol 2001; 165:600. 62. Das AK, Culley CC, Paulson DF, et al: Primary
46. Konety BR, Mitro M, Melhem M, et al: Diagnostic value carcinoma of the upper urinary tract: effect of primary
of voided urine and bladder barbotage cytology in and secondary therapy on survival. Cancer 1990;
detecting transitional cell carcinoma of the urinary tract. 66:1919.
Urol Int 1999; 62:26. 63. Blute ML, Tsushima K, Lieber MM, et al: Transitional
47. Sadek S, Soloway MS, Hook S, et al: The value of upper cell carcinoma of the renal pelvis: nuclear
tract cytology after transurethral resection of bladder deoxyribonucleic acid ploidy studied by flow cytometry.
tumor in patients with bladder transitional cell cancer. J Urol 1988; 140:944.
J Urol 1999; 161:77. 64. Haupt G: Transitional cell carcinoma of the ureter.
48. Carpinito GA, Stadler WM, Briggman JV, et al: Urinary J Endourol 2001; 15:409.
nuclear matrix protein as a marker for transitional cell 65. Strong DW, Pearse HD: Recurrent urothelial tumors
carcinoma of the urinary tract. J Urol 1996; 156:1280. following surgery for transitional cell carcinoma of the
49. Walsh IK, Keane PF, Ishak LM, et al: The BTA stat test: upper urinary tract. Cancer 1976; 38:2178.
a tumor marker for the detection of upper tract 66. Vest SA: Conservative surgery in certain benign tumors
transitional cell carcinoma. Urology 2001; 58:532. of the ureter. J Urol 1945; 53:97.
50. Wu WJ, Liu LT, Huang CN, et al: The clinical 67. Chen GL, El-Gabry EA, Bagley DH: Surveillance of
implications of telomerase activity in upper tract upper urinary tract transitional cell carcinoma: the role
urothelial cancer and washings. BJU Int 2000; 86:213. of ureteroscopy, retrograde pyelography, cytology and
51. Kamai T, Takagi K, Asami H, et al: Prognostic urinalysis. J Urol 2000; 164:1901.
significance of p27 Kip1 and Ki-67 expression in 68. Keeley FX, Culp DA, Bibbo M, et al: Diagnostic
carcinoma of the renal pelvis and ureter. BJU Int 2000; accuracy of ureteroscopic biopsy in upper tract
86:14. transitional cell carcinoma. J Urol 1997; 157:33.
52. Gill WB, Lu C, Bibbo M: Retrograde brush biopsy of 69. Guarnizo E, Pavlovich CP, Seiba M, et al: Ureteroscopic
the ureter and renal pelvis. Urol Clin North Am 1979; biopsy of upper tract urothelial carcinoma: improved
6:573. diagnostic accuracy and histopathological considerations
53. Blute ML, Segura JW, Zincke H, et al: Impact of using a multi-biopsy approach. J Urol 2000; 153:52.
endourology on diagnosis and management of upper 70. Heney NM, Nocks BN, Daly JJ, et al: Prognostic factors
urinary tract urothelial cancer. J Urol 1989; 141:1298. in carcinoma of the ureter. J Urol 1981; 125:632.
284 Part III Kidney and Ureter

71. Keeley FX, Bibbo M, Bagley DH: Ureteroscopic 90. Goel MC, Mahendra V, Roberts JG: Percutaneous
treatment and surveillance of upper urinary tract management of renal pelvic urothelial tumors: long-term
transitional cell carcinoma. J Urol 1997; 157:1560. follow-up. J Urol 2003; 169:925.
72. Chen GL, Bagley DH: Ureteroscopic surgery for upper 91. Thalmann GN, Markwalder R, Walter B, et al: Long-
tract transitional cell carcinoma: complications and term experience with bacillus Calmette–Guerin therapy
management. J Endourol 2001; 15:399. of upper tract transitional cell carcinoma in patients not
73. Bagley D, Erhard M: Use of the holmium laser in the eligible for surgery. J Urol 2002; 168:1381.
upper urinary tract. Tech Urol 1995; 1:25. 92. Eastham JA, Huffman JL: Technique of mitomycin C
74. Elliott DS, Blute ML, Patterson DE, et al: Long instillation in the treatment of upper tract urothelial
term follow-up of endoscopically treated upper tumors. J Urol 1993; 150:324.
urinary tract transitional cell carcinoma. Urology 1996; 93. Zincke H, Neves RJ: Feasibility of conservative surgery
47:819. for transitional cell cancer of the upper urinary tract.
75. Englemeyer EL, Belis JA: Long-term ureteroscopic Urol Clin North Am 1984; 11:717.
management of low grade transitional cell carcinoma of 94. Pohar KS, Sheinfeld J: When is partial ureterectomy
the upper urinary tract. Tech Urol 1996; 2:113. acceptable for transitional cell carcinoma of the ureter?
76. Martinez-Pineiro JA, Matres-Garcia MJ, Martinez- J Endourol 2001; 15:405.
Pineiro L: Endourologic treatment of upper tract 95. Johnson DE, Babaian RJ: Conservative surgical
urothelial carcinoma. Analysis of a series of 59 tumors. management for non-invasive distal ureteral carcinoma.
J Urol 1996; 156:377. Urology 1979; 13:365.
77. Johnson DE: Treatment of distal ureteral tumors using 96. Zungri E, Chechile G, Algaba F, et al: Treatment of
endoscopic argon photoirradiation. Lasers Surg Med transitional cell carcinoma of the ureter: is the
1992; 12:490. controversy justified? Eur Urol 1990; 17:276.
78. Schilling A, Bowering R, Keiditsch E: Use of the 97. Maier U, Mertl G, Pummer K, et al: Organ-preserving
neodymium:Yag laser in the treatment of ureteral tumors surgery in patients with urothelial tumors of the upper
and urethral condylomata acuminata. Clinical urinary tract. Eur Urol 1990; 18:197.
experience. Eur Urol 1986; 12(Suppl 1):30. 98. Wallace DMA, Wallace DM, Whitfield HN, et al: The
79. Smith AD, Orihuela E, Crowley AR: Percutaneous late results of conservative surgery for upper tract
management of renal pelvic tumors: a treatment option urothelial carcinomas. Br J Urol 1981; 53:537.
in selected cases. J Urol 1988; 140:745. 99. Palou J, Salvador J, Millan F, et al: Management of
80. Jabbour ME, Smith AD: Primary percutaneous approach superficial transitional cell carcinoma in the intramural
to upper urinary tract transitional cell carcinoma. Urol ureter. What to do? J Urol 2000; 163:744.
Clin North Am 2000; 27:739. 100. Leitenberger A, Beyer A, Altwein JE: Organ sparing
81. Tomera KM, Leary FJ, Zincke H: Pyeloscopy in treatment for ureteral carcinoma. Eur Urol 1996;
urothelial tumors. J Urol 1982; 127:1088. 29:272.
82. Sengupta S, Harewood L: Transitional cell carcinoma 101. Booth CM, Cameron KM, Pugh RCB: Urothelial
growing along an in-dwelling nephrostomy tube track. carcinoma of the kidney and ureter. Br J Urol 1980;
Br J Urol 1998; 82:591. 52:430.
83. Sharma NK, Nicol A, Powell CS: Track infiltration 102. Kakizoe T, Fujita J, Kishi K, et al: Transitional cell
following percutaneous resection of renal pelvic carcinoma of the bladder in patients with renal pelvic
transitional cell carcinoma. Br J Urol 1994; 73:597. and ureteral cancer. J Urol 1980; 124:17.
84. Huang A, Low RK, DeVere White R: Nephrostomy 103. Strong DW, Pearse HD, Hodges CV, et al: The
tract tumor seeding following percutaneous ureteral stump after nephroureterectomy. J Urol 1976;
manipulation of a ureteral carcinoma. J Urol 1995; 115:654.
153:1041. 104. Charbit L, Gendreau MC, Cukier J, et al: Tumors of the
85. Fulgsig S, Krarup T: Percutaneous nephroscopic upper urinary tract: 10 years of experience. J Urol 1991;
resection of renal pelvic tumors. Scand J Urol Nephrol 146:1243.
1995; 172 (Suppl):15. 105. Laguna MP, de la Rosette JJ: The endoscopic approach
86. Oefelein MG, MacLennan G: Transitional cell to the distal ureter in nephroureterectomy for upper
carcinoma recurrence in the nephrostomy tract after urinary tract tumor. J Urol 2001; 166:2017.
percutaneous resection. J Urol 2003; 170:521. 106. McDonald HP, Upchurch WE, Sturdevant CE: Nephro-
87. Clark PE, Streem SB: Endourologic management of ureterectomy: a new technique. J Urol 1952; 67:804.
upper tract transitional cell carcinoma. AUA Update 107. McDonald DF: Intussusception ureterectomy: a method
Series, Vol. 18, Lession XVI, 1999. of removal of the ureteral stump at time of
88. Studer UE, Casanova G, Kraft R, et al: Percutaneous nephroureterectomy without an additional incision. Surg
bacillus Calmette-Guérin perfusion of the upper urinary Gynecol Obstet 1953; 97:565.
tract for carcinoma in situ. J Urol 1989; 142:975. 108. Palou J, Caparros J, Orsola A, et al: Transurethral
89. Okubo K, Ichioka K, Terada N, et al: Intrarenal bacillus resection of the intramural ureter as the first step of
Calmette–Guerin therapy for carcinoma in situ of the nephroureterectomy. J Urol 1995; 154:43.
upper urinary tract: long term follow-up and natural 109. Abercrombie GF: Nephroureterectomy. Proc R Soc Med
course in cases of failure. BJU Int 2001; 88:343. 1972; 65:1021.
Chapter 15 Transitional Cell Carcinoma of the Renal Pelvis and Ureter 285

110. Clayman RV, Kavoussi LR, Figenshau RS, et al: 126. Sternberg CN, Yagoda A, Scher HI, et al: Preliminary
Laparoscopic nephroureterectomy: initial case report. results of M-VAC (methotrexate, vinblastine, adriamycin,
J Laparoendosc Surg 1991; 1:343. cisplatin) for transitional cell carcinoma of the
111. Landman J, Lev RN, Bhayani S, et al: Comparison of urothelium. J Urol 1985; 133:403.
hand-assisted and standard laparoscopic radical 127. Sternberg CN, Yagoda A, Scher HI, et al: Methotrexate,
nephroureterectomy for the management of localized vinblastine, doxorubicin and cisplatin for advanced
transitional cell carcinoma. J Urol 2002; 167:2387. transitional cell carcinoma of the urothelium. Efficacy
112. Chung HJ, Chiu AW, Chen KK, et al: and patterns of response and relapse. Cancer 1989;
Retroperitoneoscopy assisted nephroureterectomy for 64:2448.
the management of upper tract urothelial cancer. Min 128. Loehrer PJS, Einhorn LH, Elson PJ, et al: A randomized
Invasive Ther 1996; 5:266. comparison of cisplatin alone or in combination with
113. Salomon L, Hoznek A, Cicco A, et al: methotrexate, vinblastine and doxorubicin in patients
Retroperitoneoscopic nephroureterectomy for renal with metastatic urothelial carcinoma: a cooperative
pelvic tumors with a single iliac incision. J Urol 1999; group study. J Clin Oncol 1992; 10:1066.
161:541. 129. Logothetis CJ, Dexeus FH, Finn L, et al: A prospective,
114. Keeley FX, Tolley DA: Laparoscopic randomized trial comparing MVAC and CISCA
nephroureterectomy: making management of upper-tract chemotherapy for patients with metastatic urothelial
transitional cell carcinoma entirely minimally invasive. tumors. J Clin Oncol 1990; 8:1050.
J Endourol 1998; 12:139. 130. Vogelzang NJ, Stadler WM: Gemcitabine and other new
115. Hobart MH, Gill IS, Sung GT, et al: Radical chemotherapeutic agents for the treatment of metastatic
nephroureterectomy for upper tract TCC: laparoscopy bladder cancer. Urology 1999; 53:243.
versus open surgery. J Endourol 1999; 13(Suppl 1):A44. 131. von der Maase H, Hansen SW, Roberts JT, et al:
116. Shalhav AL, Dunn MD, Portis AJ, et al: Laparoscopic Gemcitabine and cisplatin versus methotrexate, vinblastine,
nephroureterectomy for upper tract transitional cell doxorubicin, and cisplatin in advanced or metastatic bladder
cancer: the Washington University experience. J Urol cancer: results of a large, randomized, multinational,
2000; 163:1100. multicenter, phase III study. J Clin Oncol 2000; 18:3068.
117. McNeill SA, Chrisofos M, Tolley DA: The long-term 132. Dodd PM, McCaffrey JA, Hilton S, et al: Phase I
outcome after laparoscopic nephroureterectomy: a evaluation of sequential doxorubicin, gemcitabine then
comparison with open nephroureterectomy. BJU Int ifosfamide, paclitaxel, cisplatin for patients with
2000; 86:619. unresectable or metastatic transitional cell carcinoma of
118. Yoshino Y, Ono Y, Hattori R, et al: Retroperitoneoscopic the urothelial tract. J Clin Oncol 2000; 18:840.
nephroureterectomy for transitional cell carcinoma of 133. Anonymous: Neoadjuvant cisplatin, methotrexate, and
the renal pelvis and ureter: Nagoya experience. Urology vinblastine chemotherapy for muscle invasive bladder
2003; 61:533. cancer: a randomized controlled trial. International
119. Kawauchi A, Fujito A, Ukimura O, et al: Hand-assisted collaboration of trialists. Lancet 1999; 354:533.
retroperitoneoscopic nephroureterectomy: comparison 134. Studer UE, Bacchi M, Biedermann C, et al: Adjuvant
with the open procedure. J Urol 2003; 169:890. cisplatin chemotherapy following cystectomy for bladder
120. Keeley FX, Sharma NK, Tolley DA: Hand-assisted cancer: results of a prospective randomized trial. J Urol
laparoscopic nephroureterectomy. Br J Urol 1999; 1994; 152:81.
83:504. 135. Stockle M, Meyenburg W, Wellek S, et al: Adjuvant
121. Seifman BD, Montie JE, Wolf JS Jr: Prospective polychemotherapy of non-organ confined bladder cancer
comparison between hand-assisted laparoscopic and open after radical cystectomy revisited: long term results of a
surgical nephroureterectomy for urothelial carcinoma. controlled prospective study and further clinical
Urology 2001; 57:133. experience. J Urol 1995; 153:47.
122. Gill IS, Sobel JJ, Miller SD, et al: A novel technique for 136. Freiha FS, Reese J, Torti FM: A randomized trial of
the management of the en-bloc bladder cuff and distal radical cystectomy versus radical cystectomy plus
ureter during laparoscopic nephroureterectomy. J Urol cisplatin, vinblastine and methotrexate chemotherapy
1999; 161:430. for muscle invasive bladder cancer. J Urol 1996;
123. Shalhav AL, Elbahnasy AM, McDougall E, et al: 155:495.
Laparoscopic nephroureterectomy for upper tract 137. Bono A, Benvenuti C, Gibba A: Adjuvant chemotherapy
transitional cell cancer: technical aspects. J Endourol in locally advanced bladder cancer: final analysis of a
1998; 12:345. controlled multicenter study. Acta Urol Ital 1997;
124. McCaffrey JA, Dodd PM, Herr H, et al: Nonbladder 11:1241.
primary site of transitional cell carcinoma (TCC) does 138. Babaian RJ, Johnson DE, Chan RC: Combination
not affect probability of response to MVAC or survival. nephroureterectomy and postoperative radiotherapy for
J Clin Oncol 1998; 17(Suppl):337a. infiltrative ureteral carcinoma. Int J Radiat Oncol Biol
125. Stadler WM, Kuzel T, Roth B, et al: Phase II study of Phys 1980; 6:1229.
single agent gemcitabine in previously untreated patients 139. Brookland RK, Richter MP: Postoperative irradiation of
with metastatic urothelial cancer. J Clin Oncol 1997; transitional cell carcinoma of the renal pelvis and ureter.
15:3394. J Urol 1985; 133:952.
286 Part III Kidney and Ureter

140. Batata MA, Whitmore WF Jr, Hilaris BS, et al: Primary multicentre rare cancer network study. Eur J Cancer
carcinoma of the ureter: a prognostic study. Cancer 1999; 35:738.
1968; 35:1626. 143. Moulad-Durdux C, Dufour B, Hennequin C, et al:
141. Cozad SC, Smalley SR, Austenfeld M, et al: Transitional Postoperative radiation therapy in 26 patients with
cell carcinoma of the renal pelvis and ureter: patterns of invasive transitional cell carcinoma of the upper urinary
failure. Urology 1995; 46:796. tract: no impact on survival? J Urol 1996; 155:115.
142. Ozsahin M, Zouhair A, Villa S, et al: Prognostic factors
in urothelial renal pelvis and ureter tumors: a
C H A P T E R

16Management of Upper Urinary Tract


Transitional Cell Carcinoma
Andrew J. Dreslin, MD, and Graeme S. Steele, MD

Transitional cell carcinoma (TCC) is a malignant disease largely on the histology of the tumor. Merely 20% to
that affects the urothelium of the urinary tract. Although 30% of upper tract urothelial tumors are low-grade, pap-
TCC of the upper urinary tract accounts for only 5% of illary lesions with favorable prognosis.2 Multifocal
urothelial tumors, this incidence is increasing.1 This may tumors are common, the incidence of that is directly
be due in part to improved treatment and survival of related to tumor grade.2 TCC has a high rate of ipsilat-
patients with TCC of the bladder. In general, clinical eral recurrence following conservative management.
symptoms and signs, such as dysuria and hematuria, do Proximal-to-distal ipsilateral recurrence is common,
not differentiate between tumors of the upper and lower while recurrence in a distal-to-proximal direction
urinary tracts. However, patients with upper tract tumors remains rare.3 In a retrospective study of 252 patients
may present with flank pain due to obstruction by tumor treated for upper tract TCC, tumor stage was a signifi-
or blood clot (Figure 16-1). Constitutional symptoms of cant predictor of recurrence on multivariate analysis.4
weight loss, fatigue, and anemia may indicate more This observation lends support to radical treatment of
advanced disease. these tumors, although increasing evidence supports the
Initial evaluation of suspected upper urinary tract use of more conservative approaches for low-grade and
tumors includes complete urinary tract imaging, urinary low-stage tumors.
cytology, and direct endoscopic visualization. Traditio- TCC of the renal pelvis and ureter can invade local
nally, the intravenous pyelogram (IVP) was employed to structures or spread by lymphatic and hematogenous
detect filling defects within upper urinary tract. However, routes. Common sites of hematogenous metastasis
with increasing availability and decreasing costs, the com- include the lungs, liver, and bones. Lymphatic spread is
puted tomography (CT) urogram provides detailed images initially to regional hilar, paraaortic, and paracaval nodes
not only of the urinary tract but also of the other abdom- for renal pelvis and proximal ureteral tumors, while dis-
inal structures, including regional lymph nodes. As a tal ureteral tumors invade pelvic nodes. Thus, a thorough
result, CT urography has replaced the IVP in many cen- preoperative evaluation is necessary prior to any decision
ters, where CT technology is readily available. In addition, on treatment strategy. This includes chest x-ray, abdom-
retrograde pyelography at the time of cystoscopy to evalu- inal CT, liver function tests, and occasional bone scans.
ate gross hematuria is widely employed to evaluate upper In this chapter, several treatment strategies are discussed,
tract morphology (Figure 16-2). including open radical nephroureterectomy (NU),
Technologic improvements in visual optics have laparoscopic NU, open nephron-sparing surgery, and
allowed direct endoscopic visualization of the entire uri- endoscopic or percutaneous management. The technical
nary tract. This advancement has readily provided the aspects, as well as the indications, and advantages of each
ability by visual inspection to differentiate soft tissue fill- procedure are also discussed.
ing defects from renal and ureteral stones, in addition to
the option of biopsy of suspected lesions and in some
OPEN RADICAL NEPHROURETERECTOMY
cases definitive treatment.
Multiple treatment options now exist for tumors of Traditionally, open radical NU with excision of bladder
the renal pelvis and ureter. Treatment decisions are based cuff has been regarded as the gold-standard therapy for

287
288 Part III Kidney and Ureter

Figure 16-1 Left retrograde pyelogram (RPG) in a 66-year


old male who presented with left flank pain and gross Figure 16-2 Left retrograde pyelogram in a patient with
hematuria. RPG revealed a renal pelvic filling defect, which gross hematuria showing filling defects due to transitional cell
proved to be a well-differentiated TCC, and was managed by in the proximal ureter and renal pelvis. Metastatic evaluation
percutaneous resection. revealed pulmonary parenchymal metastatic disease.

TCC of the upper urinary tract in patients with a normal be extended towards the pelvis allowing dissection of the
functioning contralateral kidney. While this approach distal ureter and bladder cuff. Conversely, a second lower
has some advantages in certain situations, laparoscopic abdominal midline incision may be used for this part of
NU is now regarded as the more appropriate approach in the procedure. The use of an ipsilateral Gibson or
the majority of patients requiring NU for upper tract Pfannenstiel incision may be substituted for a lower mid-
TCC. line incision, although the increased muscle dissection
Open NU with excision of bladder cuff is still appropri- with this approach may contribute to further postopera-
ate in very large, invasive tumors with possible local exten- tive morbidity. The flank dissection proceeds in a
sion, including renal vein or vena caval involvement and retroperitoneal or intraperitoneal fashion. Thoraco-
gross lymphadenopathy. Furthermore, cytoreductive sur- abdominal surgical exposures are hardly ever required.
gery in the setting of metastatic disease is often best accom- Either intraperitoneal or retroperitoneal dissection can
plished by an open, rather than a laparoscopic, approach. easily accomplish exposure and dissection of the distal
Open NU with resection of bladder cuff can be per- ureter and bladder.
formed through one or two incisions. The patient is Open NU with resection of bladder cuff is com-
placed in a flank-torque position allowing access to the menced with radical nephrectomy, which includes
kidney and lower abdomen. The surgical prep includes removal of the kidney and surrounding Gerota’s fascia.
the flank, abdomen, and genitalia, providing for sterile The renal vessels are identified and tagged with a vessel
bladder catheter placement at the beginning of the pro- loop prior to ligation. The renal artery is ligated and
cedure. Table flexion or the use of a kidney rest usually transected first, followed by the renal vein. A combina-
provides improved surgical exposure of the kidney. An tion of nonabsorbable suture tie and hemostatic clips or
initial flank incision is made over the 11th or 12th rib, suture ligature is used to secure the vessel stumps. As the
removing the tip of the 11th or 12th rib usually enhances incidence of adrenal involvement is low with renal pelvis
access to the kidney and renal pedicle. This incision may and ureteral tumors, ipsilateral adrenalectomy may be
Chapter 16 Management of Upper Urinary Tract Transitional Cell Carcinoma 289

Regional lymphadenectomy should be performed dur-


ing radical NU. For the tumors of renal pelvis and prox-
imal ureter ipsilateral hilar lymph nodes are dissected.
The paraaortic nodes extending from the renal hilum to
the aortic bifurcation are included with left-sided tumors.
For right-sided tumors, paracaval and retrocaval nodes
are dissected. With distal ureteral tumors, ipsilateral
common iliac, external iliac, internal iliac, and obturator
nodes are excised. Inclusion of a regional lymphadenec-
tomy adds little additional operative time and postopera-
tive morbidity, while it may provide therapeutic benefit
for patients with local disease and identify patients who
may benefit from adjuvant chemotherapy.8 Additional
series suggest a possible therapeutic benefit for patients
with lymph node positive disease.9
In general, NU (open or laparoscopic) remains the
gold-standard therapy for patients with upper urinary
tract TCC and a normal functioning contralateral kid-
ney. Hall et al.4 reported decreased local recurrence fol-
lowing radical versus nephron-sparing surgery. In a
review of 100 patients treated for upper urinary tract
TCC, the 15-year cancer-specific survival was 69% for
patients treated with radical NU compared to 25% for
patients treated with nephron-sparing surgery.5 A 5-year
Figure 16-3 Open NU includes resection of the ureteral survival advantage following radical NU was also seen in
orifice and bladder cuff, usually accomplished with a ureteric patients with low-grade disease, while the advantage was
catheter in situ to provide traction and ureteric exposure. lost with high-grade tumors owing to the high incidence
of metastatic disease.10 The importance of nephrectomy
with total ureterectomy was seen in a series comparing
avoided in patients with distant tumors and normal pre- simple nephrectomy versus radical NU in patients with
operative imaging. TCC of the renal pelvis.11 The authors of the above-
After mobilizing the kidney and Gerota’s fascia, the mentioned references demonstrated a 5-year survival
ureter is dissected along its course to the bladder. To advantage of 84% versus 51% for the radical surgery.
reduce the risk of tumor spillage, transection of the kid- Contrary to the previous study, further benefit was seen
ney from ureter is avoided. The distal ureteral dissection in patients with high-grade tumors, with a 5-year survival
should include removal of the intramural ureter and a of 74% versus 37% for radical NU and simple nephrec-
cuff of bladder mucosa. Recurrent TCC is seen in the tomy, respectively. The benefit of NU is well defined for
ureteral stump of 23 % to 64% of patients following patients with high-grade, organ-confined disease and
incomplete ureteral dissection.5-7 An anterior cystotomy may be of value in patients with locally advanced disease
allows access to the ureteral orifice and transmural without evidence of systemic metastasis. In the setting of
ureter. Needlepoint electrocautery and counter-traction bilateral tumors, solitary kidney, or poorly functioning
facilitate this portion of the dissection. contralateral kidney, the benefit of NU must be
Upon removing the kidney, ureter, and bladder cuff, weighed against the quality of life changes following radical
the bladder defects are closed in two layers with surgery.
absorbable suture. Alternatively, extravesical dissection of
the intramural ureter avoids the creation of an anterior
LAPAROSCOPIC RADICAL
cystotomy. Traction on the dissected ureter brings the
NEPHROURETERECTOMY
surgical plane into the field. Care must be taken to
remove completely the transmural ureter and ureteral Laparoscopic NU has emerged as an effective alternative
orifice as these may be the sites of recurrence. Again, the to open surgery in most patients with upper urinary tract
bladder defect is closed in two layers with absorbable TCC. Clayman et al.12 initially reported the use of
sutures. A bladder catheter is left in situ for 7 to 10 days, laparoscopic technique for NU in a patient with TCC of
then a cystogram determines adequate healing. An the renal pelvis. Subsequently, multiple techniques have
extravesical closed suction drain is used to detect a urine been described and include transperitoneal,13 retroperi-
leak postoperatively (Figure 16-3). toneal,14 and hand-assisted laparoscopy.15 The preferred
290 Part III Kidney and Ureter

approach is largely surgeon’s preference, although body rant, midline supraumbilical, or mid-axillary subcostal.
habitus and tumor characteristics of the patient may This latter site is particularly of benefit in right-sided
guide this decision. The patient is positioned supine or tumors to assist with retraction of the liver. Initially, the
oblique and is prepped in a similar fashion to the open colon is mobilized medially by releasing the renocolic
procedure, again allowing access to a bladder catheter on ligaments, allowing access to the kidney. The ureter is
the surgical field. In laparoscopic surgery, the use of table now identified at the lower pole of the kidney. By tracing
flexion or a kidney rest can make access to the upper pole the ureter towards the kidney, the renal hilum comes into
kidney more difficult as the surgeon’s reach is limited by view. First the renal artery then the renal vein are ligated
the length of the laparoscopic instruments. If total and transected using a vascular stapling device. On freeing
laparoscopic surgery is planned, cystoscopy with electro- the kidney from its vessels, the remaining superior and
cautery dissection of the ureteral orifice and intramural lateral dissections of the kidney are performed. With the
ureter is performed at the beginning of surgery. At the kidney completely mobilized, the dissection continues in
Cleveland Clinic, laparoscopic radical NU comprises a caudal direction along the ureter. Distal to the iliac ves-
transvesical needlescopic-assisted cystoscopy detachment sels, the vas deferens in men or round ligament in women
of the en bloc bladder cuff and ureter, followed by is encountered and ligated. The ureteral dissection is con-
retroperitoneal removal of the kidney and ureter16 tinued to the bladder. Traction on the ureter will evert the
(Figure 16-4). Under cystoscopic control 2.2-mm ureteral orifice and enable a vascular stapling device to
needlescopic ports are inserted suprapubically into the ligate and transect the distal margin. The umbilical trocar
bladder site can be extended to allow removal of the specimen.
A ureteral catheter is cystoscopically passed into the Alternatively, the specimen can be morcellated in a speci-
ureteral orifice up to the renal pelvis, and the ureteral men bag and removed through the trocar site. Finally, the
orifice is grasped with a needlescopic grasper inserted fascial and skin layers of the port sites are closed.
through the contralateral suprapubic port and retracted For hand-assisted laparoscopic NU, a lower midline
anteriorly. A 24Fr resectoscope with a pointed coagulat- or ipsilateral Gibson incision is used to place the hand-
ing electrode is inserted per urethra alongside the port (Figure 16-5). The surgery is continued in a manner
ureteral catheter to resect a bladder cuff around the similar to the total laparoscopic procedure. The sur-
ureteral orifice. Anterior traction by the suprapubic geon’s hand is used for tactile sensation, retraction, and
grasper facilitates this dissection. blunt dissection. When the dissection reaches the level of
For renal and ureteric dissection trocar site placement the iliac vessels, the surgeon may choose to release the
varies by surgeon preference, but usually involves the use pneumoperitoneum and complete the distal ureterec-
of four or five trocars. A periumbilical trocar is placed tomy in an open technique through the hand-port.17
initially by the Veras needle or open technique, and This hand-port also allows easy extraction of the intact
pneumoperitoneum is established to 15 mm Hg. A 30- specimen from the abdomen. In comparison to the total
degree telescope is inserted through this port and the laparoscopic procedure, the hand-assisted approach
remaining trocars are placed under direct vision. decreases operating time without altering postoperative
Additional port sites may include ipsilateral lower quad- analgesic requirements or hospital stay.18

Figure 16-4 A, Laparoscopic NU can be performed via a retroperitoneal approach. Initially


the balloon is inflated in a cephalad direction towards the kidney to create a working space.
Finally the balloon is inflated along the anterior surface of the psoas muscle, displacing the
ureter anteriorly and creating a working space towards the iliac vessels. B, Right
laparoscopic NU showing patient position and three-port trocar approach.
Chapter 16 Management of Upper Urinary Tract Transitional Cell Carcinoma 291

The advantages of laparoscopic surgery compared to its


open counterpart include the reports of decreased analgesic
requirements, shorter hospital stay, and shorter convales-
cence.17,19-22 The efficacy of this procedure as a definitive
cancer therapy is more difficult to establish given the lim-
ited number of series reporting long-term outcomes.
McNeill et al.23 reported a series of 25 patients with upper
urinary tract TCC treated by laparoscopic NU. With a
mean follow-up of 33 months, there was no difference in
overall survival compared to those patients treated with
open surgery. In a comparable series, the crude and cancer-
specific survivals for patients treated with open versus
laparoscopic NU were similar after 24 months of follow-
up.21 This study did note a greater number of patients with
grade 4 tumors who had local retroperitoneal recurrence
following laparoscopic surgery, suggesting that the open
procedure may provide additional therapeutic benefit for
patients with high-grade tumors. For patients with upper
urinary tract TCC without local invasion, laparoscopic
techniques of NU are proving to be a safe and effective
alternative to open surgery with the advantage of shorter
hospital stays and quicker recovery (Figure 16-6).

OPEN NEPHRON-SPARING SURGERY


Figure 16-5 Trocar and abdominal incisions for right and left Open nephron-sparing surgery, including partial
hand-assisted laparoscopic NU. nephrectomy for renal pelvis tumors and partial

Laparoscopy Open p Value

Mean surgical time (mins):* 224.8 ± 64.3 280.2 ± 73.8 0.003


Cystoscopy (mins) 58.9 (35–120) —
Laparoscopy (mins) 173.5 ± 54.2 —

Mean blood loss (cc) 242 ± 267.4 696 ± 953.2 <0.0001

Mean intravenous fluids (l) 3.6 ± 1.0 5.4 ± 2.7 0.002

No. concomitant adrenalectomy (%) 28 (67) 4 (11) <0.001

Mean extraction incision (cm) 7.0 — —

Mean time to ambulation (days) 1.4 ± 1.0 2.5 ± 1.5 0.0003

Mean time to oral intake (days) 1.6 ± 1.2 3.2 ± 1.9 0.0004

Mean hospital stay (days) 2.3 ± 1.6 6.6 ± 1.9 <0.0001

Mean analgesics (mg morphine sulfate equivalent) 26 ± 24.3 228 ± 207.2 <0.0001

Mean duration Foley catheter (days) 7.6 ± 4.9 7.4 ± 3.6 0.97

Mean time to normal activities (weeks) 4.7 ± 9.4 8.2 ± 7.6† 0.002

Mean time of convalescence (weeks) 8.0 ± 10.1 14.1 ± 8.3† 0.007

*Does not include time (approximately 45 minutes) to reposition patient.


†Only available in 13 patients.

Figure 16-6 Intraoperative and postoperative data.


292 Part III Kidney and Ureter

ureterectomy for ureteral tumors, is a treatment option suture. Following adequate hemostasis, the remaining
for patients in whom preservation of renal function is renal pelvis is closed with 4-0 chromic sutures. The bed
important. Indications for conservative surgery include of dissection is cauterized with electrocautery or argon
solitary kidney, bilateral tumors, or baseline renal insuf- beam coagulator, and the parenchymal defect is closed
ficiency. The patient is placed in the flank-torque posi- with a series of 2-0 chromic mattress sutures. A closed
tion and prepped in a similar fashion to the open NU. suction drain is used to detect a urine leak postopera-
For partial nephrectomy, an incision is made superficial tively (Figure 16-7).
to the 10th or 11th rib, and the dissection proceeds in a Open segmental ureterectomy is a treatment option
retroperitoneal, extrapleural, or thoracoabdominal for invasive ureteral tumors when nephron-sparing sur-
manner. Gerota’s fascia is incised, and the kidney is gery is required or for local low-grade tumors, which are
mobilized completely. The renal vessels and ureter are too large for endoscopic management. Patients are
identified and tagged with vessel loops. Cooling the placed in a flank position for proximal and mid-ureteral
kidney in a crushed ice slurry can reduce ischemic tumors or supine for distal ureteral tumors. The type of
injury. Similarly, mannitol diuresis prior to vascular incision depends on the location of the tumor. For prox-
clamping can decrease parenchymal injury due to free imal and mid-ureteral tumors, a flank incision provides
radical formation. Upon clamping the renal vessels, the adequate exposure, while a Gibson or lower midline
margin of dissection is defined with electrocautery of abdominal incision is used for distal tumors. Upon iden-
the renal capsule. In dissection, the blunt end of a tifying the ureter and palpating the lesion, the ureter is
scalpel blade is used to excise the tumor, the involved clamped and ligated 2 cm proximal and distal to the
renal calyces, and surrounding margin of normal renal affected area. Frozen sections of each free end are per-
parenchyma. Areas of brisk bleeding are controlled with formed prior to ureteroureterostomy to ensure complete
hemostatic microclips or over-sewn with 4-0 Prolene resection of the diseased segment. The ureter is mobilized

Laparoscopy Open p Value

Number of points with complete followup greater than 1 month 35 30 —

Mean months followup (range) 11.1(1–27) 34.4 (2.5–87) <0.0001

No. bladder recurrence (on followup cystoscopy) (%) 8 (23) 11 (37) 0.42
Multifocal 5 8
Solitary 3 3

Mean months to recurrence (range) 6.6 (3–15) 10.5 (2–37)

No. subsequent bladder treatment


Transurethral bladder tumor resection 2 1
Transurethral bladder tumor resection, bacillus Calmette-Guerin 6 10
Cystectomy 0 2

Number of points with distant metastasis (%) 3 (8.6) 4 (13) 1.00


Lung 2 1
Liver 1 0
Bone 0 2
Lymph nodes 0 1

No. adjuvant systemic chemotherapy (%)* 4 (11) 4 (13) 1.00

No. deaths (%) 2 (6) 9 (30) 0.39

Mean months to death (range) 4 (2–6) 24 (2.5–67)

% Crude survival 97 94 0.59

% Ca specific survival 97 87 0.59

There were no retroperitoneal or port site/incisional recurrences with either technique.


*Comprises 3 to 4 cycles of methotrexate, vinblastine, doxorubicin and cisplatin therapy.

Figure 16-7 Follow-up data comparing laparoscopic NU to open NU.


Chapter 16 Management of Upper Urinary Tract Transitional Cell Carcinoma 293

to allow sufficient length to enable a tension-free repair mental ureterectomy, no difference in overall survival
(Figure 16-8). If necessary, the kidney or bladder can be was observed.26 The use of open nephron-sparing sur-
mobilized to provide additional ureteral length. gery is a viable treatment option for patients with low-
Ureteroureterostomy is performed by spatulating each grade, low-stage TCC of the upper urinary tract who
free end and anastomosing around a ureteral stent with require preservation of renal function.
4-0 absorbable suture. When performing a distal
ureterectomy, the repair proceeds with creation of an
ENDOSCOPIC MANAGEMENT
ureteroneocystostomy. This is performed through an
anterior cystotomy in a refluxing or nonrefluxing manner Endoscopic evaluation of the upper urinary tract by ante-
using 4-0 absorbable suture. The use of a psoas bladder grade or retrograde approach is useful for evaluation of
hitch may be required to lessen tension across the symptoms, diagnosis of pathology, and more recently,
anastomosis.24 Again, a closed suction drain is used to definitive therapy of upper tract malignancy. Diagnostic
monitor for a urine leak postoperatively (Figure 16-9). ureteroscopy is performed on patients with lateralizing
In the setting of a solitary kidney, bilateral tumor, or hematuria or abnormal cytology, a filling defect observed
baseline renal insufficiency, nephron-sparing surgery on radiographic evaluation and tumor at the ureteral ori-
may be employed for the treatment of upper urinary tract fice.2 It is also a critical tool employed for surveillance of
TCC. Ziegelbaum et al.25 reported the use of nephron- patients treated for upper tract malignancy with renal-
sparing surgery for patients with TCC of the renal pelvis. sparing methods. With the advances in optical technol-
The indication for conservative surgery was a solitary ogy, smaller and flexible ureteroscopes are able to access
kidney in the majority of patients, and the tumors were most areas of the upper urinary tract including the renal
primarily of low grade and low stage. In this series, 62% pelvis and calyces.
of patients remained disease-free up to 5 years postoper- Ureteroscopy provides access to the upper urinary
atively. For distal ureteral tumors, 83% disease-free sur- tract for evaluation and treatment of pathology while
vival has been observed following distal ureterectomy.24 maintaining a closed urinary system. The rigid uretero-
In a large series of 224 patients with low-stage upper uri- scope provides excellent visualization of the distal and
nary tract TCC treated by NU, nephrectomy, or seg- mid-ureter, while flexible ureteroscopes are used to

Figure 16-8 Surgical technique for uretero-ureterostomy. Ureteral spatulation of well-


vascularized ureteral ends and tension-free reapproximation with fine absorbable suture
material usually results is a good surgical result. Intraoperative frozen section analysis is
imperative to establish negative surgical margins.
294 Part III Kidney and Ureter

Figure 16-9 Distal ureterectomy and psoas hitch ureteral reimplantation is a well-recognized
surgical option for patients with distal ureteral tumors, when the proximal upper tract is
tumor free. Surgical principles once again include well-vascularized distal ureter and a
tension free anastomosis.

access the proximal ureter and renal pelvis. Access to the forceps or resectoscope. Bugbee electrocautery of the
lower pole calyx may be limited by the infundibulopelvic resected tissue bed fulgurates residual tumor and mini-
angle, which can exceed the deflection capability of flex- mizes bleeding. Alternatively, various laser fibers may be
ible ureteroscopes. Patients are placed in the lithotomy used to ablate tumors following tissue biopsy. The
position. Cystoscopy is performed initially for evaluation neodymium:yttrium-argon-garnet (Nd:YAG) and
of the bladder mucosa and to gain access to the ureter holmium:YAG laser sources are commonly used. The
using a guide wire. Although not always necessary with 200 or 365 μm flexible fibers pass easily through the
the use of small diameter instruments, atraumatic dila- working channel of both rigid and flexible ureteroscopes,
tion of the ureteral orifice and intramural ureter allows without significantly altering deflection. The holmium:
easy passage of ureteroscopes into the upper urinary YAG laser at 0.6 to 1 J and 10 Hz provides tissue ablation
tract. This is accomplished by inserting an inflatable bal- to a maximum of 5 mm.27 This is especially well suited
loon dilator over a guide wire across the ureteral orifice. for the thin-walled ureter. The Nd:YAG laser induces
The balloon is inflated to 14 cm H2O for 1 to 2 minutes. coagulation necrosis with subsequent sloughing of
Following dilation, a rigid ureteroscope is passed into the necrotic tumor. Dose characteristics of this laser source
ureter under direct vision, or the flexible scope is passed have been described following studies in the canine
over a guide wire using fluoroscopic guidance. High- ureter.28 Consistent transmural necrosis and ureteral
pressure irrigation or pulse-irrigators used at a maximum perforation were observed at doses of 20 W for 2 sec-
of 40 cm H2O allows good visualization without the risk onds. At higher power (60 W) and shorter duration
of ureteral perforation. (0.5 seconds), thermal injury extended into the muscle of
The entire intraluminal ureter and renal pelvis are the ureter without perforation and resulted in nonstric-
inspected on initial and subsequent evaluations. Several tured healing. In humans, settings of 20 W for 1 to 2
biopsy instruments are available for sampling areas of seconds are commonly used for tissue penetration of 5
pathology. These include brush-biopsy forceps, cold-cup to 6 mm.29 At the completion of treatment, a double-J
biopsy forceps, and resectoscopes, in which use is limited ureteral stent is placed and left in situ until the tumor bed
to larger rigid ureteroscopes. Complete resection of is completely healed, typically for 4 to 6 weeks (Figure
abnormal lesions can be accomplished with the biopsy 16-10).
Chapter 16 Management of Upper Urinary Tract Transitional Cell Carcinoma 295

Figure 16-11 Nephrostomy tract puncture sites. Peripheral


Figure 16-10 Therapeutic options for upper tract TCC also
calyceal tumor sites are best approached directly, while renal
include ureteroscopy and laser ablation, using an
pelvic tumors are usually easily approached via lower or
holmium:YAG laser. Careful follow-up, including ureteroscopy,
interpolar calyces.
brushings and cytology, is important to ensure a good
outcome.
tions for nephron-sparing surgery or who have a solitary,
Percutaneous antegrade access to the renal sinus is low-grade, and superficial tumor. Endoscopy is used only
employed to evaluate lesions inaccessible by flexible for biopsy and tissue diagnosis in patients who have sus-
ureteroscopy or to resect large tumors of the renal pelvis. pected invasive disease, high-grade or multifocal tumor.
Under fluoroscopic guidance, a percutaneous nephros- In a comparison of patients with TCC of the upper uri-
tomy tract is established. With placement of a 34F nary tract treated by percutaneous resection or open NU,
Amplatz sheath, the renal pelvis is accessed with a rigid there was no difference observed in the cancer-specific
or flexible nephroscope. Again, cold-cup biopsy forceps survival rates (Figure 16-12).31 Following tumor treat-
or a resectoscope is used to sample and resect the lesion. ment, a high rate of recurrence is observed with reported
As with ureteroscopy, electrocautery or laser ablation is rates ranging from 24% to 65%.1,3,30,32-35 The rate of
used to fulgurate the tumor bed following resection. recurrence has been directly associated with increasing
Insertion of a 24F reentry nephrostomy tube allows easy grade of tumor in a several studies.1,2 The development
access to the pelvis on a second-look procedure. of subsequent bladder tumors is also a concern with
Nephroscopic examination of the renal pelvis with ran- reported rates of 30% to 43% of patients, similar to other
dom biopsies should always be performed within 4 weeks renal-sparing procedures.1,32 With a high rate of tumor
of tumor resection. The nephrostomy tube is removed recurrence, a strict surveillance protocol is employed for
after no suspicious areas are noted on nephroscopic all patients treated with nephron-sparing surgery. This
examination and all random biopsies are negative.30 includes cystoscopy with retrograde pyelography and
Percutaneous management is best reserved for tumors ureteroscopy every 3 months until tumor-free. This is fol-
with a low risk of recurrence. These include solitary, pap- lowed by cystoscopy and retrograde pyelography every 3
illary tumors that are less than 2 cm, low-grade, and low- months and ureteroscopy every 6 months.1 NU remains a
stage.30 Other tumors are most appropriately managed treatment option for those patients with multiple recur-
by ureteroscopy if possible (Figure 16-11). rences. Overall, the preservation of renal units by complete
Endoscopic management of upper urinary tract TCC endoscopic management ranges from 73% to 86% when
is reserved for patients with previously mentioned indica- primary tumors are low-grade and noninvasive.30,32,36
296 Part III Kidney and Ureter

Figure 16-12 Endoscopic technique for percutaneous removal of renal pelvic


tumors. A, The bulk of the tumor is removed with grasping forceps. Following
that the remaining tumor is resected using a standard resectoscope and
cutting loop. B, Medial tumors pose increased risk because of renal
vasculature and are therefore best managed by debulking biopsy followed by
holmium:YAG laser ablation. C, Finally, ureteroscopy and laser ablation can
be achieved with the flexible cystoscopy and a standard lower pole
approach. D, Intraoperative and postoperative data comparing laparoscopic
NU to open NU.
Chapter 16 Management of Upper Urinary Tract Transitional Cell Carcinoma 297

References nephroureterectomy for transitional cell carcinoma of the


upper urinary tract. Urol 2001; 58:930–934.
1. Chen GL, Bagley DH: Ureteroscopic management of 18. Landman J, Lev RY, Bhayani S, et al: Comparison of hand
upper tract transitional cell carcinoma in patients with assisted and standard laparoscopic radical
normal contralateral kidneys. J Urol 2000; 164:1173–1176. nephroureterectomy for the management of localized
2. Huffman JL: Ureteroscopic management of transitional transitional cell carcinoma. J Urol 2002; 167:2387–2391.
cell carcinoma of the upper urinary tract. Urol Clin 19. Goel A, Hemal AK, Gupta NP: Retroperitoneal
North Am 1988; 15:419–424. laparoscopic radical nephrectomy and
3. Palou J, Salvador J, Millan F, et al: Management of nephroureterectomy and comparison with open surgery.
superficial transitional cell carcinoma in the intramural W J Urol 2002; 20:219–223.
ureter: what to do? J Urol 2000; 163:744. 20. Seifman BD, Montie JE, Wolf JS Jr: Prospective
4. Hall MC, Womack S, Sagalowsky AI: Prognostic factors, comparison between hand-assisted laparoscopic and open
recurrence, and survival in transitional cell carcinoma surgical nephroureterectomy for urothelial cell carcinoma.
of the upper urinary tract: a 30-year experience in 252 Urol 2001; 57:133.
patients. Urology 1998; 52:594–601. 21. Shalhav AL, Dunn MD, Portis AJ: Laparoscopic
5. Racioppi M, D’Addessi A, Alcini A, Destito A, Alcinin E: nephroureterectomy for upper tract transitional cell
Clinical review of 100 consecutive surgically treated cancer: the Washington University experience. J Urol
patients with upper urinary tract transitional tumors. 2000; 163:1100–1104.
Br J Urol 1997; 80:707–711. 22. Keeley FX Jr, Tolley DA: Laparoscopic
6. Kakizoe T, Fujita J, Murase T, Matsumoto K, Kishi K: nephroureterectomy: making management of upper-tract
Transitional cell carcinoma of the bladder in patients transitional-cell carcinoma entirely minimally invasive.
with renal pelvic and ureteral cancer. J Urol 1980; 124:17–19. J Endourol 1998; 12:139–141.
7. Strong DW, Pearse HD, Tank ES Jr, Hodges CV: The 23. McNeill SA, Chrisofos M, Tolley DA: The long-term
ureteral stump after nephroureterectomy. J Urol 1976; outcome after laparoscopic nephroureterectomy: a
115:654–655. comparison with open nephrureterectomy. Br J Urol Int
8. Miyake H, Hara I, Gohji K, Arakawa S, Kamidono S: The 2000; 86:619–623.
significance of lymphadenectomy in transitional cell 24. Johnson DE, Babaian RJ: Conservative surgical
carcinoma of the upper urinary tract. Br J Urol 1998; management for noninvasive distal ureteral carcinoma.
82:494–498. Urol 1979; 13:365–367.
9. Komatsu H, Tanabe N, Kubodera S, Maezawa H, Ueno 25. Ziegelbaum M, Novick AC, Streem SB, et al:
A: The role of lymphadenectomy in the treatment of Conservative surgery for transitional cell carcinoma of the
transitional cell carcinoma of the upper urinary tract. renal pelvis. J Urol 1987; 138:1146–1149.
J Urol 1997; 157:1622–1624. 26. Murphy DM, Zincke H, Furlow WL: Primary grade 1
10. Murphy DM, Zincke H, Furlow WL: Management of transitional cell carcinoma of the renal pelvis and ureter.
high-grade transitional cell cancer of the upper urinary J Urol 1980; 123:629–631.
tract. J Urol 1981; 125:25–29. 27. Bagley D, Erhard M: Use of the holmium laser in the
11. Johansson S, Wahlquist L: A prognostic study of upper urinary tract. Tech Urol 1995; 1:25–30.
urothelial renal pelvic tumors: a comparison between the 28. Smith JA Jr, Lee RG, Dixon JA: Tissue effects of
prognosis of patients treated with intrafascial neodymium:YAG laser photoradiation of canine ureters.
nephrectomy and perifascial nephroureterectomy. Cancer J Surg Oncol 1984; 27:168–171.
1979; 43:2525–2531. 29. Schmeller NT, Hofstetter AG: Laser treatment of ureteral
12. Clayman RV, Kavoussi LR, Figenshau RS, Chandhoke PS, tumors. J Urol 1989; 141:840–843.
Albala DM: Laparoscopic nephroureterectomy: initial 30. Orihuela E, Smith AD: Percutaneous treatment of
clinical case report. J Laparoendosc Surg 1991; transitional cell carcinoma of the upper urinary tract. Urol
1:343–349. Clin North Am 1988; 15:425–431.
13. Shalhav AL, Elbahnasy AM, McDougall EM, Clayman 31. Lee BR, Jabbour ME, Marshall FF, Smith AD, Jarrett
RV: Laparoscopic nephroureterectomy for upper tract TW: 13-year survival comparison of percutaneous
transitional-cell cancer: technical aspects. J Endourol and open nephroureterectomy approaches for
1998; 12:345–353. management of transitional cell carcinoma of renal
14. Gill IS, Soble JJ, Miller SD, Sung GT: A novel technique collecting system: equivalent outcomes. J Endourol 1999;
for management of the en bloc bladder cuff and distal 13:289–294.
ureter during laparoscopic nephroureterectomy. J Urol 32. Elliott DS, Segura JW, Lightner D, Patterson DE, Blute
1999; 161:430–434. ML: Is nephroureterectomy necessary in all cases of upper
15. Keeley FX Jr, Sharma NK, Tolley DA: Hand-assisted tract transitional cell carcinoma? Long-term results of
laparoscopic nephroureterectomy. Br J Urol Int 1999; conservative endourologic management of upper tract
83:504–505. transitional cell carcinoma in individuals with a normal
16. Gill IS, Sung GT, Hobart MG, et al: J Urol. 2000; 164(5): contralateral kidney. Urol 2001; 58:174–178.
1513–1522. 33. Bagley DH: Ureteroscopic laser treatment of upper
17. Chen J, Chueh SC, Hsu WT, Lai MK, Chen SC: urinary tract tumors. J Clin Las Med Surg 1998;
Modified approach of hand-assisted laparoscopic 16:55–59.
298 Part III Kidney and Ureter

34. Martinez-Pineiro JA, Matres JG, Martinez-Pineiro L. treated upper urinary tract transitional cell carcinoma.
Endourological treatment of upper tract urothelial Urology 1996; 47:819–825.
carcinomas: analysis of a series of 59 tumors. J Urol 1996; 36. Okada H, Eto H, Hara I, et al: Percutaneous treatment of
156:377–385. transitional cell carcinoma of the upper urinary tract. Int
35. Elliott DS, Blute ML, Patterson DE, Bergstralh EJ, J Urol 1997; 4:130–133.
Segura JW: Long-term follow-up of endoscopically
C H A P T E R

17 Diagnosis and Staging of Bladder


Cancer
Michael J. Droller, MD

The standard approach in diagnosing and staging bladder new cases are diagnosed annually and over 12,000 indi-
cancer initially incorporates cystoscopic examination of the viduals die of bladder cancer each year.5
bladder and examination of urinary cytology to determine Efforts to diagnose the possible presence of a urothe-
the presence of malignancy. Once the presence of a lesion lial cancer are generally initiated by occurrence of either
is established, resection and pathologic evaluation of the gross or microscopic hematuria.6 The degree of hema-
tumor specimen (and occasionally of mucosal biopsies) are turia has not been observed to correlate with either size,
needed to document disease and characterize grade and grade, or stage of a particular tumor. On the other hand,
stage (extension of the neoplasm into the bladder wall). the presence of a bladder tumor may be more likely when
Preliminary information on prognosis is inferred by the gross hematuria, rather than microscopic hematuria, is
“snapshot” obtained through histopathologic examination the inciting event.
and staging of the resected specimen (Figure 17-1).1,2 Irritative symptoms suggestive of a cystitis with a
However, a deeper understanding of the implications of a sense of increased urgency and urinary frequency may
particular tumor is based on an appreciation of where this occasionally be reported by a patient with bladder cancer,
snapshot fits into the natural history and pathogenesis of most often in the setting of carcinoma in situ (see later).
the specific diathesis represented by this lesion in the con- The location of this entity at the trigone of the bladder,
text of different developmental pathways of bladder cancer the bladder neck, or in the urethra should be suspected
(Figure 17-2).3 Further, these are ultimately determined by when irritative symptoms occur in the setting of urothe-
genetic and molecular changes that correspond to intrinsic lial cancer.
biologic behaviors of specific tumors and the potential for Most often, however, hematuria is not accompanied
recurrence or progression (Figure 17-3).4 Characterization by other symptoms. It is therefore referred to as “silent.”
of a particular tumor’s pathogenesis may thus be as impor- The occurrence of hematuria requires evaluation of the
tant clinically as the pathologic stage at the time of its entire urinary tract since its source may not be exclusive
resection in the individualization of treatment approaches, to the bladder. Nor is the occurrence of hematuria nec-
assessment of results of various “standardized” treatments, essarily indicative of the presence of a malignancy.7
and design of new therapeutic approaches. Therefore, general imaging studies are performed to
This chapter discusses standard methods used to diag- evaluate the kidney cortex, the renal pelvis, the ureters,
nose and stage bladder cancer. It will also review con- and the bladder to search for malignancy or to identify
cepts that are being used increasingly to assess the another possible etiology. Although it has become
intrinsic biologic potential of various forms of bladder increasingly common to perform sonography initially in
cancer and to enhance our understanding of molecular such an evaluation, the efficacy of ultrasound in detecting
changes that may determine their pathogenesis. upper tract urothelial lesions depends on the extent of
the abnormality, as well as the experience and thorough-
ness of the radiologist performing the procedure.8 The
INITIAL DIAGNOSIS OF BLADDER CANCER
major value of sonography in this setting lies in its ability
Bladder cancer is the fourth most prevalent noncuta- to detect a renal mass and to determine whether it is solid
neous malignancy in the United States.5 Over 54,000 or cystic. Additionally, it may be useful in identification

301
302 Part IV Bladder

Figure 17-1 Staging systems for bladder cancer are based on correlations between the
layer to which a particular cancer has penetrated the bladder wall and the prognosis of that
type of cancer. The original staging system (developed by Jewett HJ, Strong GH: J Urol
1946; 55:366) suggested distinctions between superficial and muscle-invasive disease. The
superficial classification was subsequently divided into tumors confined to the mucosa
(stage Ta) and tumors that penetrate the lamina propria (stage T1). Carcinoma in situ (stage
Tis) is a superficial disease composed of neoplastic cells that replace the normal urothelium
or undermine the normal urothelium and extend along the plane of the urothelium. Deep
bladder tumors penetrate the muscularis propria either superficially (stage T2a) or deeply
(stage T2b) or penetrate through the muscularis propria into the perivesical soft tissue either
microscopically (stage T3a) or extensively (stage T3b). These distinctions are a change in the
World Health Organization (WHO) classification system, which now combines all muscle-
invasive tumors into one category (stage T2) rather than maintaining the separation that
formerly had characterized the staging system (stage T2 for superficial invasion and stage
T3a for deep muscle invasion). The involvement of lymph nodes is designated by the N
category and involvement of adjacent structures is categorized as stage T4. This format
implies a simple pattern of sequential development according to which early cancers appear
as lower stages, and then progress to higher stages in sequence. However, this is not
necessarily what characterizes the different forms of bladder cancer seen clinically. Rather, a
variety of pathways that do not necessarily occur in sequence but are possibly interrelated
may more accurately reflect the biology of the different forms of bladder cancer.

of a lesion within the renal pelvis or within the course of any areas that appear abnormal or may not have been
the ureter, a dilated possibly obstructed ureter, or an adequately visualized on IVP.
intraluminal-filling defect of the bladder itself (largely Computed tomography (CT)11-13 and magnetic reso-
for tumors larger than 1 cm).9 nance imaging (MRI)14-17 can also be used to identify
Intravenous pyelography (IVP) provides definition of upper tract lesions, the presence of dilated ureter(s), and
the contours of the upper tracts better than that obtained masses in the bladder. To this point, neither has been
with sonography. However, pyelography may not reliably used as initial means of evaluating the cause of hematuria
permit identification of small lesions of the renal or as a first-line method in diagnosing bladder cancer.
parenchyma or renal pelvis or of bladder lesions <1 cm in However, recent interest has focused on the use of CT
diameter.10 Imaging of the bladder in various positions as scanning in providing 3D imaging of the urinary tract.
it fills with contrast may allow visualization of one or Spiral CT scanning provides not only the quality of
multiple bladder tumors, and full distention of the blad- images achievable to visualize the kidney, ureter, and
der may be of particular usefulness to the radiologist in bladder on intravenous pyelogram but also provides
imaging a tumor on ultrasound. cross-sectional imaging and 3D reconstruction, which
Retrograde pyelography may be useful in a patient can be used as an advantage in possibly staging a lesion if
with renal compromise or with contrast allergy in defin- one is uncovered. The use of air insufflation to facilitate
ing the anatomy of the upper tracts and in characterizing imaging of lesions in the bladder has been suggested to
Chapter 17 Diagnosis and Staging of Bladder Cancer 305

the upper tracts or the bladder and penetrated through to guishing between tumors that are of low malignant
the soft tissues. potential and those that are of high malignant potential
MRI enhancement has been attempted by the use of (see later). The substantial overlap in labeling, however,
gadolinium, T-weighted spin modalities, and use of a may currently prevent full exploitation of this possibility.
number of other agents in imaging a primary tumor and At the same time, use of these assays in conjunction with
assessing its extent and penetration. MRI (and positron urinary cytology may permit the more reliable detection
emission tomography [PET] scanning) has been used in of urothelial cancers of all grades.
identifying whether enlarged lymph nodes represent Both the ImmunoCyt and the Vysion assays have a
metastatic disease.15-21 In such instances, fine needle lower specificity than does urinary cytology. In this, uri-
aspiration has been used to confirm whether or not nary cytology remains the gold standard.31 It is therefore
metastatic disease is present (see later).22,23 the balance between enhanced sensitivity of these assays
Former teaching held that when an upper tract source in detecting disease of low malignant potential versus the
of gross hematuria was suspected, upper tract imaging increased sensitivity and specificity of urinary cytology in
studies should be followed immediately by cystoscopy to detecting disease of high malignant potential that must
facilitate localization of disease by visualizing blood ema- be considered in using these assays appropriately.
nating from a ureteral orifice. However, as upper tract During the past decade, much interest has been focused
lesions can now be visualized with greater reliability on the use of assays that detect tumor “markers” in the
using current imaging techniques, the immediate need urine. One prominently advocated was an assay for blad-
for cystoscopy has become less pressing. der tumor “antigen” (BTA), a complement-related protein
On initial imaging evaluation of a patient with hema- released from the lamina propria in the presence of tumor
turia, it may be of value to obtain urine for cytologic and inflammation.32 Unfortunately, the latter condition
assessment. Although not particularly efficacious in the severely compromised this assay’s specificity.33 Moreover,
detection of low-moderate-grade disease (with sensitivi- the BTA assay also had inadequate sensitivity in detecting
ties ranging between 10% and 50%), urinary cytology is higher-grade disease.32 This led to the development of a
useful in diagnosing urothelial cancers that are of high point-of-care assay (BTA stat) and then a double-antibody
grade.24,25 Therefore, if a urinary cytology is positive, sandwich assay (BTA trac) in assessing a complement-
and upper tract imaging is negative, it may be expedient related factor released into the urine in association with
to proceed directly to cystoscopy under anesthesia so that the presence of a urothelial malignancy.33 However, speci-
multiple biopsies (see later) and resection of visible ficity of these assays was also affected by inflammation,
lesions can be performed. The same approach may be compromising their usefulness.
reasonable if a bladder tumor has been visualized on A urinary assay for nuclear matrix protein (NMP-22)
imaging studies. Further assessment with barbotage irri- had an improved sensitivity for detecting low-moderate-
gation specimens may produce clusters of cells that sug- grade disease and its specificity was better than that
gest low-moderate-grade malignancy, but this may also obtained with either of the BTA tests.34 Its validation in
correspond to instrumentation artifact. Therefore, barb- the detection of high-grade disease and its use in the set-
otage has generally been used more specifically to provide ting of inflammatory conditions, however, appeared simi-
increased numbers of cells for analysis by flow cytometry, larly to limit its use. Other assays (microsatellites,
which may detect abnormalities in individual cells when a telomerase, hyaluronic acid/hyaluronidase, DD-23, fibrin
urinary cytology has been interpreted as negative.26-28 gradation products [FDP], and many others) have shown
Two products have recently been introduced to promise in preliminary studies.35 However, validation
enhance detection of low-moderate grade urothelial can- regarding their reliability, their consistency, and their role
cers and to complement cytology in diagnosis of high- in the diagnosis and assessment of urothelial cancer
grade disease by employing fluorescent enhancement. remains to be obtained. An evolving consensus is that their
The ImmunoCyt assay employs fluorescent-labeled anti- use may ultimately be found in monitoring for tumor
bodies to tumor antigens, obtaining a sensitivity of 70% recurrence in patients with a history of bladder cancer, and
to 80% in detecting low-moderate-grade tumors (versus particularly in the setting of tumors that have low malig-
that of urinary cytology with reports of sensitivity as low nant potential, so that the interval between sequential
as 10% to 15% for grade 1 tumors and 50% to 60% for invasive procedures for surveillance can be lengthened.
moderate-grade disease).29 The second assay, Vysion, After preliminary imaging of the upper tracts and
incorporates fluorescent in situ hybridization (FISH) bladder and an analysis of urinary cytology, cystoscopic
technology in identifying chromosomal abnormalities in examination is required to confirm the presence of
voided or barbotage urinary specimens.30 The antibodies malignancy, characterize its architecture, determine the
used in each assay are directed against molecules that multiplicity of disease, and determine whether there is
may identify cancers with specific biologic pathways. diffuse mucosal involvement. Each helps assess the
Both assays may, therefore, offer the possibility of distin- nature of the cancer diathesis.
306 Part IV Bladder

The sine qua non of diagnosis in urothelial cancer is A “solid” or nodular (as distinct from papillary) tumor
transurethral resection of the tumor and pathologic eval- appearance implies a more deeply infiltrative diathe-
uation of the surgical specimen. In addition, biopsies may sis.39,40 In such instances, the superficial (or intraluminal)
be taken in the setting of positive urinary cytology ran- portion of the solid tumor should be resected flush with
domly from several sites or directed on visualization of the plane of the mucosa. Deeper resection to include the
abnormal areas of the bladder mucosa. Directed biopsies muscularis propria should then be done to determine the
typically include areas adjacent to the presenting lesion, depth of infiltration and possibly distinguish between
areas of mucosal erythema, and areas at the bladder neck superficial and deep muscle invasion. Some have advocated
and within the prostatic urethra. Random biopsies of even deeper resection to the level of the perivesical fat.
endoscopically normal areas of the urothelium are no However, extensiveness of disease can generally be deter-
longer taken routinely as part of the standard diagnostic mined without an aggressive resection such as this, and
approach in the evaluation of a bladder cancer diathesis risk of perforation can be avoided.
when a preliminary cytology has been negative. Several Bimanual examination of the bladder under anesthesia
studies have shown that such random biopsies are not before and after transurethral resection may provide addi-
likely to be informative and that the characterization of tional information on the extent of the tumor invasion. This
the tumor diathesis by pathologic evaluation of the is most informative when there is a large nodular or sessile
resected specimen is generally sufficient for decisions tumor that may have extended through the bladder wall, or
regarding subsequent treatment.36 Thus, random biop- possibly involved adjacent structures. Inability to palpate a
sies are now generally only obtained when there is no tumor mass prior to resection suggests that the tumor is
endoscopically visible lesion, when upper tracts and ure- possibly less extensively invasive. Palpation of a mass that
thra have no radiologic or visual abnormality, and urinary is mobile suggests the presence of a nodular, infiltrative can-
cytology is persistently positive. In these instances, biop- cer that does not involve adjacent structures. Palpation of a
sies are needed to identify the source of the positive cells. mass that is fixed suggests that the cancer may have involved
The configuration of the tumor as characterized adjacent structures or the pelvic sidewalls. Palpation follow-
endoscopically may suggest whether the tumor is super- ing resection that suggests less fixation may indicate a less
ficially or more deeply invasive. A papillary lesion is more extensive involvement of adjacent structures than had been
likely to be “superficial.” However, this alone does not surmised. However, any degree of palpability is generally an
permit a distinction to be made between those tumors ominous finding. In attempting to assess the extent of inva-
that are mucosally confined (stage Ta) and those that sive disease, bimanual palpation is highly subjective. Its
infiltrate the lamina propria (stage T1) (see later).37 greatest value may be in cases of large tumors that are exten-
Occasionally, mucosally confined disease may be identi- sively infiltrative, since these are more likely to be palpable
fied by being able to remove the lesion by “scraping” it and generally indicate an adverse prognosis, especially if
with a cold loop. This can often be done with multiple they are palpable after resection.
very small papillary lesions, in which a well-developed
fibrovascular core has not formed. Generally, however,
STAGING OF BLADDER CANCER
electroresection is necessary to excise a papillary lesion.
It is important that muscle fibers be visualized and sam- Transurethral ultrasound,41 pelvic CT,42 and pelvic or
pled for accurate staging, particularly when a papillary endorectal (endovaginal) MRI43 imaging can be used to
tumor may have invaded the lamina propria. Several assess tumor extent prior to transurethral resection.
studies have observed an at least 20% involvement of the Transurethral ultrasound examination has been reported
muscularis propria on repeat resection when initial resec- to have a sensitivity of 90% and a specificity of 76% in
tion suggested only lamina propria invasion and no mus- determining whether a tumor is muscle-invasive.44 One
cle was present for accurate analysis.38 A vigorous report described that 24% of tumors staged as T1 by
resection, however, with its risk for penetration of the transurethral ultrasound had been overstaged and that
bladder wall, is generally not necessary to obtain ade- 10% of tumors staged as T2 had been understaged.
quate levels of tissue for staging. Correspondingly, 29% of stage T3b tumors (deep muscle
Papillary tumors should initially be resected flush with invasion) were understaged and 5% of stage T3a tumors
the bladder mucosa. Somewhat deeper resection can then were overstaged by this modality.
be done to permit separate evaluation of the lamina pro- The sensitivity of CT scanning in detecting extravesi-
pria and the superficial level of the muscularis propria. cal extension of a bladder cancer has ranged between 60%
Care should be taken not to resect too deeply to avoid and 96%.12,14,42,45 Its specificity has ranged between
perforation of the bladder wall. In the setting of papillary 66% and 93%. Cumulative sensitivities and specificities
disease the bladder wall may be quite thin and electrore- have been calculated at 83% and 82%, respectively,
section need not be carried more deeply than is required implying that extravesical extension remains undetected
for accurate staging of the superficial bladder layers. in 17% of instances and is over-diagnosed in 18% of
Chapter 17 Diagnosis and Staging of Bladder Cancer 307

instances. CT scanning has had limited success in distin- cancer, studies have described a sensitivity of 86% to
guishing between mucosally confined (stage Ta) and lam- 100% and a specificity of 63% to 100%. The diagnosis of
ina propria-invasive tumors (stage T1) and between lymph node involvement or extension outside of the
superficial and muscle-invasive (stage T2) diseases. It has bladder may also be achieved using PET scanning with
been more useful in distinguishing between lesions that FDG. However, studies reported to this date have
are only minimally invasive of the extravesicular soft tissues involved only small numbers of patients.
(stage T3a) and those that have involved these tissues exten- Other studies have used LCM for diagnosis of bladder
sively (stage T3b) or adjacent organs (stage T4). However, cancer and have employed calculations of specific uptake
overstaging has occurred in two-thirds of patients with values in attempts to enhance discrimination between
superficial disease, while understaging has been found in muscle invasion and extravesical extension of disease.21
30% of patients with muscle-invasive tumors. Uptake of LCM by normal bladder tissue is low.
MRI has several features that theoretically would Therefore, staging of disease is limited. Some have sug-
make it more accurate than CT scanning in the staging gested that PET scanning with LCM may be useful in
of bladder cancer. Its major advantage lies in images that monitoring for responses to neoadjuvant chemotherapy
can be obtained in multiple planes and in the demonstra- or assessing regression of metastatic foci by evaluating
tion of perivesical fat planes and boundaries of the persistence of uptake on PET scanning after treatment.
prostate and seminal vesicles.15-17,42,43 T1-weighted Further study to assess these possibilities and definition
images of the primary tumor can be contrasted with low- of the costs involved in the use of this technique will
signal intensity images of urine and high-signal intensity determine its future usefulness.
images of perivesical fat. T2-weighted images can be The staging of bladder cancer by each of these imag-
used to assess disruption of the muscle wall and invasion ing modalities is limited by their inability to discern
of other organs. The sensitivity for MRI in identifying microscopic extension of disease and the relative
extravesical tumor extension ranges between 60% and dependence of each on disruption of normal “radi-
100%, with a cumulative average of 73%. Specificity ographic” tissue planes in identifying the extension of
ranges between 60% and 100% with a cumulative aver- the cancer. The former may lead to understaging of dis-
age of 84%. Recent reports have described improved ease, since extension of the cancer, even if only micro-
sensitivity and specificity in detecting muscle invasion scopic, may be associated with a greater likelihood of
(96.2% and 83.3%, respectively) with the use of gadolin- metastasis. The latter can lead to overstaging, since
ium-DTPA enhanced MRI for tumor staging.43 In addi- images suggesting disruption of the muscularis can be
tion, improved accuracy has been obtained with newer created by prior resection, inflammatory processes, or
MRI techniques. Improved staging accuracy with sub- other therapeutic interventions.
millimeter pixel MRI, fast dynamic first pass MRI, fast The critical element in ultimate characterization of a
low-angle shot images, oblique contrast-enhanced T1 bladder cancer is its histopathologic appearance. The
weight, and rapid gradient echo sequence MRI have been extent to which the cancer penetrates the bladder micro-
used in attempts to improve the accuracy of staging both scopically,46 the histologic appearance of the tumor cells
in terms of identifying depth of penetration into and and the architectural configuration of those cells (papil-
through the bladder wall and identifying abnormalities in lary versus nodular),40,47 whether or not bladder lymphat-
the pelvic lymph nodes.15-18 ics or vasculature are involved by the cancer,40 and the
PET scanning in bladder cancer has not been studied nature of the urothelium adjacent to and at sites distant
extensively, and its role in staging is therefore unclear.19- from the presenting lesion(s) (dysplasia or carcinoma in
21 The radionuclides used are 18F-2-fluoro-2-deoxy-D- situ)48,49 are important in considering a tumor’s prognosis.
glucose (FDG) and L-(methyl)-11C-methionine (LCM). Currently, staging of bladder cancer is based on the
Both are primarily glucose derivatives and are used for depth to which the cancer has penetrated the bladder
PET scanning that exploits the hypermetabolic state of wall.1,50 Although transurethral resection may be per-
malignant tissues when compared with normal tissues. formed so as to provide specimens that permit determina-
FDG is taken up by normal and malignant cells, and the tion of the extent of involvement by the cancer of different
glucose is phosphorylated for passage through the levels, inaccuracies generally prevail and tumors are often
Embden-Meyerhof pathway.20 However, phosphorylated understaged.51 More definitive assessment may only be
FDG cannot leave the cell and cannot be metabolized. possible by analyzing a full thickness of the bladder wall
This allows its detection by PET scanning. obtained by partial cystectomy or total cystectomy speci-
Since FDG is excreted in the urine, the bladder must mens. This is clearly impractical in the majority of
be irrigated continuously to permit delineation of the patients. Several reports have described transabdominal
bladder wall from the urine. Diagnosis of a primary blad- CT-guided needle biopsies to provide full-thickness blad-
der tumor and its separation from possible urinary arti- der wall specimens to evaluate the depth of cancer infiltra-
fact, however, may be difficult. In diagnosis of a bladder tion.52 This technique has not gained general usage.
308 Part IV Bladder

Early attempts to characterize the prognosis of various the lamina propria (stage T1). The classification for mus-
forms of bladder cancer were based more on an analysis of cle invasive bladder tumors was unified in a single category
their cellular appearance than their depth of penetration. of stage T2 disease (with subcategorization into stage T2a
Broders53 described different grades of disease and corre- representing superficial invasion into muscularis propria
lated those that were poorly differentiated with a more omi- and stage T2b representing invasion into the deeper com-
nous prognosis and those that were well differentiated with ponent of the muscularis propria) and this was distin-
a more benign prognosis. Although Aschner54 subsequently guished from stage T3 disease that represented either
correlated an increasing depth to which a tumor had pene- microscopic penetration into the perivesical soft tissues
trated the bladder wall with a progressively worse progno- (stage T3a) or disease that had extensively infiltrated the
sis, this system did not obtain widespread currency.54 soft tissues surrounding the bladder (stage T3b). These
It was not until the mid-1940s that Jewett and Strong55 staging systems are identical to those that have been pre-
proposed a classification of bladder cancer based on the sented in the current American Joint Committee on
depth of involvement of the bladder wall in association Cancer’s (AJCC) staging system for 2002.
with the “curability” of the tumor by cystectomy. Initial In addition, various modifiers that characterize the
observations were based on an autopsy series, in which tumor diathesis can be included in describing the stage and
those tumors that had penetrated deeply into the bladder character of disease that is present at any particular time.
wall were found in retrospect to have been “incurable.” In For example, multiplicity of disease as this may be indica-
a subsequent study of cystectomy specimens, Jewett and tive of the increased risk for recurrence can be indicated by
Lewis56 proposed that tumors that had invaded only super- stating the number of tumors in parentheses following the
ficially were curable by cystectomy whereas those that had pathologic stage of disease. The presence of lymphatic
invaded the bladder wall more deeply were not. invasion or vascular invasion in the wall of the bladder,
Subsequent refinements in staging have been based on while not officially recognized as a component of the stag-
more precise correlations between the depths of invasion of ing system, can be indicated as a modifier of either “l” or
a tumor through the different layers of the bladder wall in “v” in parentheses following the indication of pathologic
association with prognosis, giving rise to the TNM classi- stage. Ultimately, incorporation of molecular markers may
fication.1,50 This consists of characterization of depth of be used to modify staging based on depth of invasion, and
penetration of the tumor (T), involvement of pelvic lymph grade may also be useful in determining risk of progres-
nodes (N), and whether or not metastases were present sion. It is important to recognize in each of these that a
(M). The objectives of such a system are to suggest prog- particular stage provides only a snapshot as part of the
nosis, to form a basis for selecting treatment, to provide a pathogenesis of disease that may evolve with time. A more
standard for evaluating treatment results, and to facilitate detailed discussion of a schema indicative of the different
the exchange of information through standardization of the pathways of tumor development that represents the
classification of the disease being treated. pathogenesis of disease in bladder cancer is provided in
The TNM system consists of a pretreatment clinical detail at the end of this chapter.
(c) classification and a postsurgical histopathologic (p) clas-
sification. The pretreatment clinical classification is
Biologic Activity of Different Stages of Bladder
based on imaging studies and impressions obtained both
Cancers
by endoscopic visualization of the cancer and bimanual
examination. The histopathologic classification is based Mucosally confined tumors (stage Ta), comprising 50%
on specimens obtained by transurethral resection. of all urothelial cancers at initial diagnosis (70% of all
Differences between these two classifications within indi- superficial cancers that are diagnosed as such in 70% to
vidual categories may be substantial. Moreover, the latter 75% of all urothelial cancers initially), have the best
may often be in substantial error (generally by under- prognosis.61,62 A 70% recurrence rate has been associated
staging) when compared to assessments determined by with larger and multiple tumors at diagnosis. This could
full thickness bladder wall cystectomy specimens.57-59 represent inadequate resection, increased likelihood of
Although correlating well with stage and biologic poten- tumor cell implantation, or sites that were not endoscop-
tial of disease, tumor grade has not been included in ically visible initially but that then “recurred” (becoming
these classification systems. apparent clinically).
The most recent TNM classification for bladder Most mucosally confined tumors found likely to
tumors (1998)50 maintained the classifications for superfi- progress have been of high grade. These have comprised
cial tumors that were described in the 1992 TNM classifi- only 2% to 4% of all stage Ta cancers.62 Additionally,
cation subsequently reviewed at the 4th International they have often been observed as a more diffuse
Consensus Meeting on bladder cancer in 1993.60 These micropapillary diathesis,63 or to have been accompanied
classifications included carcinoma in situ (TIS), mucosally by flat carcinoma in situ (see later) adjacent to or else-
confined cancers (stage Ta), and cancers that had infiltrated where in the bladder.48
Chapter 17 Diagnosis and Staging of Bladder Cancer 309

“Superficial” cancers that have invaded the lamina It is as yet unclear whether there are different forms of
propria (stage T1) comprise 20% of all urothelial cancers carcinoma in situ, and whether the variability and the
on initial presentation (30% of all superficial cancers manner in which carcinoma in situ presents may affect
[70%] as staged initially).61 As with mucosally confined the heterogeneity of response of superficial disease
(stage Ta) tumors, these have a 70% rate of recurrence. (whether mucosally confined or invasive of the lamina
Unlike the mucosally confined diathesis, however, stage propria) to different types of treatments.3,77 One form of
T1 disease has a 20% to 30% possibility of progres- carcinoma in situ is characterized by highly abnormal
sion.64,65 The high-grade tumors in this category (com- cells that involve the bladder mucosa diffusely and may
prising 50% of all T1 cancers) appear most likely to actually undermine the normal mucosa in spreading in
progress.62 This is seen in 50% of these cancers, or in pagetoid fashion to cover broad areas of the bladder wall,
25% of the entire group.66,67 High-grade lamina propria possibly extending as well into the prostatic urethra and
invasive tumors are often associated with urothelial the lower ureters.78-80 Patients often present with symp-
abnormalities (dysplasia or carcinoma in situ) adjacent to toms of irritability, and urinary cytology is likely to be
the presenting lesion or at distant sites.48,68,69 These positive. Other forms of carcinoma in situ may be unifo-
appear more likely to be aggressive, particularly if they cal and occasionally more well differentiated. These may
are rapidly recurrent or do not respond to adjunctive theoretically represent a less malignant form of this
intravesical BCG (see Chapter 18). diathesis.81
It is highly important that the resection specimen The ominous prognosis ascribed to carcinoma in situ
includes the muscularis propria in the staging of these may have originally been based on its association with
lesions. Several studies have documented muscle invasion concomitant muscle-invasive transitional cell cancer.82
on repeat resection in 10% to 20% of tumors initially staged The muscle-invasive cancer rather than the carcinoma in
as having only involved the lamina propria, particularly situ, at least as diagnosed at that phase of its course, was
when extensive lamina propria invasion has been seen.38,70,71 probably the component that progressed and that was
In addition, the submucosal connective tissue (lamina responsible for metastasis in the majority of such cases.
propria) contains a muscle layer (muscularis mucosae), However, knowing what we now know of the biology of
the thickness of which can vary from being either virtu- this diathesis, the specific pathway in which neoplastic
ally absent to being quite prominent.72 In the latter transformation had originally occurred and had led to
instance, this muscle layer may be confused with the this diathesis may in turn have led to the formation of
muscularis propria. Several reports have suggested the nodular or sessile muscle-invasive disease with penetra-
muscularis mucosae as a staging boundary, above which tion into the bladder wall that was the form in which can-
(stage T1a) tumors do not have a strong risk of progres- cer was diagnosed on presentation.3,83,84
sion and beneath which (stage T1b) there is a higher risk Once the entity of carcinoma in situ was appreciated
of progression to muscle invasive disease.73 Penetration and urinary cytology came into more common use, the
of the muscular mucosae as an indication of a more diagnosis of this entity in association with more superfi-
aggressive cancer and its use in staging remains to be val- cially invasive disease (a secondary form of diathesis) or
idated. At the same time, many of those tumors that as the only (primary) form of cancer that was present
invade more deeply appear to be more extensive while became more common. It therefore should not have been
those that are more superficially invasive appear to be less surprising to see an apparently prolonged and possibly
extensive and may have invaded in only microscopic nonprogressive course of carcinoma in situ when muscle-
foci.74 This in itself may be more indicative of the intrin- invasive cancer was not present. Indeed, several studies
sic biologic potential of the particular tumor diathesis suggested that this form of cancer by itself might not
rather than of its topographically “prognostic” relation to pose a major threat as the cancer cells forming carcinoma
muscularis mucosae fibers. in situ might lack the biochemical machinery necessary
In considering distinctions in the biologic potential of to become invasive.76,77,81 However, it is also possible
various forms of superficial bladder cancer, grade of the that these observations represented lead-time biases in
individual cells may be highly important as the manifes- describing the course and risk of this diathesis.
tation of a particular biologic capability.75 This may be The ultimate ominous potential of carcinoma in situ
manifested in the development of generalized changes in has been more fully appreciated on the basis of molecu-
the urothelium leading to expression of a more diffuse lar changes that characterize this diathesis.77,85-87 This
neoplastic diathesis known as carcinoma in situ.76 The has been supported further by its association with those
ultimate natural history of this entity may reflect forms of high-grade papillary cancer that may not have as
the intrinsic biology of the neoplastic diathesis and yet invaded the muscularis propria. These have an omi-
should be taken into account in the staging of a particu- nous prognosis, rapid recurrence, and progression to
lar bladder cancer and in the implementation of the muscle-invasion and metastasis soon after initial diagno-
TNM staging system. sis. In long-term follow-up of carcinoma in situ, the
310 Part IV Bladder

incidence of progressive disease has been reported as these progress, leaving 20% of stage T1 tumors
75% to 80% in patients with diffuse disease. (equaling 6% of all superficial tumors or 4% of all
Several additional observations are relevant. First, car- urothelial cancers).62,65,67
cinoma in situ has been found to penetrate the lamina
propria microscopically in 20% to 30% of cases.83 Approximately 50% of patients who present with muscle
Second, high-grade carcinoma in situ appears to be more invasive disease are thought to have occult metastasis at
commonly associated with papillary and papillo-nodular diagnosis, these generally being expressed within 2 years
tumors that are invasive of the lamina propria than with regardless of aggressive locoregional treatment.89
papillary tumors that are mucosally confined.48,76,84 When The directness of these associations and inaccuracies
associated with the latter, the tumors may be micropapil- of staging have led to a broad variability in suggested cor-
lary, and usually are high grade. Since those tumors that relations. For example, tumors that have penetrated only
have invaded the lamina propria are more likely to the superficial muscle (stage T2) may have a better prog-
become progressive, the carcinoma in situ associated nosis than those that have penetrated only the lamina
with these may reflect a more aggressive diathesis as well. propria (stage T1) but have done this extensively.90 Those
Third, several reports have suggested the development of that have a papillary configuration and have invaded in a
solid or nodular tumors from foci of carcinoma in situ.84 broad front may have a better prognosis than those that
If carcinoma in situ is the direct antecedent of nodular have a nodular configuration and that have manifested a
tumors, which are more deeply invasive when they tentacular form of infiltration.91,92 Furthermore, cancers
become clinically apparent than are papillary tumors, the staged as T2 may have been infiltrative of only the mus-
diagnosis of this type of carcinoma in situ may signify a cularis mucosae (the muscle layer within the lamina pro-
particularly ominous neoplastic diathesis. pria) and not of the muscularis propria. And, even if truly
The correct placement of carcinoma in situ within the invasive of the muscular propria, many of these may only
generally accepted staging system remains unclear. All have been very superficially invasive. Thus, despite hav-
forms of bladder cancer have their origin in intraepithelial ing had the ability to infiltrate, they may have invaded at
neoplastic transformation, which, by definition, produces a less rapid rate and may have lacked the ability to pene-
“carcinoma in situ.” Indeed, pathologists often identify trate lymphatics and vasculature and metastasize.92
papillary mucosally confined tumors as papillary carcinoma Correspondingly, cancers staged as T1 may have been
in situ, notwithstanding low-moderate grade cells and the understaged in 10% to 20% of instances, since repeat
implication of disease with low malignant potential. resection has found residual cancer that was actually
However, despite its confinement technically to the invasive of the muscularis propria.38,70,71 These, as well as
mucosa, this diathesis is clearly quite different from the those staged as T1 that are extensive in their penetration,
form of flat carcinoma in situ that is associated with lamina may have a particularly aggressive biologic potential.65-67
propria-invasive high-grade or muscle-invasive disease. When recurrent (or persistent) they may already have
Descriptions of cancers as “invasive” have generally extended into the muscularis and even metastasized.
implied their penetration of the bladder wall muscle At the 1993 bladder cancer consensus conference in
(muscularis propria). Progressive depth of penetration Antwerp, a proposal was made that stage T3a tumors
into and through this layer has been correlated with an (those with deep muscle invasion) and stage T2 tumors
increased likelihood of metastasis. A majority of patients (those with superficial muscle invasion) be combined in a
diagnosed with muscle invasive disease are diagnosed at single category of T2.93 Stage T2a would then comprise
this stage of disease at their initial presentation.88,89 those tumors that had invaded only the superficial muscle
These comprise 20% to 30% of all urothelial cancers at (less than one-half the depth of the muscle layer) and stage
initial presentation. Muscle invasive cancers that arise T2b would comprise those that had invaded the deep mus-
from initially superficial tumors comprise only an addi- cle (deeper than half the depth of the muscle layer). Stage
tional 10% to 12%. This reflects calculations based on T3 would then designate tumor penetration into the
observations in stage T1 disease (since only 2% to 4% of perivesical tissue. Stage T3a would comprise those tumors
stage Ta tumors progress) as follows: that demonstrated only microscopic invasion beyond the
muscularis and stage T3b would comprise those that had
1. 20% to 30% of T1 tumors (comprising 30% of all invaded more extensively into the extravesical tissues.
superficial tumors on initial diagnosis) are actually The rationale for including all muscle-invasive cancers
muscle-invasive on repeat resection (equals 6% to in one category reflected in part the problem of accu-
9% of all superficial tumors or 4% to 6% of all rately determining the depth of muscle invasion by
urothelial cancers);61,62 transurethral resection, as well as the impression that all
2. 40% of initially high-grade T1 cancers are at risk for cancers invading the muscle wall were likely to behave in
progression (representing an original 50%, less 20% a similar aggressive fashion regardless of their depth of
which are already muscle invasive) but only 50% of penetration.94-96 It also reflected the concept that
Chapter 17 Diagnosis and Staging of Bladder Cancer 311

muscle-invasive tumors did not exhibit the same poten- urethra and prostatic ducts, prognosis will be less omi-
tial biologic behavior as did tumors that had extended nous than if the cancer has infiltrated into the prostatic
through the full thickness of the bladder wall into the stroma.80 An analogous situation may pertain in women.
perivesical fat.97-99 If there has been direct penetration only to the outer
Some have suggested that this new categorization fails aspect of the anterior vaginal wall, prognosis may be less
to take into account several observations. First, those ominous than if there has been more extensive involve-
tumors that have penetrated only the superficial muscle ment of the vagina or penetration of the uterus. Evidence
appear to be less likely to be associated with metastasis validating this suggestion has not been obtained. Direct
and appear also to be more likely to be cured by a variety involvement of other adjacent structures or fixation to
of surgical approaches.97,98 For example, many of these the pelvic sidewalls is generally tantamount to the cancer
have shown no remaining cancer in the cystectomy being incurable. Distant metastatic disease is nearly
specimen after initial extensive transurethral resec- invariably present in these situations even if only occult.
tion.89,100,101 In addition, treatment outcomes have often Therefore, locoregional treatments generally fail.99,105,106
been more successful either in terms of time to disease Involvement of regional lymph nodes has generally
recurrence or survival as for more deeply invasive dis- been interpreted as indicating an ominous prognosis.107
ease.101-103 That more superficial muscle invasion has Several recent reports have suggested that prognosis may
been associated more often with a papillary configura- not be as grave when only microscopic metastases are
tion, invasion in a broad front, and less frequent involve- found and only involve 1 to 2 regional lymph nodes.108,109,111
ment of bladder wall lymphatics and vasculature than Under these circumstances, aggressive surgery with
deep muscle-invasive cancer is supportive of distinctions meticulous lymphadenectomy has been reported to pro-
between these diatheses (see later).91,92 Second, those duce 5-year survivals as high as 35%.108 Moreover, although
tumors that have invaded the muscle layer more deeply gross involvement of the lymph nodes or involvement of
appear to have an ominous prognosis similar to that of more than two lymph nodes has previously been associ-
tumors that have involved perivesical soft tissues micro- ated with a poor prognosis, recent studies have suggested
scopically.93,94,99 This harks back to earlier staging cate- that more extensive dissection of lymph nodes beyond
gories that combined deep muscle invasion with extension the pelvis and inclusive of all pelvic nodes resulted in
into perivesical fat.55 Differences in clinical manifestation improved outcomes.110 Whether staging subdivisions
and treatment outcomes may be less restrictive than dis- according to the degree of lymph node involvement,
tinctions between these categories imply. number of lymph nodes involved, proportion of nodes
Superficial muscle invasion has been associated with a involved per total number of lymph nodes removed, or
papillary configuration and a pattern of infiltration that differences between macroscopic versus microscopic dis-
has been described as “broad front,” in which tumor cells ease play a role in outcomes and should adjust surgical
penetrated the bladder wall muscle in large clusters of treatment approaches remains to be validated.
cells with a broad infiltrative margin.91,92 Such tumors Distinctions in organ involvement by distant metas-
have appeared to involve the bladder wall vasculature and tases have not been associated with variability of out-
lymphatics in only one-third of cases.47 In contrast, comes and survival. Therefore, staging subdivisions
tumors that have invaded more deeply have been associ- according to organ involvement have not been included
ated with a more nodular type of configuration and a pat- in standard staging systems.
tern of infiltration that has been described as
“tentacular,” in which tumor cells have appeared to “per-
PATHOGENIC PATHWAYS IN BLADDER CANCER
colate” through the bladder wall in smaller clusters or as
finger-like projections.47,91,92 Such tumors have been Schematic depictions of bladder cancer staging systems
found to involve the bladder wall lymphatics and vascu- in association with likelihood of progression have created
lature in at least two-thirds of instances.47 the impression that the various cancer diatheses fall into
Stage T4 disease has been compartmentalized into a sequence of stages in their pathogenesis (see Figure
stage T4a, representing tumor invasion of the prostate, 17-1). This has implied further that progression from the
uterus, or vagina, and stage T4b representing tumor most superficial appearance histologically to the most
invasion of the pelvic or abdominal wall. Conflicting extensive invasion is inexorable.
observations have made definitive staging in these cate- Such schema, however, as they imply a sequence of
gories somewhat unclear. For example, 40% of cystec- stages, do not take into account issues in the pathogene-
tomy specimens have cancer involvement of the sis of disease. The natural history of various forms of
prostate.80,104 Traditionally thought to indicate an omi- bladder cancer may instead reflect developmental path-
nous prognosis, prostatic involvement may actually influ- ways that, although interrelated, may be distinct from
ence outcomes in a variable manner. If the cancer is one another (see Figure 17-2). A schema that incorpo-
mucosally confined while extending into the prostatic rates the concept of a variety of different developmental
312 Part IV Bladder

pathways can complement the standard concept of a cells that extend along the plane of the bladder
sequence of stages in understanding the pathogenesis of mucosa,75,76,79 possibly undermine it, cause it to slough,
different forms of bladder cancer. and then themselves slough. Such cells lack cohesive and
In this construct, stage of disease can be viewed as a adhesive ability. Through proliferation and continued
snapshot in the evolving pathogenesis of disease. Although pagetoid spread, a high-grade flat neoplastic diathesis
stage may remain consistent with time, changes may also could occur that in some instances might lead to the
occur. The stage at resection as manifest in the snapshot development of either papillary tumors that extend into
provided by the resection specimen may be used to suggest the lumen of the bladder or, more likely, nodular tumors
prognosis, but this may profitably be understood in the that penetrate into the lamina propria and muscu-
context of the potential pathogenesis of a particular tumor laris.59,68,81,83 Some have suggested that carcinoma in situ
diathesis. Distinctions in the potential biologic behavior of leads directly to infiltration of nodular disease into the
a particular cancer at a specific point in time (either at ini- bladder wall, so that silent carcinoma in situ is diagnosed
tial diagnosis or at the time of recurrence) may be of use in initially in its muscle-invasive manifestation.84
determining whether aggressive treatment is indicated The various staging systems have not as yet incorpo-
(even if a tumor appears to be at an “early” stage) or rated the genetic changes that characterize different can-
whether conservative treatment may be appropriate (even cers. Although many reports have focused on specific
if a tumor appears to be more advanced). chromosomal and molecular changes that correlate with
In considering a schema in which different pathways the propensity either towards proliferation or to invasion
portray the biology of various types of bladder cancer, and metastasis,112–116 these have not as yet been fully val-
several developmental terms may be applied to describe idated. Therefore, they have not been incorporated into
the different processes that give rise to various forms of staging systems.
tumor diathesis. For example, papillary tumors may arise They have, however, been correlated with different
through a process that can be described as proliferative. developmental pathways as suggested in schema for tumor
Accordingly, transformed epithelial cells proliferate and pathogenesis.2,4,77,85 For example, aberrations of chromo-
produce tumors with a multilayered epithelium in papil- some 9 have been seen in both low-grade, low-stage blad-
lary configuration surrounding a central fibrovascular der tumors and higher grade, invasive tumors. This has
core. The process of proliferation by itself may produce suggested that loss of heterozygosity of chromosome 9
tumors that contain cells with a histologically normal may occur early in the genesis of a variety of bladder can-
appearance (low grade). Such tumors may lack the cers.117–120 It has also been shown that chromosome 9 is
machinery necessary to invade the lamina propria aggres- lost in each of several multifocal tumors within an individ-
sively, progress, and metastasize. ual patient, supporting the concept of a field change in the
Tumors that are comprised of histologically abnormal genesis of bladder cancer.121,122 Abnormalities in chromo-
(high grade) cells may occur as the result of a process that some 9 have been associated with a proliferative diathe-
can be termed “dysplasia” superimposed on the prolifer- sis.117,120 Specific foci for these changes have been variable,
ative process. It is unusual to see papillary tumors that are but all have been correlated with neoplastic transforma-
high grade and truly mucosally confined. Indeed, this has tion that is reflected in a proliferative diathesis and that
been reported to occur in only 2% to 4% of all mucos- characterizes the course of these tumors as one of recur-
ally confined tumors.61,62 rence rather than progression.120
A high-grade histologic appearance in papillary Loss of heterozygosity of chromosomes other than
tumors is more commonly seen when penetration of the chromosome 9 has been observed in high-grade, high-
basement membrane and invasion of the lamina propria stage tumors.77,86,87 Mutations in chromosome 17 with
has occurred.64–66 This difference in appearance concep- expression of higher levels of p53 have been identified in
tually suggests a developmental process that can be approximately half of all high-grade high-stage bladder
termed dysplasia. It may indicate a different biologic cancers.86,123 Abnormalities in chromosome 17 have also
capability of these tumors such that they can proliferate been associated with tumors more likely to invade and
but also infiltrate. Although low-grade, mucosally con- metastasize.124–126
fined tumors could conceivably change during the course Several studies have also shown that carcinoma in situ
of their proliferation to become high-grade tumors that expresses high levels of p53 mutations, and that it is not
have acquired the biochemical machinery to penetrate characterized by loss of heterozygosity of chrom-
into the sub-epithelial stroma, dysplasia may hypotheti- osome 9.86,87,127 This is in keeping with the apparently
cally have been the primary result of neoplastic transfor- aggressive course implied by the presence of carcinoma
mation, leading to the development of a papillary in situ either alone as “primary” or in the setting of high-
high-grade urothelial cancer capable of invasion. grade lamina propria or muscle invasive cancer as “sec-
The neoplastic pathway characterized predominantly ondary.”81,83,84 Other studies have demonstrated that
by dysplasia could also lead to the development of cancer only those lamina propria invasive tumors that express
Chapter 17 Diagnosis and Staging of Bladder Cancer 313

p53 progress to muscle invasion suggesting a commonal- T2-weighted MR imaging, dynamic gadolinium-
ity of pathways with the more aggressive tumor diatheses enhanced imaging, and late gadolinium-enhanced
expressing abnormalities in chromosome 17. imaging. Radiology 1994; 193:239–245.
Most studies have been based on immunohistochemical 14. Husband JE, Olliff JFC, Williams MP, et al: Bladder
cancer staging with CT and MR imaging. Radiology
studies. Some have been validated with analysis of selected
1989; 173:435.
sequence conformational polymorphisms. Taken together,
15. Tanimoto A, Yuasa Y, Imai Y, et al: Bladder tumor
initial changes in chromosome 9 may be followed by mul- staging: comparison of conventional and gadolinium-
tiple sequential changes of other genes that may then lead enhanced dynamic MR imaging and CT. Radiology
to an invasive and potentially metastatic phenotype. 1992; 185:741–747.
Although these situations are complex, it may ulti- 16. Nishimura K, Hido S, Nishio Y, et at: The validity of
mately be possible to superimpose a molecular profile for MRI in the staging of bladder cancer: comparison with
a particular cancer on the snapshot stage of a tumor as CT and transurethral US. Am J Clin Oncol 1922;
determined by histopathologic analysis to determine not 18:217.
only the type of therapy that may be effective in a partic- 17. Narumi Y, Kadota T, Inoue E, et al: Bladder tumors:
ular cancer diathesis but also to predict the outcome of staging with gadolinium-enhanced oblique MR imaging.
Radiology 1993; 187:145–150.
particular treatment regimens in the context of the natu-
18. Barentsz JO, Jager G, Mugler JP III, et al: Staging
ral history and propensity of that diathesis based on its
urinary bladder cancer: value of T1-weighted three-
predicted pathogenesis. dimensional magnetization prepared-rapid gradient-echo
and two-dimensional spin-echo sequences. Am J
Roentgenol 1995; 164:109–115.
REFERENCES 19. Ahlstrom H, Malmstrom PU, Letocha H, et al:
Positron emission tomography in the diagnosis and
1. Green FL, Pate DL, Fleming ID, et al: AJCC cancer staging of urinary bladder cancer. Acta Radiol 1996;
staging manual. Philadelphia, Springer, 2002. 37:180–185.
2. Droller MJ: Bladder cancer: state of the art care cancer. 20. Bender H, Schomburg A, Albers P, et al: Possible role of
Cancer J Clin 1998; 48:269. FDG-PET in the evaluation of urologic malignancies.
3. Droller MJ: Bladder cancer. Curr Prob Surg 1981; 18:205. Anticancer Res 1997; 17:1655–1660.
4. Jones PA, Droller MJ: Pathways of development and 21. Heicappel R, Muller-Mattheis V, Reinhardt M, et al:
progression in bladder cancer: new correlations between Staging of pelvic lymph nodes in neoplasms of the
clinical observations and molecular mechanisms. Semin bladder and prostate by positron emission tomography
Urol 1993; 11:177. with 2-[(18)F]-2-deoxy-D-glucose. Eur Urol 1999;
5. Jemal A, Thomas A, Murray T, et al: Cancer statistics, 36:582–587.
2002. CA Cancer J Clin 2002; 52:23. 22. Boccon Gibod L, Katz M, Cochand B, et al:
6. Messing EM, Young TB, Hunt VB, et al: Urinary tract Lymphography and percutaneous fine needle node
cancers found by home screening with hematuria aspiration biopsy in the staging of bladder carcinoma.
dipstick in healthy men over 50 years of age. Cancer J Urol 1984; 132:24–26.
1989; 64:2361. 23. Chagnon S, Cochand-Pirollert B, Gzaeil M, et al: Pelvic
7. Messing EM, Young TB, Hunt VB, et al: The cancers: staging of 139 cases with lymphography and
significance of asymptomatic microhematuria in men 50 fine-needle aspiration biopsy. Radiology 1989;
or more years old: findings of a home screening study 173:103–106.
using urinary dipsticks. J Urol 1987; 137:919–922. 24. Brown FM: Urine cytology. Is it still the gold standard
8. Resnick MI: Overview: imaging of the genitourinary for screening? Urol Clin N Am 2002; 27:25–37.
tract. In Droller MJ (ed): Surgical management of 25. Gregoire M, Fradet Y, Meyer F, et al: Diagnostic
urologic disease, an anatomic approach, p 69. St. Louis, accuracy of urinary cytology, and deoxyribonucleic acid
Mosby-Yearbook, 1992. flow cytometry and cytology on bladder washings during
9. Denkhaus H, Crone-Munzebrock W, Huland H: follow-up for bladder tumors. J Urol 1997;
Non-invasive ultrasound in detecting and staging bladder 157:1660–1664.
carcinoma. Urol Radiol 1985; 7:121. 26. Badalament RA, Fair WR, Whitmore WF Jr, et al:
10. Hatch TR, Barry JM: The value of excretory urography The relative value of cytometry and cytology in the
in staging bladder cancer. J Urol 1986; 135:49. management of bladder cancer. Semin Urol 1988;
11. Barentsz JO, Witjes JA, Ruijs JH: What is new in bladder 6:22.
cancer imaging. Urol Clin N Am 1997; 24:583–602. 27. Billerey C, Lamy B, Bettard H, et al: Flow cytometry
12. Caterino M, Giunta S. Finocchi V, et al: Primary cancer versus urinary cytology in the diagnosis and follow-up of
of the urinary bladder: CT evaluation of the T bladder tumors: critical review of a 5-year experience.
parameter with different techniques. Abdom Imaging World J Urol 1993; 11:156.
2001; 26:433–438. 28. Bastacky S, Ibrahim S, Wilczynski SP, Murphy WM:
13. Kim B, Semelka RC, Ascher SM, et al: Bladder tumor The accuracy of urinary cytology in daily practice.
staging: comparison of contrast-enhanced CT, T1- and Cancer 1999; 87:118–128.
314 Part IV Bladder

29. Mian C, Pycka A, Wiener W, et al: ImmunoCyt: a new 44. Koraitim M, Kamal B, Metwalli N, et al: Transurethral
tool for detecting transitional cell cancer of the urinary ultrasonographic assessment of bladder carcinoma: its
tract. J Urol 1999; 161:1486–1489. value and limitation. J Urol 1995; 154:375–378.
30. Holling KC, King W, Sokolova IA, et al: A comparison 45. Beer M, Schmidt H, Riedl R: The clinical value of
of cytology and fluorescence in situ hybridization for the preoperative staging of bladder and prostatic cancers
detection of urothelial carcinoma. J Urol 2002; with nuclear magnetic resonance and computerized
164:1768–1775. tomography. Urologe A 1989; 28:65–69.
31. Cajulis RS, Haines GK III, Frias-Hidvegi D, et al: 46. Herr HW: Routine CT scan in cystectomy patients: does
Cytology, flow cytometry, image analysis, and interphase it change management? Urology 1996; 47:324–325.
cytogenetics by fluorescence in situ hybridization in the [Published erratum appears in Urology 1996; 47:785].
diagnosis of transitional cell carcinoma in bladder 47. Slack NH, Prout GR Jr: Heterogeneity of invasive
washes: a comparative study. Diagn Cytopathol 1995; bladder carcinoma and different responses to treatment.
13:214–223. In Bonney WW, Prout GR (eds): Bladder Cancer, AUA
32. Sarosdy MF, de Vere White RW, Soloway MS, et al: Monographs, Vol. 1, p 212. Baltimore, Williams &
Results of a multicenter trial using the BTA test to Wilkins, 1980.
monitor for and diagnose recurrent bladder cancer. 48. Althausen AF, Prout GR Jr, Daly JJ: Non-invasive
J Urol 1995; 154:379–383. papillary carcinoma of the bladder associated with
33. Leyh H, Marberer M, Conort P, et al: Comparison of carcinoma in situ. J Urol 1976; 116:575–580.
the BTA stat test with voided urine cytology and bladder 49. Vicente J, Laguna MP, Duarte D, et al: Carcinoma in
wash cytology in the diagnosis and monitoring of situ as a prognostic factor for G3pT1 bladder tumors. Br
bladder cancer. Eur Urol 1999; 35:52–56. J Urol 1991; 68:380.
34. Soloway MS, Briggman V, Carpinito GA, et al: Use of a 50. Epstein JL, Amin MB, Reuter VR, et al: The World
new tumor marker, urinary NMP22, in the detection of Health Organization/International Society of Urological
occult or rapidly recurring transitional cell carcinoma of Pathology consensus classification of urothelial
the urinary tract following surgical treatment. J Urol (transitional cell) neoplasms of the urinary bladder.
1996; 156:363–367. Bladder Consensus Conference Committee. Am J Pathol
35. Konety BR, Getzenberg RH. Urine based markers of 1998; 22:1435–448.
urological malignancy. J Urol 2001; 165:600–611. 51. Paik ML, Scolieri MJ, Brown SL, et al: Limitation of
36. Van der Meijden MA, Oosterlinck W, Brausi M, et al: computerized tomography in staging invasive bladder
Significance of bladder biopsies in Ta, T1 bladder tumors: cancer before radical cystectomy. J Urol 2000;
a report from the EORTC Genito-Urinary Tract Cancer 163:1693–1696.
Cooperative Group. EORTC-GU Group Superficial 52. Malmstrom PU, Lonnemark M, Busch C, et al: Staging
Bladder Committee. Eur Urol 1999; 35:267–271. of bladder carcinoma by computer tomography-guided
37. Angulo JC, Lopez JI, Grignon DJ, et al: Muscularis mucosa transmural core biopsy. Scan J Urol Nephrol 1993;
differentiates two populations with different prognosis in 27:193.
stage T1 bladder cancer. Urology 1995; 44:47–53. 53. Broders AC: Epithelioma of the genitourinary organs.
38. Klan R, Loy V, Huland H. Residual tumor discovered in Ann Surg 1922; 75:574.
routine second transurethral resection in patients with 54. Aschner PW: The pathology of vesical neoplasms. Its
stage T1 transitional cell carcinoma of the bladder. evaluation in diagnosis prognosis. JAMA 1928; 91:1697.
J Urol 1991; 146:316–318. 55. Jewett HJ, Strong GH: Infiltrating carcinoma of the
39. Kakizoe T, Tobisu K, Takai K, et al: Relationship bladder: relation of depth of penetration of the bladder
between papillary and nodular transitional cell carcinoma wall to incidence of local extension and metastases.
in the human urinary bladder. Cancer Res 1988; J Urol 1946; 55:366.
48:2299. 56. Jewett HJ, Lewis E: Infiltrating carcinoma of bladder:
40. Friedell GN, Parija GC, Nagy K, et al: The pathology of curability by total cystectomy. J Urol 1948; 60:107.
human bladder cancer. Cancer 1980; 45:1823. 57. Fuller WA Jr: Staging of advanced bladder cancer.
41. Tomita Y, Kobayashi K, Saito T, et al: Use of miniature Current concepts and pitfalls (Review). Urol Clin N Am
ultrasonic probe system for intravesical ultrasonography 1992; 19:663.
for transitional cell cancer of the urinary tract. Scand 58. Herr HW: Staging invasive bladder tumors (Review).
J Urol Nephrol 2000; 34:313–316. J Surg Oncol 1992; 51:217.
42. Kim B, Semelka RC, Ascher SM, et al: Bladder tumor 59. Kotake T, Flanigan RC, Kirkels W, et al: The current
staging: comparison of contrast-enhanced CT, T1- and TNM-classification of bladder carcinoma: Is it as good
T2-weighted MR imaging dynamic gadolinium- as we need it to be. Int J Urol 1995; 2:36.
enhanced imaging and late gadolinium-enhanced 60. Hermanek P, Sobin LH: International Union Against
imaging. Radiology 1994; 193:239–245. Cancer: TNM Classification of Malignant Tumours, 2nd
43. Scattoni V, Da Pozzo LF, Colombo R, et al: Dynamic revision, 4th edition. Berlin, Springer-Verlag, 1992.
gadolinium-enhanced magnetic resonance imaging in 61. Heney NM, Nocks BN, Daly JJ, et al: Ta and T1
staging of superficial bladder cancer. J Urol 1996; bladder cancer; location, recurrence, and progression.
155:1594–1599. Br J Urol 1982; 54:152.
Chapter 17 Diagnosis and Staging of Bladder Cancer 315

62. Heney NM, Ahmed S, Flanagan MJ, et al: Superficial specimens removed for bladder cancer. J Urol 1989;
bladder cancer: progression and recurrence. J Urol 1983; 141:346.
130:1083. 81. Herr HW: When is a cystectomy necessary in carcinoma
63. Cheng L, et al: Natural history of urothelial dysplasia of in situ? In Murphy GP, Khoury S (eds): Therapeutic
the bladder. Am J Surg Pathol 1999; 23:443–447. Progress in Urologic Cancers, p 511. New York, Alan R.
64. Jakse G, Loidle W. Seeber G: Stage T1, grade 3 Liss, 1989.
transitional cell carcinoma of the bladder: an unfavorable 82. Melicow M: Histological study of vesical epithelium
tumor: J Urol 1987; 137:39. intervening between gross neoplasms in total cystectomy.
65. Abel PD, Hall RR, Williams G: Should T1 transitional J Urol 1952; 68:261.
cell carcinoma of the bladder still be classified as 83. Farrow GM, Utz DC: Observations on microinvasive
superficial?: J Urol 1988; 62:235. transitional cell carcinoma of the urinary bladder. Clin
66. Holmang S, Hedelin H, Anderstrom C, et al: The Oncol 1982; 1:609.
importance of the depth of invasion in stage G1 bladder 84. Kakizoe T, Tobisu K, Takai K, et al: Relationship
carcinoma: a prospective cohort study. J Urol 1997; between papillary and nodular transitional cell carcinoma
157:800–804 [comment]. in the human urinary bladder. Cancer Res 1988;
67. Cheng I, Neumann RM, Weaver Al, et al: Predicting 48:2299.
cancer progression in patients with stage T1 bladder 85. Tsai YC, Nichols PW, Hiti Al, et al: Allelic losses of
carcinoma. J Clin Oncol 1999; 17:3182–3187. chromosome 9, 11 and 17, in human bladder cancer.
68. Kakizoe T, Matumoto K, Nishio Y, et al: Significance Cancer Res 1990; 50:44.
of carcinoma in situ in association with bladder cancer. 86. Sarkis AS, Dalbagni G, Cordon-Cardo C, et al: Nuclear
J Urol 1985; 133–395. overexpression of p53 protein in transitional cell bladder
69. Vicente J, Laguna MP, Duarte D, et al: Carcinoma in carcinoma: a marker for disease progression. J Natl
situ as a prognostic factor for G3pT1 bladder tumours. Cancer Inst 1993; 85:53.
Br J Urol 1991; 68:380–382. 87. Hartmann A, Schlake G, Zaak D, et al: Occurrence of
70. Koloszy Z: Histopathological “self-control” in chromosome 9 and p53 alterations in multifocal
transurethral resection of the bladder tumours. Br J Urol dysplasia and carcinoma in situ of human urinary
1991; 67:162. bladder. Cancer Res 2002; 62:809–818.
71. Langenstroer P, See W: The role of a second 88. Kaye KW, Lange PH: Mode of presentation of invasive
transurethral resection for high-grade bladder cancer. bladder cancer: Reassessment of the problem. J Urol
Curr Urol Rep 2000; 1:204–207. 1982; 128:31–33.
72. Engel P, Anagnostaki L, Braendstrup O. The muscularis 89. Slack NH, Prout GR Jr: Heterogeneity of invasive
mucosae of the human urinary bladder. Implications for bladder carcinoma and different responses to treatment.
tumor staging on biopsies. Scan J Urol Nephrol 1992; J Urol 1980; 123:644.
26(Suppl):249–252. 90. Jewett HJ: Comments on the staging of invasive bladder
73. Angulo JC, Lopez, JI, Grignon DJ, et al: Muscularis cancer: two B’s or not two B’s: that is the question
mucosa differentiates two populations with different (apologies to Shakespeare, Hamlet, act 2, sc I, lines
prognosis in stage T1 bladder cancer. Urology 1995; 1056). J Urol 1978; 119:39 (editorial).
45:47–53. 91. Fridell GN, Parija GC, Nagy K, et al: The pathology of
74. Platz CE, Cohen MB, Jones MP, et al: Is microstaging of human bladder cancer. Cancer 1989; 45:1823.
early invasive cancer of the urinary bladder possible or 92. Soto EA, Friedell GH, Tiltman AJ: Bladder cancer as
useful? Mod Pathol 1996; 9:1035–1039 [comment]. seen in giant histologic sections. Cancer 1977; 39:447.
75. Jordan AM, Weingarten J, Murphy WM: Transitional 93. Schroder FH, Cooper EH, Debruyne FMJ, et al: TNM
cell neoplasms of the urinary bladder. Can biological classification of genitourinary tumors 987-position of
potential be predicted from histologic grading? Cancer the EORTC Genitourinary Group. Br. J Urol 1988;
1987; 60:2761. 62:502.
76. Lamm DL: Carcinoma in situ. Urol Clin N Am 1992; 94. Hendry WF, Rawson NSB, Turney L, et al:
19:499. Computerization of urothelial carcinoma records: 16
77. Spruck CH, Ohneseit PF, Gonzalez-Zulueta M, et al: years’ experience with the TNM system. Br J Urol 1990;
Two molecular pathways to transitional carcinoma of the 65:583.
bladder. Cancer Res 1994; 54:784. 95. Herr HW: A proposed simplified staging system of
78. Jones PA, Droller MJ: Pathways of development and invasive bladder tumors. Urol Int 1993; 50:17.
progression in bladder cancer: new correlations between 96. Herr HW, Scher HI: Surgery of invasive bladder cancer:
clinical observations and molecular mechanisms. Semin is pathologic staging necessary. Semin Oncol 1990;
Urol, 1993; 11:177–192. 17:590–597.
79. Weinstein R, Miller AW III, Pauli BV: Carcinoma in 97. Frazier HA, Robertson JE, Dodge RK, et al: The value
situ: comments on the pathology of a paradox. Urol Clin of pathologic factors in predicting cancer specific
N Am 1980; 18:523. survival among patients treated with radical cystectomy
80. Wood DP, Montie JE, Pontes JE, et al: Transitional cell for transitional cell cancer of the bladder and prostate.
carcinoma of the prostate in cysto-prostatectomy Cancer 1993; 71:3993.
316 Part IV Bladder

98. Wishnow KI, Levinson AK, Johnson DE, et al: Stage B bladder cancer detected by comparative genomic
(P2/3A/N0) transitional cell carcinoma of bladder hybridization. Cancer Res 1997; 57:2860–2864.
highly curable by radical cystectomy. Urology 1992; 114. Chow NH, Cairns P, Eisenberger CF, et al: Papillary
39:12–16. urothelial hyperplasia is a clonal precursor to papillary
99. Roehrborn CG, Sagalowsky AI, Peters PC. Long-term transitional cell bladder cancer. Int J Cancer 2000;
patient survival after cystectomy for regional metastatic 89:514–518.
transitional cell carcinoma of the bladder. J Urol 1991; 115. Presti JC, Jr., Reuter VE, Galan T, et al: Molecular
146:36–39. genetic alterations in superficial and locally
100. See WA, Fuller JR: Staging of advanced bladder cancer: advanced human bladder cancer. Cancer Res 1991;
current concepts and pitfalls. Urol Clin N Am 1992; 51:5405–5409.
19:663. 116. Baithun SI, Naase M, Blanes A, et al: Molecular and
101. Herr HW, Whitmore W Jr, Morse MJ, et al: kinetic features of transitional cell carcinomas of the
Neoadjuvant chemotherapy in invasive bladder cancer: bladder: biological and clinical implications. Virchows
the evolving role of surgery. J Urol 1990; 144:1083–1088 Arch 2001; 438:289–297.
[review]. 117. Miyao N, Tsai YC, Lerner SP, et al: The role of
102. Herr HW: Conservative management of muscle chromosome 9 in human bladder cancer. Cancer Res
infiltrating bladder cancer: prospective experience. 1993; 53:4066.
J Urol 1987; 138:1162–1163. 118. Eleuteri P, Grollino MG, Pomponi D, et al:
103. Solsona E, Iborra I, Ricos, JV, et al: Feasibility of Chromosome 9 aberrations by fluorescence in situ
transurethral resection for muscle-infiltrating carcinoma hybridization in bladder transitional cell carcinoma. Eur
of the bladder: prospective study. J Urol 1992; J Cancer 2001; 37:1496–503.
147:1513–1515. 119. Stadler WM, Steinberg G, Yang X, et al: Alterations of
104. Schellhammer PF, Bean MA, Whitmore WF: Prostatic the 9p21 and 9q33 chromosomal bands in clinical
involvement by transitional cell carcinoma: pathogenesis, bladder cancer specimens by fluorescence in situ
patterns and prognosis. J Urol 1988; 118:399. hybridization. Clin Cancer Res 2001; 7:1676–1682.
105. Fossa SD, Ous S, Berner A: Clinical significance of the 120. Tsukamoto M, Matsuyama H, Oba K, et al: Numerical
“palpable mass” in patients with muscle-infiltrating aberrations of chromosome 9 in bladder cancer. A
bladder cancer undergoing cystectomy after pre- possible prognostic marker for early tumor recurrence.
operative radiotherapy. Br J Urol 1991; 67:54–60. Cancer Genet Cytogenet 2002; 134:41–45.
106. Gospodarowicz MK, Rider WD, Keen CW, et al: 121. Hafner C, Knuechel R, Zanardo L, et al: Evidence for
Bladder cancer: long-term follow-up results of patients oligoclonality and tumor spread by intraluminal seeding
treated with radical radiation. Clin Oncol 1992; in multifocal urothelial carcinomas of the upper and
3:155–161. lower urinary tract. Oncogene 2001; 20:4910–4915.
107. Smith JA Jr, Whitmore WF Jr: Regional lymph node 122. Vriesema JL, Aben KK, Witjes JA, et al: Superficial and
metastases from bladder cancer. J Urol 1981; 126:591. metachronous invasive bladder carcinomas are clonally
108. Skinner DG: Management of invasive bladder cancer: a related. Int J Cancer 2001; 93:699–702.
meticulous pelvic lymph node dissection can make a 123. Olumi AF, Tsai YC, Nicholes PW, et al: Allelic loss of
difference. J Urol 1982; 128:34. chromosome 17p distinguishes high grade from low
109. Lerner SP, Skinner DG, Lieskovsky G, et al: The grade transitional cell carcinomas of the bladder. Cancer
rationale for en bloc pelvic lymph node dissection for Res 1990; 50:7081.
bladder cancer patients with nodal metastases: long-term 124. Fujimoto K, Yamada Y, Okajima E, et al: Frequent
results. J Urol 1993; 149:758–765. associations of p53 gene mutations in invasive bladder
110. Paulson AL, Horn T, Steven K: Radical cystectomy: cancer. Cancer Res 1992; 52:1393.
extending the limits of pelvic lymph node dissection 125. Esrig D, Spruck CH III, Nichols PW, et al: p53 nuclear
improves survival for patients with bladder cancer protein accumulation correlates with mutations in the
confined to the bladder wall. J Urol 1998; p53 gene, tumor grade, and stage in bladder cancer. Am
160:2015–2019. J Pathol 1993; 143:1389–1397.
111. Wishnow KI, Johnson DB, Ro YJ, et al: Incidence, 126. Dalbagni G, Ren ZP, Herr H, et al: Genetic alterations
extent, and location of unsuspected pelvic lymph node in p53 in recurrent urothelial cancer: a longitudinal
metastasis in patients undergoing radical cystectomy for study. Clin Cancer Res 2001; 7:2797–2801.
bladder cancer. J Urol 1987; 137:408–412. 127. Edwards J, Duncan P, Going JJ, et al: Loss of
112. Theodorescu D: Commentary on genetic prognostic heterozygosity on chromosomes 11 and 17 are markers
markers for transitional carcinoma of the bladder: from of recurrence in TCC of the bladder. Br J Cancer 2001;
microscopes to molecules. J Urol 1996; 155:2. 85:1894–1899.
113. Richter J, Jiang F, Gorog JP, et al: Marked genetic
differences between stage pTa and stage pT1 papillary
C H A P T E R

18Superficial Transitional Cell Carcinoma


of the Bladder: Management and
Prognosis
Murugesan Manoharan, MD, FRCS, and Mark S. Soloway, MD

Superficial transitional cell carcinoma (TCC) of the uri- discomfort, as an office examination.1 The anterior blad-
nary bladder refers to tumor confined to the mucosa (Ta, der wall and bladder neck regions are well visualized with
Tis) or submucosa (T1). This is a heterogeneous disease flexible cystoscopy. Cold-cup biopsy is possible, although
with a variable natural history. At one end of the spec- sufficient tissue to determine depth of invasion is difficult
trum, low-grade Ta tumors have a low progression rate to obtain with small forceps. Hence, we prefer cys-
and require initial endoscopic treatment and surveillance toscopy under anesthesia to obtain satisfactory biopsy.
but rarely represent a threat to the patient. At the other Upper tract radiologic visualization should be per-
extreme, high-grade T1 tumors have a high malignant formed in all cases. Intravenous urography is sensitive in
potential with significant progression and cancer death detecting papillary tumors, as well as in providing func-
rates. Seventy percent of bladder tumors present as tional and anatomic information. Ultrasound has also
superficial disease. Approximately 70% of these tumors been used for upper tract assessment, with the advantages
present as Ta lesions, 20% as T1, and 10% as Tis (carci- of avoiding contrast reactions, radiation, and intestinal
noma in situ). The true natural history of untreated non- preparation. However, ultrasound is less sensitive in
invasive disease is not fully known. detecting small tumors. Retrograde ureteropyelography,
Many characteristics of TCC have been studied in an with cytologic washings at the time of tumor resection, is
attempt to predict this variable tumor behavior. These useful to further assess suspicious or poorly imaged areas.
include pathologic features, cytologic analysis, and bio- In selected cases, CT scan may be useful in the initial
logic and molecular markers. Although thorough endo- evaluation. It is more sensitive in detecting small renal
scopic tumor resection remains the principal treatment, masses, urinary calculi and nonurologic lesions. The dis-
intravesical agents have become important in the sub- advantages include inability to visualize small urothelial
group of tumors that are at risk of progression. In order lesions, cost, availability issues, and the need for contrast
to tailor treatment appropriately, the urologist should agents.
review the various prognostic factors and define the If there is suspicion of a high-grade tumor, cytologic
behavior of the tumors as precisely as possible. assessment is important. Bladder washings have a better
yield than voided urine cytologies.2 Gentle barbotage
using 50 ml of sterile saline with prompt cytology evalu-
INITIAL EVALUATION
ation is suggested.
Patient evaluation begins with a thorough history includ-
ing exposure to smoking and other known carcinogens,
ENDOSCOPIC MANAGEMENT
physical examination, and urine analysis. Endoscopic
assessment of the entire urethra and urinary bladder Well-performed transurethral resection (TUR) remains
remains the most important diagnostic procedure. the most important method for the diagnosis and
Modern flexible endoscopes with high-quality optics treatment of primary and recurrent bladder tumors,
allow this to be performed safely, with minimal patient despite the evolving role of intravesical therapy.

317
318 Part IV Bladder

Technologic improvements have dramatically improved Although close follow-up with endoscopic management
this procedure.3 of recurrences is appropriate for most low-grade Ta
Videoendoscopy has been a major advance in all areas tumors, the adjuvant use of intravesical chemotherapeu-
of endourology but particularly for bladder tumor resec- tic and immunotherapeutic agents is appropriate for
tion. In addition to minimizing the surgeon’s exposure to high-grade T1 and CIS. In order to identify patients at
body fluids, it allows all areas of the bladder, including higher risk of progression and recurrence, who may ben-
the dome and anterior wall, to be resected with greater efit from such therapy, numerous prognostic factors have
ease and improved magnification. It also provides docu- been proposed (Table 18-1). These include well-
mentation and improved teaching opportunities. established pathologic features, such as grade and stage,
Continuous-flow resection allows the urologist to con- endoscopic features, cytologic characteristics, tumor cell
trol the degree bladder filling, thus reducing the risk of products, and molecular genetic markers, such as p53.
perforation, obturator nerve excitation, and resection Although some of these prognostic factors have an estab-
time. lished clinical role, some of the newer markers remain
Adequate anesthesia is important for safe, controlled investigational.
resection, and patient comfort. In males, we prefer
beginning the procedure with an optical dilator that pro-
TUMOR GRADE
vides calibration and dilation of the urethra with visual
assessment prior to placement of the resectoscope.4 Tumor grade is well established as an important prognos-
Saline barbotage for cytologic analysis should be per- tic factor for both new tumor occurrence and progression
formed first if high-grade tumor is suspected. The entire to invasion.5,6 Mostofi et al.7 introduced a World Health
bladder should be examined using the 12-degree or 30- Organization’s (WHO) classification system for grading
degree and 70-degree lenses. The nature, number, loca- TCC as grades 1, 2, and 3 based on the degree of cellular
tion, and size of the lesions should be noted. The differentiation. The WHO/International Society of
capacity of bladder, ureteral orifices and adjacent urothe- Urologic Pathology (ISUP) consensus classification of
lium should be assessed. 1998 distinguishes between papilloma, papillary urothe-
Papillary tumors should be systematically resected lial neoplasm of low-grade potential (PUNLMP), low-
with a right angle or bladder wall loop, as appropriate. and high-grade carcinoma. A solitary papillary, tumor
The bladder wall or angled loop is designed for resecting with a fibrovascular core and no more than either seven
tumors located on the high lateral or posterior walls. The or eight normal-appearing cell layers, is regarded by
aim is to achieve complete resection of all tumors. Biopsy some as a papilloma. Others include these with grade 1
of the tumor base should be done either with cold-cup papillary carcinomas.8,9 The WHO’s grading system
forceps or formal resection, which may involve the mus- takes into account the degree of anaplasia as determined
cle. Specimens should be retrieved for histopathology. by increased cellularity, nuclear crowding, disturbance of
Suspicious mucosal areas should be biopsied with a cold-
cup forceps to minimize cautery artifact. The roller ball
electrode is used to fulgurate all suspicious areas and to Table 18-1 Prognostic Factors for Superficial
Bladder Cancer
achieve hemostasis.
A history or the presence of a high-grade tumor Number of tumors
necessitates transurethral biopsy of the prostatic urethra.
Tumor size
This is best performed with the resectoscope, as opposed
to cold-cup forceps. One should sample the mucosa and Tumor grade
underlying stroma from the bladder neck to the veru-
montanum. A bimanual examination should be per- Tumor T stage
formed. Catheter drainage is necessary if there is
Lymphatic invasion
bleeding requiring irrigation, or if a deep resection has
been performed. The majority of procedures can be per- Carcinoma in situ
formed on an outpatient or overnight basis.
Urine cytology

PATHOLOGIC PROGNOSTIC FACTORS DNA ploidy

After complete endoscopic resection, approximately 30% Blood group antigens


to 80% of patients will develop additional tumors, usually
of similar grade and stage. The majority of these lesions Tumor cell products-EGFR, AMFR, PCNA
are new occurrences, as opposed to true recurrences,
Molecular genetic markers-p53; c-erb B-2; c-myc
reflecting a generalized instability of the urothelium.
Chapter 18 Superficial Transitional Cell Carcinoma of the Bladder 319

cellular polarity, the absence of differentiation from base risk of stage progression is clearly higher for T1 tumors.
to surface, pleomorphism, variations in nuclear shape and Between 0% and 4% of Ta tumors and 27% to 46% of
chromatin pattern, mitotic counts, and the presence of T1 tumors will progress to muscle invasion at 3
giant cells.7 This pattern is reproducible at the extremes years.12,19–22 Long-term survival for Ta tumors is excel-
(grades 1 and 3) but leaves grade 2 as a heterogeneous lent. Ten-year survival rates are 95% for grade 1, 89% for
intermediate group.10,11 grade 2, and 84% for grade 3 tumors.23 Twenty-year
There is a strong correlation between higher grade cancer-specific survival for Ta tumors is 89%, compared
and invasion, disease progression, metastases, and sur- with 30% for T1 tumors.
vival.5,6 Mortality from progression of grade 1 tumors is Clearly, careful evaluation for lamina propria invasion
low (0% and 4% at 10 years).6,12 Another 3% recur as is important. Muscularis mucosa is identifiable as a land-
grade 3 tumors.1 Grade 3 tumors frequently progress to mark in 70% to 80% of TUR specimens.25 When absent,
muscle invasion, however. This has been documented in the larger arteries of the deep lamina propria are a useful
50% and 62% of cases, with a 35% 10-year actuarial sur- landmark. The surgeon should attempt to minimize
vival rate.6 cautery artifact by using cold-cup biopsies or reducing
Predicting the behavior of grade 2 tumors is less well the coagulation current. With papillary disease, the
defined. For all grade 2 tumors, between 18% and 33% tumor base should be sampled carefully, submitting the
are reported to progress, with a 10-year actuarial survival underlying tissue as a separate specimen if necessary.
of 87%.5,6 In an attempt to clarify the behavior of these The significance of the depth of lamina propria inva-
heterogeneous tumors, Carbin et al.10,11 subclassified sion has varied mainly due to difficulties in identifying
them into 2a and 2b based on nuclear pleomorphism and the muscularis mucosae. In a series with three review
the number of mitoses. When progression was assessed, pathologists, the concurrence rate for depth of invasion
grade 2a tumors faired much better, with a 92% 5-year was only 50%.26 Despite this interobserver error, two
survival compared with 43% for grade 2b. This prognos- series have used T1 subclassification and found it useful
tic discrimination has been demonstrated elsewhere.13 for prognostic discrimination. Younes et al.27 used the
Nuclear size (area) has also been used to stratify interme- presence of invasion to, and beyond, the muscularis
diate-grade tumors.14 mucosae to subclassify T1 tumors. Superficial invasion,
The major criticism of multiple subclassification for termed T1a, had a 75% 5-year survival compared with
clinical application has been subjectivity. The subclassifi- 14% for deep invasion (T1b). Using the same classifica-
cation of Carbin et al.10 was demonstrated to be highly tion system, Hassui et al.25 reported a 7% progression
reproducible, with an interobserver agreement of 90%. rate for T1a and 53% for T1b, independent of tumor
However, inter- and intraobserver errors ranging from grade, number, and size. The deep lamina propria is rich
50% to 87% have been reported for grade using reference in lymphatics, and this may explain why it is an important
pathologists in controlled trials.15 In clinical practice, most factor in progression to deep muscle invasion. The prac-
pathologists describe the tumor as low or high grade. tical role of T1 subclassification needs further study to
The categorization of grade becomes increasingly clarify its clinical application.
important for T1 tumors. This group has a much higher Vascular and lymphatic invasion in general confer a
risk of progression to muscle invasion than Ta tumors. In poorer prognosis. In a study including Ta and T1 tumors,
a series of T1 tumors, 50% of grade 3 and 22% of grade 2 7% had documented vascular or lymphatic invasion, with
tumors have progressed.12 a 30% 6-year survival. These results were independent of
grade.23 Clearly, it is important to exclude muscularis
propria invasion. If there is any doubt, the urologist
Histologic Stage
should resect the tumor site again to be certain of the
Depth of tumor invasion was first included as a marker of stage. In a series of 46 patients who had a second TUR
outcome by Jewett and Strong.16 It is now commonly within 2 weeks of their first resection, 43% demonstrated
used as a prognostic indicator and incorporated into stag- residual tumor in the deep resection.28 Given the impli-
ing systems including the current American Joint cation of muscle invasion, adequate tissue sampling to
Committee on Cancer (AJCC) system.17 include muscle is essential.
There is a significant difference in prognosis between
Ta tumors, which are confined to the mucosa (not pene-
Reproducibility of Grade and Stage
trating the basement membrane), and T1 tumors, which
infiltrate the lamina propria. This difference has been As indicated, there are limitations with the current stag-
documented for both tumor recurrence and progression. ing and grading system for superficial tumors. This has
The difference in recurrence risk between the two stages implications for tailoring treatment for an individual
is minimal. Grade, multiplicity, and size are more impor- patient, and for the analysis of prognostic factors in clin-
tant in determining the recurrence rate.18 However, the ical trials.
320 Part IV Bladder

Ooms et al.15 with seven review pathologists, reported reduced the role of mucosal biopsies. In an attempt to define
a high inter- and intraobserver error of grade. A similar the role of random biopsies in predicting recurrence and
discrepancy in stage with an intraobserver error of 50% progression, Kiemeney et al.8 demonstrated that random
has also been demonstrated.11,29–31 In reviewing a series biopsies did not significantly influence patient outcome.
of multicenter trials by tile Dutch Uro-Oncology Group, Biopsies gave no additional information to predict tumor
which utilized review pathology, the error of grading was behavior beyond stage, grade, tumor size, and number.
30%, with a tendency for local pathologists to under- Most patients with high-grade tumor will receive
grade.32 The error in T stage was 20%, with a tendency adjuvant treatment, regardless of the information from
for local pathologists to overstage. mucosal biopsies. Therefore, we infrequently perform
The same problem has been reported in interpreting mucosal bladder biopsies outside of clinical trials.
biopsies of normal-appearing urothelium in patients with
superficial tumors. In one study, biopsies reported as
ENDOSCOPIC FEATURES
“dysplasia” were reproduced only 60% of the time.33
In randomized trials, such distortion of pathologic The number of primary tumors is a strong predictor of
results is balanced in both treatment and control arms recurrence. The presence of two or more tumors
and hence does not have a major impact on the outcome increases the recurrence rate approximately 2-fold, and
of a study.32 This variation becomes more significant, decreases the time to first recurrence.5,45–48 Heney et al.5
however, when comparing results between studies of showed that the recurrence rate for multiple tumors
“similar” pathologic stage. It also has an important impli- increased from 18% to 43% for Ta disease and from 33%
cation in the management of an individual patient, par- to 46% for T1 disease. Multiple primary tumors are also
ticularly when identifying muscle-invasive disease. These associated with a tendency for increasing grade with
limitations in staging and grading must be recognized. recurrence.48 There is an approximately 1.5-times
The important lesson is for the clinician to review the increased risk of progression for multiple tumors.12,45
slides with the pathologist prior to any major treatment Tumor size is also a predictor of recurrence and progres-
decision. sion. The risk of stage progression increases from 9% to
25% for tumors >5 cm.5
The time to first recurrence is also prognostic. A
Status of Bladder Mucosa Distant from the Tumor
recurrence or new occurrence at 3 months has been
Any abnormality of the mucosa distant from the primary shown to increase the risk of subsequent recurrence from
tumor provides prognostic information on recurrence 20% to between 70% and 85%.47,48 This emphasizes the
rates. Recently, there has been increasing support for importance of the disease status at the first follow-up
tumor cell implantation as one explanation for high cystoscopy.18
recurrence rates.34,35 In general, any abnormal-appearing
mucosa should be biopsied. A cold-cup technique is
CYTOLOGIC PROGNOSTIC FACTORS
preferable, as it allows adequate tissue sampling without
Urine Cytology
diathermy artifact. More controversial is the role of
biopsy of normal-appearing mucosa. The incidence of Urinary cytology performed on exfoliated urothelial cells
associated changes in normal-appearing mucosa is in urine or bladder washings can be used to detect TCC.
between 10% and 50%.5,36,37 Dysplasia is reported in Care must be taken with specimen collection and han-
approximately 15%, with carcinoma in situ more variable dling to preserve exfoliated cells and avoid bacterial con-
(10% to 50%).37In general, CIS is more common with tamination.
grade 3 tumors.37 The incidence of concurrent CIS in The yield for diagnosis of low-grade tumors is low,
the prostatic urethra is reported to be between 16% and generally <30%.50 Recognition of subtle malignant fea-
30%.39,40 We believe that the magnification associated tures requires a skilled cytopathologist. However, cytol-
with videoendoscopy lessens the likelihood of CIS being ogy is highly sensitive for high-grade disease for both
found in normal-appearing bladder mucosa. CIS and papillary tumors. Greater than 60% of papillary
Less well defined is the accuracy and reproducibility tumors and 90% of cases of CIS have positive cytology.
of dysplasia reported in a biopsy, and the true risk of pro- The reduced cellular adherence of high-grade tumors
gression for dysplasia.41–44 Progression for Ta and T1 results in a higher proportion of cells shed into the urine
tumors has been shown to increase from 8% to 30% and thus detectable with cytology. This complements
when moderate or severe mucosal dysplasia was also cytoscopy in the detection of high-grade TCC. Bladder
present on biopsy.5 Others have suggested that moderate wash cytology may provide more information on the
dysplasia like mild dysplasia is not a risk factor.41 presence or absence of CIS than random biopsies. It is
More widespread use of cytology and improved endo- important to provide relevant clinical history for accurate
scopic optics, particularly video magnification, have interpretation by the cytopathologist.
Chapter 18 Superficial Transitional Cell Carcinoma of the Bladder 321

In addition to identifying the method of specimen improved sensitivity and specificity64 They have not as
collection, it is important to note any history of yet found a role in clinical practice.
previous treatment (radiation, chemotherapy, or
immunotherapy), urinary infection, stones, recent sur-
gery, or catheterization. All of these conditions may Tumor Cell Products
produce cells with nuclear features that may be con- There have been numerous cell proteins that appear to
fused with malignancy.57 correlate with tumor behavior. They perform various
The interpretation of a positive urine cytology functions but often act as cell receptors. These include
requires knowledge of tumor status of the bladder. The desmosomal glycoprotein,65transferrin receptor,66 and
absence of cancer in the bladder requires exclusion of human milk fat globulin.67 Epidermal growth factor
CIS or high-grade disease in the upper tracts or prostatic (EGF) receptor and basement membrane tissues have
urethra. Cytology also has a valuable role in follow-up of been more extensively studied.
high-grade superficial tumors. EGF receptor is reported to be an independent pre-
dictor for stage progression in Ta and T1 tumors.68 In
DNA Ploidy addition, an increased recurrence rate from 7% to 17%
has been noted for receptor-positive patients.69 This
DNA ploidy as assessed by flow cytometry has been used marker would appear to have potential clinical applica-
to assess TCC. In general, all grade 1 and most of the tion on the basis of results to date.
grade 2 tumors are diploid. Some grade 2 tumors are Various connective tissue components of the basement
tetraploid, while others, and most of the grade 3 tumors membrane have been studied as potential markers. The
are nontetraploid aneuploid. Variation in DNA content loss of laminin and type IV collagen in the basement
within a tumor is a significant limitation in the applica- membrane underlying noninvasive tumors correlates
tion of ploidy studies. with increasing tumor progression.70
There is a correlation between ploidy and tumor
behavior. Of 229 grade 1 and grade 2 superficial tumors,
Gustafson et al.53 showed that none of the diploid tumors Cytogenic and Molecular Genetic Markers
progressed, while 35% of the aneuploid tumors pro- Although the majority of invasive bladder tumors com-
gressed with a 21% death rate. Aneuploid tumors have monly contain nondiploid and morphologically abnor-
also been shown to recur more frequently.54 mal DNA, most superficial tumors are near diploid.
There is conflicting evidence as to the ability of ploidy Sandberg71 first identified abnormal DNA in Ta and T1
to independently predict progression over grade tumors, and demonstrated a relationship with risk of
alone.55,56 Ploidy remains expensive, and has yet to estab- recurrence and progression. Chromosomal analysis for
lish a clinical role. We do not use this in decision-making. solid tumors has been simplified with the development of
specific probes, such as fluorescent in situ hybridization
BIOLOGIC PROGNOSTIC MARKERS (FISH) for DNA sequence analysis. RNA amino acid
Blood Group Antigens sequencing with polymerase chain reaction (PCR) has
also contributed to the understanding of TCC. Both
There has been long-standing recognition of the prog- FISH and PCR can be performed on paraffin sections.
nostic importance of the loss or absence of blood group Specific chromosomal abnormalities for bladder can-
antigens from the tumor cell surface.57,58 This can be cer have been identified on chromosomes 1, 5, 7, 9, 11,
assessed by immunohistochemistry or red-cell adherence and 17.72–75 Abnormalities of chromosome 7 include
tests. However, the results are usually qualitative and the alteration to the c-erb B oncogene, which codes for the
methodology subjective. The result is also dependent on EGF receptor.
the patient’s blood group, with group O being prone to Loss of heterozygosity at chromosomes 9q, 11p, and
false-negative results. An increased risk for progression 17p correlates with known tumor suppressor genes,
has been documented with loss of antigen expression.58 namely, the Wilms’ tumor gene (WTG) and p53. Studies
This information appears to be independent of grade and on the role of oncogenes and carcinogenesis have tradi-
stage.59 There is also an association with tumor recur- tionally used rodent tumors, particularly the ras fam-
rence rate60 and response to bacille Calmette-Guérin ily.78–80 Unfortunately, it has not proven useful in the
(BCG) treatment.61 study of human TCC.
Other cell surface antigens have also been correlated Promising results have been demonstrated through
with prognosis. These include the Lewis surface antigens molecular genetic work with oncogenes. The p53 onco-
and the Thompson-Friedenreich antigen.62,63 Advances gene (17p13-1 locus) codes for a nuclear phosphoprotein,
in immunostaining with monoclonal antibodies have whose major role appears to be in transcriptional regula-
allowed assessment of these antigens in urine, with tion. The p53 is involved in DNA repair or induction of
322 Part IV Bladder

apoptosis for irreversibly damaged DNA. Hence, it is been resected and urinary cytology, if performed, is neg-
thought to function as a tumor suppressor gene.86 There ative. Intravesical therapy can be subdivided into
are two forms of this gene product—the wild, or unal- chemotherapy and immunotherapy. The decision to use
tered type, and the mutant form.82 Mutation of wild type intravesical treatment is dependent on numerous factors,
results in the accumulation of mutant p53 protein within including: tumor stage, grade, size, and multiplicity; the
the tumor cell nucleus, due to a prolonged half-life. This presence of CIS or positive urinary cytology; the side-
leads to alterations in modulating activity.83 Mutant p53 effect profile of each agent; and other patient-specific
can be easily detected immunohistochemically using factors (Table 18-2). Economic factors may also play a
monoclonal antibodies on fresh or fixed tumor speci- part in the decision process.
mens.84 Immunohistochemical changes have been shown The ideal intravesical agent would have an antitumor
to correspond to abnormalities with DNA sequence effect against TCC, show no phase specificity in the cell
analysis.85–88 It must be recognized that the limitation of cycle and have limited systemic and local toxicity on an
monoclonal immunohistochemistry is false-negative acute and chronic basis. Much of the systemic toxicity
results,89 although this is reduced with the monoclonal associated with intravesical therapy is due to drug
antibody Pab-1801.89 absorption. Factors associated with increased absorption
Changes in p53 are the most commonly recognized include low molecular weight of the instilled drug, tem-
genetic alterations in human malignancy, and correlate poral relationship to the TUR, and extent of resection.
with tumor behavior.90 Mutations have been observed in Local toxicity with intravesical therapy is frequent and
50% of high-stage bladder tumors.85–87,91 usually consists of irritative voiding symptoms or hema-
However, p53 changes and their relationship to tumor turia.104 Delaying the initiation of intravesical therapy for
behavior in superficial tumors are less clear. Studies have 10 or more days after resection will allow healing of the
utilized both immunohistochemistry and nucleic acid resected urothelium and may lessen the local and sys-
sequence analysis for assessment of Tis, Ta, and T1 temic side effects.
tumors. Mutations have been observed in up to 65% of Increasing exposure of the urothelium to the intraves-
biopsies of primary Tis (pTis).92 Positivity for p53 has ical agent would seem to be important. This can be done
been demonstrated as an independent predictor of by increasing the concentration and contact time of the
progression in a series of 33 pTis patients followed for drug. Some advocate asking the patient to lie in the
124 months.89 prone position for part of the treatment.105 Others sug-
The results for Ta and T1 diseases are more variable. gest that sterile water, instead of saline, as the diluent
Positive mutant p53 staining has ranged between 8% and decreases the osmolality of the solution, thereby increas-
95%,85,93–97 with a strong correlation between positivity ing the intracellular concentration of the drug.106
and tumor grade.94,95,98 Several recent institutional Recommended dosages, administration schedule, and
reviews of Ta and T1 tumors utilizing multivariate analy- duration of intravesical therapy have varied and are
sis have demonstrated p53 status as a statistically signifi- largely based on empiric data. Patients who receive
cant prognostic factor for progression-free and overall intravesical therapy should be monitored for response.
survival.99,100 The disparity between numerous investiga- This includes endoscopy and bladder wash cytology.
tors is difficult to explain. Technical factors may play a Appropriate end-points include recurrence, time to
role. In summary, the oncogene p53 is an encouraging recurrence, progression in grade or stage, and time to
prognostic marker for bladder cancer. Its clinical role in progression. Patients are monitored for response 3
superficial disease will remain unclear until further large months after initiation of treatment. A complete
series with multivariate analyses are performed. The response (CR) to treatment is defined as no tumor on
overexpression of other oncogenes, c-erb B-2 and c-myc, endoscopy, negative cytology, and negative bladder biop-
has also been observed and is more frequently in high-
grade, high-stage TCC.101–103 Their role in superficial
TCC has not been defined. Table 18-2 Indications for Intravesical Therapy
Cumulative tumor size >5 cm

INTRAVESICAL THERAPY Multiple tumors


Rationale
Multiple recurrences
The high incidence of subsequent tumors after initial
TUR, whether true recurrences or new occurrences, has Stage T1 or high-grade TCC
led to the use of intravesical instillation of antineoplastic
CIS or positive cytology
agents. This form of treatment can be used either thera-
peutically, to eradicate residual tumor after an incom- Residual tumor
plete TUR, or prophylactically, after all visible tumor has
Chapter 18 Superficial Transitional Cell Carcinoma of the Bladder 323

sies. Anything less than a CR is considered as a treatment Soloway and Ford116 found that only 4% of patients suf-
failure, as partial responders have the same incidence of fered this side effect and none of the consequences were
progression as nonresponders.107 severe. Irritative voiding symptoms are common as with
If any question exists as to the persistence of tumor, other intravesical agents. Overall, thiotepa is relatively
resection or biopsy is performed. In the face of a positive safe and inexpensive, and is most effective for low-grade
biopsy or cytology after intravesical treatment, it is less tumors. We believe that it is a reasonable choice for pro-
likely that another course with the same agent will be phylaxis in patients with multiple, recurrent grade 1, Ta
effective, and an alternative treatment should be used. bladder tumors.
BCG may be an exception, as a second 6-week course is
beneficial in some instances. The stage and grade of the
tumor recurrence will ultimately be the determinant of Mitomycin C
future treatment (e.g., cystectomy, TUR alone). Mitomycin C (MMC) is a 329-kDa antitumor antibiotic,
which inhibits DNA synthesis. There has never been a
Intravesical Chemotherapy thorough dose response study with MMC. Most studies
Thiotepa have used from 20 to 40 mg in 20 to 40 ml of water,
weekly, for 8 weeks. Many studies have used MMC for
Thiotepa is an antineoplastic alkylating agent related to residual TCC after incomplete resection (i.e., treatment
nitrogen mustard that has been shown to eradicate exist- rather than prophylaxis). Most patients in these studies
ing tumors and delay the development of new tumors. were at high risk for recurrence, having a history of prior
The usual dosage is 30 to 60 mg in 30 to 60 ml of distilled TCC. A study by the National Bladder Cancer Group
water. The National Bladder Cancer Study Group found involved treatment with 40 mg of MMC for 8 consecutive
that the CR rate for 30 mg was equal to that for 60 mg. weeks.117 The study included 117 patients with CIS or
Hence, the lower dose is recommended.12 Duration of grade 1 to grade 3 Ta or T1 disease. Each patient had
treatment is usually weekly for 6 to 8 weeks when given failed a course of intravesical thiotepa. At 3 months, 27%
therapeutically and weekly for 4 to 6 weeks when given pro- of patients had a CR as defined by negative endoscopy,
phylactically. If maintenance is given, a monthly schedule biopsy, and cytology. Another 9% had negative endoscopy
is usually selected. A randomized cooperative study by and cytology but were not biopsied again. Despite a neg-
the Medical Research Council found no statistically ative endoscopy, an additional 12% were not considered
significant difference in tumor recurrence rates in as complete responders, since their cytology was positive.
patients receiving: (1) no intravesical therapy, (2) a single Overall, 18% of patients with T1 disease, 29% with Ta
instillation of thiotepa at the time of resection, or (3) a disease, and 35% with CIS were rendered tumor free.
single thiotepa instillation at the initial resection and The National Bladder Cancer Collaborative Group
3-month intervals for a total of 5 treatments in a year. also compared MMC and thiotepa for treatment of
Median follow-up was almost 9 years. Thus, it appears TCC.118 Study participants received either 40 mg of
that it is necessary to give more than 5 treatments of MMC in 40 ml or 30 mg of thiotepa in 30 ml of sterile
thiotepa in a year to make a significant difference in water, weekly, for 8 weeks. MMC patients had a statisti-
tumor recurrence. cally significant higher CR rate (39% versus 27% with
When used as a therapeutic agent, CR rates of 35% to thiotepa, p = 0.02). Ta grade 1 tumors responded best as
45% have been reported.14,115 Long-term use of thiotepa in previously reported thiotepa studies.
prophylaxis was evaluated by the National Bladder Soloway119 reported a series of 80 patients who
Cancer Collaborative Group.112 They reported that 53% received 8 weekly 40-mg instillations of MMC for treat-
of patients were tumor free at 2 years compared to only ment with evaluation for response at 12 weeks. All com-
27% of untreated patients. The benefits were only noted plete and some partial responders received monthly
in patients with grade 1 lesions (51% treated with MMC for 1 year. Average follow-up was 40 months.
thiotepa were tumor free at 2 years versus 14% of the Complete responses were noted in 37% (15 of 41) of Ta
control group). Recurrence rates of grade 2 and grade 3 patients, 33% (7 of 21) of CIS patients, and 44% (8 of 18)
tumors were not significantly different than controls, of T1 patients. Twenty-six percent of patients eventually
suggesting that thiotepa is most effective in treating had a cystectomy and 15% developed muscle invasion.
patients with low-grade tumors. Nine percent (7 of 80) of the patients eventually died of
Due to thiotepa’s low molecular weight (189 Da), it is bladder cancer.
absorbed more than any other intravesical agent. The initial response to MMC was predictive of the
Myelosuppression occurs in up to 9% of patients, usually patient’s eventual outcome. Of those who had persistent
in the form of leukopenia or thrombocytopenia. Thus, TCC at 3 months, 34% later required cystectomy com-
white blood cell and platelet counts must be monitored pared to only 13% of initial complete responders. Death
during the course of therapy. In a study of 670 patients, from carcinoma of the bladder occurred in 12% of early
324 Part IV Bladder

treatment failures compared to only 3% of initial com- There have been a number of randomized studies
plete responders. It thus may be prudent to consider cys- using doxorubicin for prophylaxis. Niijima et al.125
tectomy for patients with high-grade TCC who do not reported findings from a randomized study of doxoru-
have a CR with intravesical treatment. bicin (30 mg in 30 ml, twice a week, for 4 weeks) plus
Huland et al.120 reported a randomized prospective TUR versus TUR alone. At a minimum 12 months’ fol-
study of MMC used for prophylaxis. Patients received low-up, 70% (104 of 149) of patients receiving doxoru-
either 20 mg of MMC in 20 ml every other week for up bicin were recurrence free compared to 55% (77 of 139)
to 18 months or had a TUR alone. Only 10% of MMC- who had a TUR alone. Kurth et al.126 found that 64% (58
treated patients recurred compared to 51% of controls. of 86) of patients randomized to receive doxorubicin
Maier and Hobarth121 reported a study of 63 patients (50 mg in 30 ml, weekly, for 1 month then monthly for
who received MMC prophylaxis over a 2-year period. 1 year) in addition to TUR were recurrence free versus
The mean follow-up was 50 months. Fifty-two percent 52% (39 of 69) in the control group. Doxorubicin was
developed a recurrence. One-third (21 of 63) died while not shown to be superior to thiotepa for prevention of
receiving MMC. Of the remaining 42 patients, 26% superficial tumor recurrence in other controlled double-
recurred at an average of 14 months after MMC was dis- blind studies.127,128
continued. The remaining 31 patients (48% of the origi- In a randomized trial comparing the efficacy of dox-
nal 63) remained tumor free for a mean 26 months after orubicin and BCG for CIS, Lamm et al.129 reported a
MMC was completed. 34% CR rate for doxorubicin. Median length of time to
The Medical Research Council has reported the failure was 5 months. BCG was superior for treatment of
7-year follow-up of a 502-patient randomized multicen- CIS. Others have demonstrated up to a 70% CR rate
ter trial involving the use of early instillation of MMC. with doxorubicin, but follow-up was short.130
After complete resection of Ta and TI tumors, patients Due to doxorubicin’s high molecular weight, absorp-
were randomized to receive either no further treatment tion is low and myelosuppression rare. Cystitis is the pri-
until the next endoscopy, a single instillation of 40 mg mary toxicity, occurring in 25% of patients.104,124,131
MMC within 4 hours of resection, or an instillation Diminished bladder capacity occurs in up to 9% of
within 24 hours of resection and at 3-month intervals for patients and anaphylactic reactions have been seen.124,130
1 year (5 total doses). They found that the overall recur- Overall, doxorubicin has been shown to be an effective
rence rates were lower and the interval to recurrence agent for treatment and prophylaxis of TCC.
prolonged in patients receiving 1 or 5 instillations of
MMC compared to controls. The estimated 5-year
Valrubicin (AD-32)
reduction in recurrence risk was 5% and 23% for the
1- and 5-dose regimens, respectively.122 AD-32 (N-trifluoroacetyladriamycin-14-valerate), an
Due to MMC’s molecular weight, systemic toxicity is anthracycline derivative, is a semisynthetic analog of dox-
rare. Local symptoms, primarily chemical cystitis, are orubicin. AD-32 (MW 723) differs from doxorubicin
relatively common (10% to 17%).117,120,123 A desquamat- (Adriamycin) in that it lacks cardiotoxicity, it is associated
ing rash of the palms and genitalia is unique to MMC. It with less local toxicity, and it is lipophilic. AD-32 does
occurs in approximately 5% of patients and is thought to not bind DNA but is an active inhibitor of DNA and
be a form of contact dermatitis. Occasionally, the rash is RNA syntheses.
diffuse. Severe bladder contracture requiring cystectomy Studies have focused on treating patients with CIS or
(4%) has also been reported myelosuppression is superficial disease who failed with BCG treatment. Phase 1
rare.104,119,124 trials of patients treated with AD-32 have been com-
In summation, MMC appears to be more effective pleted. Of the 35 courses given, 69% had some local tox-
than thiotepa. The toxicity of MMC is acceptable. The icity, 31% grade 2 or grade 3. Irritative voiding
widespread use of MMC as a first-line therapy, however, symptoms accounted for all but three adverse events.
is limited in the United States by its cost. Reducing the alcohol content in the diluent decreased
these symptoms. There was negligible systemic absorp-
tion.133 Valrubicin has been approved by the FDA for
Doxorubicin
treatment of BCG refractory carcinoma in situ, and fur-
Best known as a systemic chemotherapeutic drug, this ther clinical trials in this group are in progress.
antitumor antibiotic has been shown to be an active intrav-
esical agent. Doxorubicin is a 380-kDa intercalating agent.
Immunotherapy
The vast majority of the literature comes from Japan,
Bacille Calmette-Guérin
where doxorubicin is the most frequently used intravesical
agent. The doses varied widely from 10 to 100 mg at var- Since BCG was initially identified as an effective intrav-
ious intervals. Therefore, there is no standard dose. esical agent for superficial bladder cancer, much has been
Chapter 18 Superficial Transitional Cell Carcinoma of the Bladder 325

learned about its actions. Still more remains to be eluci- follow-up of 48 months. Only 117 of responders subse-
dated.134 BCG is a live attenuated tuberculosis organism quently required cystectomy versus 55% of nonrespon-
first developed from cultures at the Pasteur Institute of ders. However, no survival difference was noted between
Lille.135 Its mechanism of action remains ill defined, but responders and nonresponders. Lamm et al.129 in the
at least part of its effectiveness is due to an immunologic aforementioned randomized study of BCG versus dox-
host response. T-cell-deprived animals do not respond to orubicin for CIS, reported a 70% CR rate for BCG ver-
BCG. Furthermore, BCG must bind to urothelial cells to sus 34% for doxorubicin. The mean interval to
be active and binds by attaching to fibronectin.136 An treatment failure was 39 months for BCG compared to
immunologic cascade of events occurs causing a strong 5 months for doxorubicin.
inflammatory response. The inflammatory response itself
may have a deleterious effect on tumor cells. In addition,
BCG for Prophylaxis
various cytokines are released including interleukins,
which have known antineoplastic activity.134,136 Several randomized studies have compared the efficacy of
The urologic literature is replete with reports on the TUR alone versus TUR plus BCG. Herr142 recently
efficacy of BCG. There is marked variability. This vari- summarized the results of 5 such studies encompassing
ability may be due to differences in the completeness of 437 patients. Overall, 70% of patients who received BCG
resection, tumor stage and grade, prior history of TCC, prophylaxis were tumor free compared to 31% of
prior intravesical therapy, associated CIS, the number of patients who had TUR alone. Follow-up ranged from 12
instillations, the BCG substrain used, host immunologic to 60 months.
factors, and length of follow-up. To address the impact of BCG on disease progression,
The optimal schedule for BCG administration has yet Herr et al.143 reviewed the 10-year follow-up of 86
to be established. The number of milligrams per colony patients with recurrent Ta, T1, and Tis diseases, who
count differs among the numerous BCG substrains. The were randomized to either TUR plus 6 weeks of Armand-
concept of 6 weekly instillations is arbitrary. An induc- Frappier BCG or TUR alone. Crossover was available for
tion phase is necessary for the development of the patients in the TUR group who recurred. The 10-year
immunologic response in the bladder. While most progression-free and overall survival were 62% and 75%,
patients develop an inflammatory response with 6 instil- respectively, for patients who received BCG and 37% and
lations, some will require fewer and some may require 53% for patients who had a TUR alone. The median pro-
more.137 Studies have shown that 19% to 26% of patients gression-free survival was not reached for the BCG group
treated, who do not respond to an initial 6-week course and was 46 months for the “control” group. Fifteen of 18
of BCG, will respond to 6 additional weekly doses.108,109 patients who crossed over and received BCG did not have
Maintenance therapy is also controversial. Early studies tumor progression. The authors concluded that BCG
showed no clear benefit to maintenance BCG to justify delayed both tumor progression and death in patients
the increased risk of side effects.137 However, Lamm with superficial bladder cancer.
et al.129 recently reported results of a randomized, BCG has also been compared to the most commonly
prospective Southwest Oncology Group’s (SWOG) trial used intravesical chemotherapeutic agents. Brosman144
of maintenance BCG for CIS, Ta, and TI diseases. After and Marfinez-Pineiro,145 in separate studies, found BCG
a 6-week induction course, patients received BCG in to be superior to thiotepa in reducing tumor recur-
3 weekly doses at 3 months, 6 months, and every 6 months rence.109 Lamm et al.146 reported the results of a
for 3 years. In this study, maintenance therapy further SWOG’s comparison of BCG and doxorubicin in
reduced the tumor recurrence rate.138 However, the opti- patients with rapidly recurring superficial TCC. The
mal timing of these maintenance doses whether monthly, mean interval to recurrence in the patients with papillary
3-monthly, or even yearly has not been established. tumors receiving BCG was 22 months versus 110 months
in patients receiving doxorubicin.
Vegt et al.147 recently reported the results of a ran-
BCG for CIS
domized prospective study of 435 patients with pTa or
The literature strongly supports the use of BCG for pT1 disease comparing the efficacy of TICE BCG,
treatment of CIS. CR rates of 70% have been RlVM-BCG, and MMC. Mean follow-up was 36
reported.139–142 Dejager et al.141 reported on 123 months. Patients underwent TUR followed by either 6
patients from 6 phase 11 studies; patients received at weeks of TICE or RIVM-BCG or 4 weeks of MMC fol-
least 6 weekly instillations of TICE BCG and 12 lowed by monthly doses for 6 months. Results are shown
monthly maintenance doses. A 76% complete remission in Table 18-3. They concluded that MMC was equivalent
rate was reported, including a 71% (45 of 63) CR rate to BCG in efficacy. Drug-induced cystitis, the most com-
for patients who failed intravesical chemotherapy. A mon local toxicity, and systemic side effects were signifi-
durable CR was seen in 50% of responders at a mean cantly less in the MMC group.
326 Part IV Bladder

Table 18-3 Randomized Trial of TICE and RIVM BCG and MMC
TICE BCG RIVM-BCG MMC

Recurrence Ta/TI 75 of 117 (64%) 62 of 134 (46%) 58 of 136 (43%)

Recurrence CIS 17 of 23 (74%) 9 of 15 (60%) 8 of 12 (67%)

Progression at recurrence 7 (5%) 8 (6%) 8 (6%)

From Vegt PDJ, Witjes JA, Wities WPJ, et al: J Urol 1995; 153:929, with permission.

In regard to T1 disease, Eum et al. reported a series of be used if the patient is septic. In addition, the coadmin-
30 patients with high-grade T1 disease treated with a istration of antituberculin agents with BCG to lessen side
6-week course of Armand-Frappier BCG. After one effects of treatment has been proposed.150
course, 47% (14 of 30) had a negative cytology and
biopsy at 6 months. Another 6 patients responded to a
Interferon
second 6-week course, for an overall CR rate of 66%.
Four patients required a cystectomy for progression or Interferons (IFN) are biologic response modifiers that
recurrent TI tumor, and one patient had metastasis. All are integrally associated with the immunologic cascade.
had failed a first course of BCG. Thus, the overall pro- IFN has antiproliferative and antiviral properties. It stim-
gression rate was 17%. ulates cytokine release and inhibits nucleotide synthesis.
Cookson and Sarosdy148 studied BCG for T1 disease It stimulates macrophage function, enhances natural
in 86 patients. Patients underwent TUR prior to receiv- killer (NK) cell cytolytic activity, and activates B and T
ing a 6-week course of Pasteur BCG. Some patients lymphocytes.151
received booster doses at 3, 6, and 12 months. Patients The success of IFN-α-2b has been limited in several
with recurrence had a repeat TUR followed by additional previously reported series of superficial TCC. The
BCG either weekly or monthly. At a median follow-up of Northern California Oncology Group reported that 4 of
59 months (range 9 to 149), 9170 were disease free. Of 16 patients (25%) with superficial TCC had a CR with
the 91%, 69% responded to the initial induction course intravesical recombinant IFN-α-2b. In the same study, 6
and 22% were recurrence free after reresection and addi- of 19 patients with CIS had a CR. Of the 10 CRs in the
tional BCG. Progression to T2 disease occurred in only study, 5 were durable at 18 to 37 months.153
7% of patients. They concluded that BCG was effective Glashan154 reported on a series of 87 patients with
in the treatment of T1 tumors. Some patients in their CIS. Patients received either high-dose (100 million
study, however, were listed as having grade 1, T1 tumors. units) or low-dose (10 million units) IFN-α-2b, weekly
Grade 1, TI disease rarely, if ever, exists. This under- for 12 weeks, then monthly up to a year. In the high-dose
scores the importance of pathology review. group, 20 of 47 (43%) had a CR compared to only 2 of
Nadler et al.149 reported that 23 of 66 (35%) patients 38 (5%) in the low-dose group. Interestingly, 6 of 9
who were disease free 2 years after treatment with BCG patients who failed a course of BCG had a CR with IFN-
had tumor recurrence after 2 to 11 years of follow-up. α2b. Seven patients in each group eventually required a
Thus, lifelong monitoring after BCG is essential even cystectomy, 13 for disease progression. The median
with an excellent initial response to treatment. interval to cystectomy was 32 weeks for the high-dose
Cystitis is the most common side effect of BCG, group and 18 weeks for the low-dose group. The primary
occurring in up to 90% of patients. Hematuria occurs in toxicity was a flulike syndrome occurring in 17% of high-
up to one-third of patients and can be problematic. dose and 8% of low-dose patients. Local symptoms did
Major adverse reactions include fever over 103 ˚F (3%), not occur and no patient discontinued therapy due to
granulomatous prostatitis (0.9%), and pneumonitis or side effects. Though the efficacy of IFN is inferior to
hepatitis (0.7%). BCG sepsis, the most serious complica- BCG, it is being used more commonly in BCG failure
tion, occurs in 0.4% of patients. Thus far, 10 deaths from patients.
BCG sepsis have been reported. Caution should be used
in administering BCG to patients who are immunocom-
Keyhole-Limpet Hemocyanin
promised or have liver disease. Treatment depends on the
clinical situation but includes isoniazid 300 mg and Keyhole-limpet hemocyanin (KLH) is a high molecular-
rifampin 600 mg daily. In advanced cases, ethambutol weight immunogenic protein collected from the
1200 mg daily is useful. Cycloserine 250 to 500 mg twice hemolymph of the mollusk Megathura crenulata. In
daily in combination with isoniazid and rifampin should humans, KLH induces both cell-mediated and humoral
Chapter 18 Superficial Transitional Cell Carcinoma of the Bladder 327

responses. KLH was initially used as a skin test to evalu- and subsequent 6-week BCG instillation. The immediate
ate delayed-type hypersensitivity responses in humans.152 instillation of MMC decreases the tumor cell implanta-
Olsson et al.155 first reported the use of KLH tion and BCG should further reduce the recurrence and
immunotherapy for superficial TCC of the bladder. progression rate.190
Patients immunized with KLH were noted to have fewer
tumor recurrences. In a prospective trial of 19 patients
Recommendations
with a history of superficial TCC, prophylactic KLH
injections were given after TUR. Of the 9 immunized From the above data it is clear that BCG plus TUR is
patients, only 1 had a recurrence over 204 patient-months. more beneficial than TUR alone for patients with high-
In the nonimmunized group, 7 of 10 had a recurrence grade TCC and T1 disease. Furthermore, BCG has been
over 228 patient-months. shown to prolong the interval to progression, while
In a randomized controlled study, Jurinic et al.156 intravesical chemotherapy has not. BCG or MMC
found that KLH was more effective than MMC in pro- appears to be the agent of choice for CIS.
phylaxis against recurrent TCC. KLH (10 mg) was given For recurrent, low-grade Ta tumors, the optimal
intravesically following a 1-mg intracutaneous dose. treatment is less clear. The likelihood of progression to
Twenty-three patients received 20 mg of intravesical muscle-invasive disease is very low (<5%). We recom-
MMC. Fourteen percent of KLH patients recurred com- mend either no adjuvant therapy or intravesical
pared to 39% in the MMC group. In a subsequent non- chemotherapy. In this situation, the role of intravesical
randomized single arm study, Jurinic et al. reported that chemotherapy would be to prolong the tumor-free inter-
17 of 81 patients (21%) had recurrence of TCC after the val and decrease the cost and morbidity of frequent sur-
same KLH regimen. No adverse effects were reported.152 gical procedures. Some studies show no significant
difference between BCG and chemotherapy for low-
grade, low-stage disease,139 and as described above, the
Bropirimine
side-effect profile of BCG is not insignificant. Thus, we
Bropirimine is a pyrimidine that induces interferon, pre- do not suggest BCG for grade 1 TCC.
sumably leading to antineoplastic and immunomodulatory For grade 2 or grade 3 Ta tumors treatment should be
activities.157 Bropirimine is a unique agent for urothelial individualized based on recurrence patterns. Either BCG
neoplasms, since it can be given orally in a phase 1 trial for or intravesical therapy may be appropriate in these situa-
treatment of CIS and papillary TCC; Sarosdy et al.158 tions. If intravesical chemotherapy fails, BCG would be a
reported that 6 of 26 patients who could be evaluated had reasonable next step. If a patient were to fail a course of
a CR at 3 months. Five of 11 patients with CIS had a CR. BCG, a second 6-week course could be considered. This
The drug was most effective when given at high doses, 3 may help avoid the morbidity of cystectomy in a sub-
g/day for 3 consecutive days, weekly for 12 weeks. Only 1 group of patients. Yet, this delay may allow disease pro-
of 10 papillary tumors regressed completely. gression and loss of curability with radical treatment.
In a subsequent phase 2 trial using high-dose bropir- Herr et al.160 reported a series of 61 patients with high-
imine, 20 of 39 patients (51%) with CIS had a CR. This grade superficial bladder cancer who were initially
included 6 of 13 patients (46%) who were BCG failures. treated with BCG. At a follow-up ranging from 10 to 13
CRs lasted as long as 17 months. Flulike symptoms were years, 12 patients (20%) died of metastatic urothelial can-
the most common adverse effect occurring in 62% of cer. Thus, in many cases, despite diligent follow-up,
patients.159 Further phase 2 trials of bropirimine alone patients progress and die of bladder cancer.
and in combination with BCG are in progress. Biologic immune response modifiers other than BCG
have been used to treat TCC. These cytokines can be
administered orally or intravesically. Their mechanisms
Combination Therapy
of action are not clearly defined. A host response appears
Recent studies have shown that BCG and IFN-α have to be central to their efficacy. Further elucidation of the
complementary and synergistic immunomodulatory and mechanism of action of these agents, as well as controlled
antitumoral activity. This allows reduction in the BCG studies of various drug combinations at various doses,
dose used. This combination is efficacious in patients will better define their role in the treatment of Ta and TI
who failed BCG therapy. O’Donnell et al.189 reported TCC and CIS.
63% and 53% disease-free rates at 12 and 24 months in
BCG refractory patients.189
TRANSITIONAL CELL CARCINOMA
Clinical studies suggest that combination of BCG and
OF THE PROSTATIC URETHRA
MMC is not advantageous over mitomycin alone. In
high-grade tumors, complete resection of the tumor TCC of the prostatic urethra is usually found concomi-
should be followed by immediate instillation of MMC tant with or subsequent to TCC of the bladder. It is
328 Part IV Bladder

rarely primary. Analysis of cystoprostatectomy specimens prostatic urethra, several series have documented CRs
has identified TCC of the prostate in 10% to 50% of using BCG.168,169
cases. Involvement is more common with high-grade Cystoprostatectomy is recommended for most cases
bladder tumors, occurring in 70% of men with primary of TCC involving the prostatic ducts or stroma.162
CIS of the bladder. Since the presence of TCC of the Concurrent urethrectomy is recommended because
prostate may alter the treatment strategy, a TUR biopsy there is a 30% to 50% risk of urethral recurrence.171
of the floor of the prostatic urethra should be performed The prognosis for urethral involvement relates to dis-
in all men with high-grade TCC of the bladder.162 ease stage, with high-T-stage lesions associated with a
The management and prognosis of TCC of the pro- grim outcome.
static urethra depend on stage and grade. Although the
UICC staging system indicates that all TCC of the
PARTIAL AND RADICAL CYSTECTOMY
prostate is stage T4, it seems prudent to substratify
patients based on involvement of the prostatic urethra, With the current efficacy of quality endoscopic instru-
ducts, or stroma.165 The prognosis for prostatic mucosal ments and multiple intravesical agents, the majority of
CIS and ductal involvement differs from that of stromal patients with superficial bladder cancer will not require
invasion. Furthermore, TCC involving the prostatic ure- radical surgery. The indications for cystectomy include
thra primarily may be less lethal than the disease from failed intravesical therapy for high-grade disease (papil-
the bladder infiltrating through the bladder wall to lary or CIS), persisting or recurrent high-grade tumor, or
involve the prostate. progression to bladder muscle or prostatic stromal inva-
Treatment of TCC of the prostatic urethra must take sion (Table 18-4). Persistently positive urine cytology
into consideration the status of the bladder (i.e., current after intravesical treatment requires a thorough reevalu-
TCC or prior history of TCC and treatment). ation of the bladder, in addition to reassessment of the
Therapeutic options include intravesical chemotherapy, upper tracts and the prostatic urethra in men.
immunotherapy, or cystoprostatectomy (Figure 18-1). Localization and accurate restaging is necessary before
BCG has been the most widely studied agent for topical considering cystectomy.
therapy. Initial concerns about obtaining effective con- Cystectomy should also be strongly considered when
tact with the prostatic urethra arose from observations of pathologic understaging is suspected. Hydronephrosis
recurrence in the prostatic urethra after BCG therapy for associated with a bladder tumor usually indicates muscle
TCC of the bladder. However, bladder neck incompe- invasion. Staging error rates as high as 55% have been
tence following TUR would seem to allow adequate tis- reported for T2 tumors.172,173 This reinforces the impor-
sue contact for treatment. For disease confined to the tance of careful pathologic review of all biopsy material.

High-grade bladder TCC (Ta, T1, CIS)

TUR biopsy of the prostatic urethra

CIS only Ductal stromal invasion Negative

Intravesical therapy Cystoprostatectomy


Cystoprostatectomy Treat bladder tumor as
(if fails, cystoprostatectomy with urethrectomy
with urethrectomy indicated
with urithrectomy) or intravesical therapy

Figure 18-1 Algorithm for evaluation and treatment of prostatic urethral TCC. (From Matzkin H,
Soloway MS, Haredeman S: J Urol 1991; 146:1210, with permission.)
Chapter 18 Superficial Transitional Cell Carcinoma of the Bladder 329

Table 18-4 Indications for Cystectomy trode at least 1 cm beyond the tip of the endoscope to
for Superficial Bladder Cancer avoid thermal damage to the fiberoptic system. Both
Persistent or recurrent high-grade papillary tumor techniques require a cooperative patient. There is con-
siderable cost benefit for this strategy in appropriate
Persistent or recurrent CIS individuals. It must be emphasized that representative
histopathology is necessary to avoid undertreatment of
Progression to muscularis propria or prostatic stromal innocent-appearing high-grade lesions.181
invasion
Protocols for cystoscopic follow-up have been pro-
TCC involving prostatic ducts posed based on known prognostic factors, such as tumor
size, number, histologic grade and stage, urine cytology,
and DNA ploidy. Parmar et al.18 concluded that the like-
Consideration should be given to concurrent ure- lihood of recurrence, and hence the need for follow-up
threctomy with cystectomy if there is widespread multi- cystoscopy, could be predicted simply and accurately by
focal CIS, involvement of the prostatic urethra in men, the number of tumors at initial presentation and the
or bladder neck and trigone in women. This probably presence of recurrent tumor at the first 3-month cys-
precludes orthotopic urinary reconstruction to the ure- toscopy risk categories and appropriate follow-up are
thra for these individuals, although some suggest that this summarized in Table 18-5. The power of this classifica-
is not an absolute contraindication with limited prostatic tion lies in the objectivity and reliability of these two
urethral involvement. prognostic factors. The safety and efficacy of this proto-
Partial cystectomy has a limited role. There is a sub- col have been demonstrated in a retrospective review of
group of superficial tumors in which this may be consid- 232 patients.182
ered, such as tumors within bladder diverticula. The duration of endoscopic follow-up for patients free
Diverticulectomy should be performed with care to of recurrence for many years has been debated. Morris
obtain adequate surgical margins and avoid tumor cell et al.,183 in a longitudinal follow-up study of 179 patients
spillage. with Ta and T1 TCC, documented recurrence risk over
Surgery alone achieves a high cure rate for superficial time. They reported that after a tumor-free interval of 2,
TCC. Amling et al.174 reported cancer-specific 5-year 5, and 10 years, the risk of recurrence is 43%, 22%, and
survival rates of 88% for Ta, 76% for T1, and 100% for 2%, respectively. No patient had a recurrence after
CIS. Pelvic lymph node metastases were present in 5.9%. remaining tumor free for 12 years. No patient free of
Similar survival figures have been reported in other large recurrence for 2 years progressed to muscle invasion or
series.175–177 metastases. Holmang et al.24 reported that only 1 of 59
patients tumor free for 5 years progressed. They also
observed that patients who had recurrences on 10 or
FOLLOW-UP
more cystoscopic studies, or multiple recurrences over
Since 30% to 80% of tumors will recur, close follow-up more than 4 years, continued to have recurrences until
is important to diagnose and treat recurrence early. death or cystectomy.
Flexible endoscopy and urinary cytology (for high-grade Urinary markers and cytology may have a role in long-
disease) are essential for follow-up. Historically this has term follow-up. Table 18-6 lists sensitivities and speci-
been performed at 3-month intervals, and then tailored ficities for potentially useful urinary markers. The ideal
according to patterns of recurrence. The disease status at
the initial 3-month cystoscopy is important in predicting
Table 18-5 Follow-Up for Superficial Bladder Cancer
future tumor behavior, and has become the basis of fol-
low-up protocols.18–48 Risk Category Recommended Follow-Up
Treatment of papillary recurrence, or suspicious areas,
Single tumor with no Annual cystoscopy
is ideally performed under anesthesia, with rigid instru- recurrence at 3 months
mentation, allowing adequate biopsies and thorough
visual assessment. The mucosa distant from the tumor Single tumor with 3-monthly cystoscopy
and the prostatic urethra in males should be sampled as recurrence at 3 months,
previously outlined for high-grade primary tumors. or multiple tumors
Laser ablation via flexible endoscopy under topical with no recurrence
anesthesia has been used to manage recurrences in
Multiple tumors with Adjuvant treatment
selected cases.178,179 Laser is well tolerated, although recurrence at 3 months
there is a recognized risk of bowel injury.178 Diathermy
has also been used180; 4-French electrodes are available From Parmar MKB, Friedman LS, Hargreave TB, et al: J Urol 1989;
to treat small lesions. Care must be taken to pass the elec- 142:284–288, with permission.
330 Part IV Bladder

Table 18-6 Urinary Markers for Follow-up of Superficial Bladder Cancer


Number Sensitivity Specificity
Marker Technique of Patients (%) (%)

LewisXantigen64 Immunocytology (bladder wash) 101 81 86

LewisXantigen184 Immunocytology 89 85 85

Basement membrane complex Latex agglutination 499 40 82


(Bard BTA)185

Autocrine motility factor (AMF) Qualitative immunoassay 70 80 75


receptor186

Nuclear matrix protein (NMP-22)* Quantitative immunoassay 69 74 78

Hyaluronidase† Quantitative ELISA assay 104 63 (high grade-99) 94

ELISA, enzyme-linked immunosorbent assay.


*Data derived from Soloway MS, Briggman J, Carpinito G, et al: Use of a new tumor marker, urinary NMP22, in the detection of occult or rapidly
recurring transitional cell carcinoma of the urinary tract following surgical treatment. Personal communication, 1995.
†Data derived from Lokeshwar BL: Personal communication, November, 1995.

Case 2
noninvasive bladder tumor marker would need to be
nearly 100% sensitive before replacing follow-up cys- A 48-year-old woman presented with a 2-cm papillary
toscopy. Voided urine cytology has excellent specificity tumor on the bladder trigone, near the ureteric orifice,
and reasonable sensitivity in high-grade lesions. But in with negative mucosal biopsies. Pathology confirmed a
low-grade lesions, the sensitivity is low (25%). At present grade 3 Ta lesion that was strongly positive for p53 on
none of these tests can completely replace cystoscopy for immunohistochemical staining. Small papillary recur-
surveillance. A positive cytology and a negative cys- rences were resected at 13 and 21 months, all of which
toscopy should prompt the clinician to investigate the remained grade 3 Ta and p53-positive. Intravesical mito-
patient further including random biopsies of the bladder, mycin (20 mg weekly for 5 weeks) was administered. The
prostatic urethra and careful assessment of upper tracts. patient remains tumor free at 10 months following the
The frequency of radiologic follow-up depends on the last TUR.
individual’s risk of developing upper urinary tract tumors.
This risk varies from 0.5% with low-grade bladder
Case 2 Discussion
tumors24,187 to 15% with CIS.188 Intravenous pyelogra-
phy should be performed every 2 to 5 years, accordingly. Recent data suggest that p53-positive papillary tumors
may be at increased risk for progression. These tumors
initially recurred as high-grade noninvasive lesions,
CASE HISTORIES
although they appear to have responded to a prophylac-
Case 1
tic course of mitomycin. BCG would have been an
A 74-year old female presented with a 4-cm papillary equally reasonable alternative and will be used with
bladder tumor. TUR confirmed a grade 3 T1 tumor with recurrence. Intravesical therapy could have been used at
invasion to the level of the muscularis mucosae but not the time of diagnosis given the high tumor grade.
involving deep muscle (stage T1a).
Case 3
Case 1 Discussion
A 66-year-old male underwent a TUR of the prostate for
Accurate staging is important when evidence of lamina symptoms associated with benign prostatic hyperplasia.
propria invasion is present on initial biopsy. Restaging Surprisingly, the pathology noted grade 3 TCC involving
biopsy is essential. Confirmed lamina propria invasion the prostatic ducts. No bladder disease was detected on
and high grade puts this patient at high risk for both biopsy at a subsequent cystoscopy. The patient declined
recurrence and progression. Intravesical BCG was further treatment. A TUR biopsy at 24 months again
administered weekly for 6 weeks, without evidence of demonstrated CIS involving prostatic ducts with no stro-
recurrence. mal invasion and no TCC of the bladder.
Chapter 18 Superficial Transitional Cell Carcinoma of the Bladder 331

Case 3 Discussion
distal ureter stricture treated with distal ureterectomy
The management options for prostatic ductal involve- and reimplantation. In follow-up she had a positive cytol-
ment include intravesical BCG or cystoprostatectomy. A ogy but no evidence of tumor on bladder biopsy She
cystoprostatectomy with urethrectomy was performed received 6 weeks of BCG. Her irritative symptoms wors-
confirming prostatic ductal CIS with extensive CIS of the ened. She later had dysplasia in the bladder documented
bladder and one focus of lamina propria invasion. This by biopsy and received another 6-week course of MMC.
demonstrates the problems of disease understaging, She remained tumor free but presented with incapacitat-
which is the major concern with intravesical treatment ing irritative voiding symptoms and incontinence and a
for both bladder and prostatic urethral TCC. severely contracted bladder. She underwent a cystectomy
and ileal conduit for severe bladder contracture. There
was no cancer in the specimen.
Case 4
A 65-year old male presented with a 1-cm grade 3 Ta
Case 6 Discussion
tumor involving the left wall of the bladder, which was
completely resected. A large, wide-mouthed bladder This is a classic case of overtreatment with intravesical
diverticulum located on the right wall of the bladder was therapy and incapacitating iatrogenic morbidity. While
filled with a second grade 3 papillary tumor. Random intravesical agents may prolong recurrence-free survival,
bladder and prostatic urethral biopsies were negative. A the side effects of treatment should not be underestimated.
TUR biopsy at 8 weeks revealed no evidence of tumor in
the left wall. A partial cystectomy removing the bladder
Case 7
diverticulum was performed, confirming a grade 3 Ta
tumor with associated CIS but negative surgical margins. A 75-year old woman presented with a history of super-
ficial, low-grade TCC in 1983. She had been treated pre-
viously for interstitial cystitis. In 1988, she underwent a
Case 4 Discussion
left nephroureterectomy for T1, grade 2 TCC of the
Residual or high-grade noninvasive tumor involving a ureter. In 1991, she was found to have CIS in the bladder
bladder diverticulum could be managed with divertic- and superficial tumor in the right distal ureter. A stent
ulectomy or cystoprostatectomy. Care must be taken to was placed and she received intravesical MMC. Four
exclude and treat other foci of tumor, if bladder preser- months later she had a resection of a T1, grade 3 bladder
vation is chosen. tumor. She was given a second 6-week course of BCG.
Nine months later she had a TURBT, with pathology
revealing a high-grade, muscle-invasive tumor with
Case 5
involvement of the right ureteral orifice and invasion of
A 74-year old female presented with a history of CIS and the bladder neck and urethra. Cystectomy with urinary
T1 grade 3 TCC of the bladder. After resection she was diversion was performed. Pathology revealed a grade 3
treated with 6 weeks of BCG. T3 TCC. No adjuvant chemotherapy was given.

Case 5 Discussion Case 7 Discussion


This case prompts consideration as to whether to give This case illustrates the progression of a superficial lesion
maintenance BCG. Early randomized studies showed no to a high-grade invasive tumor despite intravesical
clear-cut advantage to giving maintenance BCG over a MMC, BCG, and close surveillance.
standard 6-week inductive course. Lamm et al.138 recently
reported that not only can maintenance therapy reduce
tumor recurrence but may also increase survival. The
optimal timing of maintenance doses has not been clearly REFERENCES
established. Given the patient’s age and risk of side effects,
no maintenance therapy was given in this case. 1. Soloway MS: Flexible cystourethroscopy: alternative to
rigid instruments for evaluation of the lower urinary
tract. Urology 1985; 25:472.
Case 6 2. Matzkin H, Moinuddin S, Soloway MS: Value of urine
cytology versus bladder washing in bladder cancer.
A 77-year old woman was diagnosed with multifocal CIS Urology 1992; 39(3):201.
in 1990. She was treated with 6 weekly doses of intraves- 3. Soloway MS, Patel J: Surgical techniques for endoscopic
ical MMC. At that time she had frequency and urgency, resection of bladder cancer. Urol Clin North Am 1992;
as well as perineal discomfort. In 1993, she had a right 19(3):467.
332 Part IV Bladder

4. Soloway MS: An optical dilator to obviate blind urethral 22. Birch BRP, Harland SJ: The pT1 G3 bladder tumor.
dilation prior to endoscopic resections. Urol 1988; 31:427. Urology 1989; 64:109.
5. Heney NM, Ahmed S, Flanagan MJ, et al: Superficial 23. Anderstrom C, Johansson S, Nilsson S: The significance
bladder cancer: progression and recurrence. J Urol 1983; of lamina propria invasion on the prognosis of patient
130:1083. with bladder tumors. J Urol 1980; 124:23.
6. Jordan AM, Weingarten J, Murphy WM: Transitional 24. Holmdng S, Hedelin H, Anderstrom C, et al: The
cell neoplasms of the urinary bladder: can biological relationship among multiple recurrences, progression
potential be predicted from histological grading? Cancer prognosis of patients with stages Ta and T1 transitional
1987, 60:2766. cell carcinoma of the bladder followed for at least 20
7. Mostofi FK, Sobin LH, Torlini H: Histological typing of years. J Urol 1995; 153:18.
urinary bladder tumors. International Histological 25. Hassui Y, Osada Y, Kitada S, et al: Significance of
Classification of Tumors No. 10. Geneva, World Health invasion of the muscularis mucosa on the progression of
Organization, 1973. superficial bladder cancer. Urology 1994; 43:782.
8. Murphy WM, Beckwith JB, Farrow G: Tumors of 26. Dixon JS, Gosling JA: Histology and fine structure
the kidney, bladder and related urinary structures, muscularis mucosa of the human urinary bladder. J Anat
pp 198–224. Washington, DC, Armed Forces Institute 1983; 136:265.
of Pathology, 1994. 27. Younes M, Sussman J, True LD: The usefulness level of
9. Ayala AG, Ro JY: Premalignant lesions of the urothelium muscularis mucosa in the staging of in, transitional cell
and transitional cell tumors. In Young RH (ed): carcinoma of the urinary bladder. Cancer 1990; 66:543.
Pathology of the Urinary Bladder, pp 65–101. New York, 28. KIan R, Loy V, Huland H: Residual tumor discovered
Churchill Livingstone, 1989. routine second TUR in patients with T1 transition
10. Carbin BE, Elkman P, Gustafson H, et al: Grading of carcinoma of the bladder. J Urol 1991; 146:316.
human urothelial neoplasms based on nuclear atypia and 29. Lynch CF, Platz CE, Jones MP, et al: Cancer registry
mitotic frequency 1: histological description. J Urol problems in classifying invasive bladder cancer. J Natl
1991; 145:968. Inst 1991; 83:429.
11. Carbin BE, Elkman P, Gustafson H, et al: Grading of 30. Abel PB, Henderson D, Bennett MK, et al: Differing
human urothelial neoplasms based on nuclear atypia a interpretations by pathologists of the pT category grade
mitotic frequency. II: prognostic importance. J Urol of transitional cell carcinoma of the bladder. Urol 1988;
1991; 145:972. 62:339.
12. Kaubisch S, Lum BL, Reese J, et al: Stage TI bladder 31. Olsen LH, Overgaard S, Frederiksen P, et al: The
cancer: grade is the primary determinant for risk of reliability of staging and grading of bladder tumors: in
muscle invasion. J Urol 1991; 146:28. misinformation of the pathologists’ diagnosis. Scand
13. Pauwels RP, Schapers REM, Smeets AWGB, et al: Grad J Urol Nephrol 1993; 27:349.
in superficial bladder cancer 1: morphological criteria. 32. Witjes JA, Kiemeney LALM, Schaafsma HE, et al:
J Urol 1988; 61:129. Influence of review pathology on study outcome
14. Blomjous CEM, Vos W, Schipper NW: The prognostic randomized multicenter superficial bladder cancer trial.
significance of selective nuclear morphometry in urine Br J Urol 1994; 7(3):172.
bladder carcinoma. Hum Pathol 1990; 21:409. 33. Richards B, Parmar MKB, Anderson CK, et al:
15. Ooms ECM, Anderson WAD, Alons CL, et al: Anal of Interpretation of biopsies of “normal” urothelium in
the performance of pathologists in the grading of patients with superficial bladder cancer. Br J Urol 1991;
bladder tumors. Hum Pathol 1983; 14:140. 67:369.
16. Jewett HJ, Strong GH: Infiltrating carcinoma of the 34. Sidransky D, Frost P, Von Eschenbach A, et al:
bladder: relation of depth of penetration of bladder wall Clonal 5 origin of bladder cancer. N Engl J Med 1992;
incidence of local extension and metastases. J Urol 1978 326:737.
55:366. 35. Harris AL, Neal D: Bladder cancer-field versus clonal
17. Beahrs OH, Henson DE: The Manual for Staging of origin. N Engl J Med 1992; 326:759.
Cancer. AJCC. Philadelphia, Lippincott Raven, 1993. 36. Soloway MS, Murphy W, Rao MK, et al: Serial multiple
18. Parmar MKB, Friedman LS, Hargreave TB, et al: site biopsies in patients with bladder cancer. J Urol 1978;
Prognostic factors for recurrence follow-up policies in 120:57.
treatment of superficial bladder cancer: report from 37. Kakizoe T, Maturnoto K, Nishio Y, et al: Significance of
British Medical Research Council Subgroup on carcinoma in situ and dysplasia in association with
superficial bladder cancer. J Urol 1989; 142:284. bladder cancer. J Urol 1985; 133:395.
19. Abel PD, Hall RR, Williams G: Should pT1 transitional 38. Kiemeney LALM, Witjes JA, Heijbroek RP, et al:
cell carcinoma of the bladder still be classified as Should random urothelial biopsies be taken from
superficial? Br J Urol 1988; 62:235. patients with primary superficial bladder cancer? A
20. Heney NM, Nocks BN, Daley JJ, et al: Ta and T1 decision analysis. Br J Urol 1994; 73:164.
bladder cancer: location, recurrence, and progression. 39. Hardeman SW, Perry A, Soloway MS: Transitional cell
Br J 1982; 54:152. carcinoma of the prostate following intravesical therapy
21. Williams JL, Hammonds JC, Saunders N: T1 bladder for transitional cell carcinoma of the bladder. J Urol
tumors. Br J Urol 1977; 49:663, 1988; 140:289.
Chapter 18 Superficial Transitional Cell Carcinoma of the Bladder 333

40. Montie JE, Mersky H, Levin HS: Transitional cell 58. Richie JP, Blute RD, Waisman J: Immunological
carcinoma of the prostate in a series of cystectomies: indicators of prognosis in bladder cancer: the importance
incidence and staging problems. J Urol 1986; 135:243A. of cell surface antigens. J Urol 1980; 123:22.
41. Wolf H, Hojgaard K: Urothelial dysplasia concomitant 59. Blasco E, Tarrado J, Belloso L, et al: T antigen: a
with bladder tumors as a determining factor for future prognostic indicator of high recurrence index in transitional
new occurrences. Lancet 1983; 2(8342):134. cell carcinoma of the bladder. Cancer 1988; 61:1091.
42. Althausen AF, Prout GR, Daly JJ: Noninvasive papillary 60. Bergman S, Javadpour N: The cell surface antigen as an
carcinoma of the bladder associated with carcinoma-in- indicator of malignant potential in stage A bladder
situ. J Urol 1976; 116:575. cancer. J Urol 1978; 119:49.
43. Smith G, Elton RA, Beynon LL, et al: Prognostic 61. Sanders H, McCue P, Graham SD: ABO (H) antigens
significance of biopsy results of normal looking mucosa and beta-2 microglobulin in transitional cell carcinoma:
in cases of superficial bladder cancer. Br J Urol 1983; predictors of response to intravesical BCG. Cancer 1991;
55:665. 67:3024.
44. Olsen PR, Wolf H, Schoeder T, et al: Urothelial atypia 62. Johl BR, Hartzen SH, Hainau B: Lewis A antigen in
and survival rate of 500 unselected patients with primary transitional cell tumors of the urinary bladder. Cancer
transitional cell carcinoma of the urinary bladder. Scand 1986; 58:222.
J Urol Nephrol 1988; 22:257. 63. Langkilde NC, Wolf H, Milgard P, et al: Frequency and
45. Lerman RL, Hulter RVP, Whitmore WF: Papilloma of mechanism of Lewis antigen expression in human
the urinary bladder. Cancer 1970; 25:333. urinary bladder and colon carcinoma patients.
46. Cutler SJ, Heney NM, Friedell GH: Longitudinal study Br J Cancer 1991; 63:583.
of patients with bladder cancer: factors associated with 64. Golijanin D, Scherman Y, Shapiro A: Detection of
disease recurrence and progression. In Bonney WW, bladder tumors by immunostaining of Lewis X antigen
Prout JR (eds): Bladder Cancer, p 35. Baltimore, in cells from voided urine. Urology 1995; 46:173.
Williams & Wilkins, 1982. 65. Conn IG, Vilela MJ, Garrod DR, et al:
47. Lutzeyer W, Rubben H, Damm H: Prognostic Immunohistochemical staining with monoclonal
parameters in superficial bladder cancer: an analysis of antibody 32-213 to desmosomal glycoprotein 1: its role
315 cases. J Urol 1982; 127:250. in the histological assessment of urothelial carcinomas.
48. Fitzpatrick JM, West AB, Butler MR, et al: Superficial Br J Urol 1990; 65:176.
bladder tumors (stage pTa grades, 1&2): the importance 66. Smith NW, Stutton GM, Walsh MD, et al: Transferrin
of recurrence pattern following initial resection. J Urol receptor expression in primary superficial bladder tumors
1986; 135:920. identifies patients who develop recurrences. Br J Urol
49. Pagano F, Garbeglio A, Milani C, et al: Prognosis of 1990; 65:339.
bladder cancer 1: risk factors in superficial transitional 67. Conn IG, Crocker J, Emtage LA, et al: Human milk fat
cell carcinoma. Eur Urol 1987; 13:145. globulin-2 as a prognostic indicator in superficial bladder
50. Murphy WM, Soloway MS, Jukkola AF, et al: Urine cancer. J Clin Pathol 1988; 41:1191.
cytology and bladder cancer. Cancer 1984; 53:1555. 68. Neal DE, Scharples L, Smith K, et al: The epidermal
51. Murphy WM: Current status of urine cytology in the growth factor receptor and the prognosis of bladder
evaluation of bladder neoplasms. Hum Pathol 1990; cancer. Cancer 1990; 65:1619.
21:886. 69. Mellow K, Wright C, Kelly P, et al: Long term outcome
52. De Vita R, Forte D, Maggi F, et al: Cellular DNA related to epidermal growth factor receptor status in
content and proliferative activity evaluated by flow bladder cancer. J Urol 1995; 153:919.
cytometry versus histopathological staging classifications 70. Schapers RFM, Pauwels RPE, Havenith MG, et al:
in human bladder tumors. Eur Urol 1991; 19:65. Prognostic significance of type IV collagen and laminin
53. Gustafson H, Tribukait B, Esposti PL: DNA pattern, immunoreactivity in urothelial carcinomas of the
histological grade, and multiplicity related to recurrence bladder. Cancer 1990; 66:2583.
rate in superficial bladder tumors. Scand J Urol Nephrol 71. Sandberg AA: Chromosome markers and progression in
1982; 16:135. bladder cancer. Cancer Res 1977; 37:2950.
54. Gustafson H, Tribukait B, Esposti PL: DNA profile in 72. Waldman FM, Carroll PR, Kerschmann R, et al:
tumor progression in patients with superficial bladder Centromeric copy number of chromosome 7 is strongly
tumors. Urol Res 1982; 10:13. correlated with tumor grade and labeling index in human
55. Masters JRW, Camplejohn RS, Parkinson MC, et al: bladder cancer. Cancer Res 1981; 51:3807.
DNA ploidy and the prognosis of stage pT1 bladder 73. Fearon ER, Feinberg AP, Hamilton SH, et al: Loss of
cancer. Br J Urol 1989; 64:403. genes on the short arm of chromosome 11 in bladder
56. Murphy WM, Chandler RW, Trafford RM: Flow cancer. Nature 1985; 318:377.
cytometry of deparaffinised nuclei compared to 74. Hopman A, Moesker O, Smeets A, et al: Numerical
histological grading for the pathological evaluation of chromosome 1, 7, 9, 11 aberrations in bladder cancer
transitional cell carcinoma. J Urol 1986; 135:694. detected by in situ hybridization. Cancer Res 1991; 51:644.
57. Newman AJ, Carlton C, Johnson S: Cell surface A,B, O 75. Summers JL, Falor W, Ward R: A 10 year analysis of
blood group antigen as an indicator of malignant potential chromosomes in noninvasive papillary carcinoma of the
in stage A bladder carcinoma. J Urol 1980; 124:27. bladder. J Urol 1981; 125:177.
334 Part IV Bladder

76. Tsai YC, Nichols PW, Hiti AL, et al: Allelic losses of 95. Lipponen PK: Overexpression of p53 nuclear
chromosomes 9, 11, and 17 in human bladder cancer. oncoprotein in transitional cell bladder cancer and its
Cancer Res 1990; 50:44. prognostic value. Int J Cancer 1993; 53:365.
77. Olumi AF, Tsai YC, Nichols PW, et al: Allelic loss of 96. Gardiner RA, Walsh MD, Allen S, et al:
chromosome 17p distinguishes high grade from low Immunohistological expression of p53 in primary pT1
grade transitional cell carcinoma of the bladder. Cancer transitional cell bladder cancer in relation to tumor
Res 1990; 50:7081. progression. Br J Urol 1994; 73:526.
78. Agnantis NJ, Constantinidou A, Poulios C, et al: 97. Sarkis A, Dalbagni G, Cordon-Cardo C, et al: Nuclear
Immunohistochemical study of the ras oncogene overexpression of p53 protein in transitional cell bladder
expression in human bladder endoscopy specimens. Eur carcinoma: a marker of disease progression. J Natl
J Surg Oncol 1990; 16:153. Cancer Inst 1993; 85:53.
79. Viola MV, Fromowitz F, Oravez S, et al: Ras oncogene 98. Esrig D, Spruck C, Nichols P, et al: p53 nuclear protein
p2l. expression is increased in premalignant lesions and accumulation correlates with mutations in the p53 gene,
high grade bladder cancer. J Exp Med 1985; 161:1211. tumor grade, and stage, in bladder cancer. Am J Pathol
80. Feinberg AP, Vogelstein B, Droller MJ, et al: Mutation 1993; 143:1389.
affecting the 12th amino acid of the c-Ha-ras oncogene 99. Dalbagni G, Cordon-Cardo C, Reuter V, et al: Tumor
product occurs infrequently in human cancer. Science suppressor gene alterations in bladder carcinoma
1983; 220:1175. translational correlates to clinical practice. Surg Oncol
81. Lane DP: p53 guardian of the genome. Nature 1991; Clin North Am 1995; 4:231.
358:15. 100. Esrig D, Elmajian D, Groshen S, et al: Accumulation of
82. Kasten MB, Onyekwere O, Sidransky D, et al: nuclear p53 and tumor progression in bladder cancer.
Participation of p53 protein in the cellular response to N Engl J Med 1994; 331:1259.
DNA damage. Cancer Res 1991; 51:6304. 101. Sato K, Moriyama M, Mori S, et al: An
83. Reich NC, Oren M, Levine AJ: Two distinct mechanisms immunohistological evaluation of c-er17 B-2 gene
regulate the levels of cellular tumor antigen p53. Mol product in patients with urinary bladder carcinoma.
Cell Biol 1993; 3:2143. Cancer 1983; 70:2493.
84. Harris AL: Telling change of base. Nature 1990; 102. Moriyarria M, Akiyama T, Yamamoto T, et al:
350:377. Expression of c-erli B-2 gene product in urinary bladder
85. Fujimoto K, Yamada Y, Okajima E, et al: Frequent cancer. I Urol 1991; 145:423.
association of p53 gene mutation in invasive bladder 103. Masters JR, Vessey SG, Minn CF: c-nzyc oncoprotein
cancer. Cancer Res 1992; 52:1393. levels in bladder cancer. Urol Res 1988; 16:341.
86. Sidransky D, von Eschenbach A, Tsai YC: Identification 104. Lamm DL, Lamm LM: Benefits of intravesical
of p53 gene mutations in bladder cancers and urine chemotherapy for superficial disease. Contemp Urol
samples. Science 1991; 252:706. 1989:3.
87. Lunec J, Challen C, Wright C, et al: c-erb B-2 105. Walker MC, Masters JR, Parris CN, et al: Intravesical
amplification and identical p53 mutations in concomitant chemotherapy: in vitro studies on the relationship
transitional carcinoma of the renal pelvis and urinary between dose and cytotoxicity. Urol Res 1986; 14:137.
bladder. Lancet 1992; 339:439. 106. Goos E, Masters JR: Intravesical chemotherapy: studies
88. Iggo R, Gatter K, Bartek J, et al: Increased expression of on the relationship between osmolality and cytotoxicity.
mutant forms of p53 oncogene in primary lung cancer. J Urol 1986; 136:399.
Lancet 1990; 335:675. 107. Cant JD, Murphy WM, Soloway MS: Prognostic
89. Sarkis A, Dalbagni G, Cordon-Cardo C, et al: significance of urine cytology on initial follow up after
Association of p53 nuclear overexpression and tumor intravesical mitomycin C for superficial bladder cancer.
progression in carcinoma-in-situ of the bladder. J Urol Cancer 1986; 57:2119.
1994; 152:388. 108. Haaf EO, Dresner SM, Ratliff TL, et al: Two courses of
90. Sidransky D, Mikkelsen T, Schwechheimer K, et al: intravesical bacillus Calmette-Guérin for transitional cell
Clonal expansion of p53 mutant cells is associated with carcinoma of the bladder. J Urol 1986; 136:820.
brain tumor progression. Nature 1992; 355:846. 109. Eure GR, Cundiff MR, Shellharnmer PF: Bacillus
91. Oka K, Ishikana J, Bruner JM, et al: Detection of loss of Calmette-Guérin therapy for high risk stage TI
heterozygosity in p53 gene in renal cell carcinoma and superficial bladder cancer. J Urol 1992, 152:376.
bladder carcinoma using the polymerase chain reaction. 110. Soloway MS, Perito PE: Superficial bladder cancer:
Mol Carcinog 1991; 4:10. diagnosis, surveillance and treatment. J Cell Biochem
92. Spruck CH, Ohneseit PF, Gonzales-Sulueta M, et al: 1992; 161(Suppl):120.
Two molecular pathways to transitional cell carcinoma of 111. Soloway MS: Surgery and intravesical chemotherapy in
the bladder. Cancer Res 1994; 54:784. the management of superficial bladder cancer. Semin
93. Yoskimura I, Kudoh J, Saito S: p53 gene mutation in Urol 1983; 11:23.
recurrent superficial bladder cancer. J Urol 1995; 112. Prout GR Jr, Koontz WW Jr, Coombs LJ, et al: Long-
153:1711. term fate of 90 patients with superficial bladder cancer
94. Uchida T, Wada C, Ishida H, et al: p53 mutations and randomly assigned to receive or not to receive thiotepa.
prognosis in bladder tumors. J Urol 1995; 153:1097. J Urol 1983; 130:677.
Chapter 18 Superficial Transitional Cell Carcinoma of the Bladder 335

113. MRC Working Party on Urological Cancer, Subgroup 129. Lamm DL, Blumenstein BA, Crawford ED, et al: A
on Superficial Bladder Cancer: The effect of intravesical randomized trial of intravesical doxorubicin and
thiotepa on tumor recurrence after endoscopic treatment immune-therapy with bacille Calmette-Guérin for
of newly diagnosed superficial bladder cancer. A further transitional cell carcinoma of the bladder. N Engl J Med
report with long-term follow-up of a Medical Research 1991; 325:1205.
Council randomized trial. Br J Urol 1994; 73:632. 130. Jackse G, Hofstadter F, Marsberger H: Topical
114. Soloway MS: Rationale for intensive intravesical doxorubicin hydrochloride therapy for carcinoma in situ
chemotherapy for superficial bladder cancer. J Urol of the bladder: a follow-up. J Urol 1984; 131:41.
1980; 123:461. 131. Ausfield A, Beer M, Muhlethale J, et al: Adjuvant
115. Soloway MS, Jordan AM, Murphy WM: Rationale for intravesical chemotherapy of superficial bladder cancer
intravesical chemotherapy in the treatment and with monthly doxorubicin of intensive mitomycin: a
prophylaxis of superficial transitional cell carcinoma. comparison of two consecutive series. Eur Urol 1987;
Prog Clin Biol Res 1989; 310:215. 13:10.
116. Soloway MS, Ford KS: Thiotepa-induced 132. Sweatman TW, Parker RF, Israel M: Pharmacologic
myelosuppression: review of 670 bladder instillations. rationale for (AD-32): a preclinical study. Cancer
J Urol 1983; 130:889. Chemother Pharmacol 1991; 28:1.
117. Koontz WW Jr, Heney NM, Barton B, et al: Intravesical 133. Greenberg R, O’Dwyer F, Patterson L, et al: Intravesical
effects of mitomycin C in patients with superficial AD-32 (N-trifluoroacetyladriamycin-14-valerate) in the
bladder carcinoma who have previously failed therapy treatment of patients with refractory bladder
with thiotepa. Urology 1985; 26:30. carcinoma—clinical efficacy, pharmacology, and safety.
118. Heney NM: First-line chemotherapy of superficial J Urol 1995; 153:233A.
bladder cancer: mitomycin C versus thiotepa. Urology 134. Morales AM, Eidinger D, Bruce AW: Intracavitary
1985; 26:27. bacillus Calmette-Guérin in the treatment of superficial
119. Soloway MS: Diagnosis and management of superficial bladder tumors. J Urol 1976; 116:180.
bladder cancer. Semin Surg Oncol 1989; 5:247. 135. Guerin C: The history of BCG. In Rosenthal SR (ed):
120. Huland H, Otto J, Droese M, et al: Long-term BCG Vaccination Against Tuberculosis, p 35. Boston,
mitomycin C instillation after transurethral resection of Little Brown & Co, 1957.
superficial bladder carcinoma: influence on recurrence, 136. Ratliff TL, Haaff EO, Catalona WJ: Interleukin 2
progression, and survival. J Urol 1984; 132:27. production during intravesical bacillus Calmette-Guérin
121. Maier U, Hobarth K: Long-term observation after therapy for bladder cancer. Clin Immunol
intravesical metaphylaxis with mitomycin C in patients Immunopathol 1986; 40:375.
with superficial bladder tumors. Urology 1991; 37(5):481. 137. Brosman S: Bacillus Calmette-Guérin immunotherapy:
122. Tolley DA, Parmar MKB, Grigor KM, et al: The effect techniques and results. Urol Clin North Am 1992;
of intravesical mitomycin C on recurrence of newly 19(3):557.
diagnosed superficial bladder cancer: a further report 138. Lamm DL, Crawford ED, Blumenstein B, et al:
with 7 years’ follow-up. J Urol 1996; 155:1223. Maintenance BCG immunotherapy of superficial bladder
123. Nissenkorn I, Herrod H, Soloway MS: Side effects cancer: a randomized prospective Southwest Oncology
associated with intravesical mitomycin. J Urol 1981; Group Study (Abstract 242). J Urol 1992; 147(part 2):
126:596. 274A.
124. Batts CN: Adjuvant intravesical therapy for superficial 139. Lamm DL: BCG in carcinoma-in-situ and superficial
bladder cancer. Ann Pharmacother 1992; 26:1270. bladder tumors. EORTC Genitourinary Group.
125. Niijima T, Koiso K, Adaza H: Randomized clinical trial Monograph 1988; 5:497.
for chemoprophylaxis of recurrence in superficial bladder 140. Herr HW, Laudone VP, Whitmore WF: An overview of
cancer. In Spitzy KH, Karrer K (eds): Proceedings of the intravesical therapy for superficial bladder tumors. J Urol
13th International Congress of Chemotherapy, 1987; 138:1363.
pp 240–246, Vienna, 1983. 141. Dejager R, Guinan F, Lamm D, et al: Long-term complete
126. Kurth KH, Debruyne FJM, Senge T, et al: Adjuvant remission in bladder carcinoma in situ with intravesical
chemotherapy of superficial transitional cell carcinoma: Tice bacillus Calmette-Guérin: overview analysis of six
an EORTC randomized trial comparing doxorubicin phase 11 clinical trials. Urology 1991; 38:507.
hydrochloride, ethoglucid and TUR-alone. Prog Clin 142. Herr HW: Intravesical BCG: current results, natural
Biol Res 1985; 162A:135. history and implications for urothelial cancer prevention.
127. Zincke H, Utz DC, Taylor WF, et al: Influence of J Cell Biochem 1992; 161(Suppl):112.
thiotepa and doxorubicin instillation at time of 143. Herr HW, Schwalb DM, Zhang ZF, et al: Intravesical
transurethral surgical treatment of bladder cancer on bacillus Calmette-Guérin therapy prevents tumor
tumor recurrence: a prospective, randomized, double- progression and death from superficial bladder cancer:
blind, controlled trial. J Urol 1983; 129:505. ten-year follow-up of a prospective randomized trial.
128. Llopis B, Gallego J, Mompo JA, et al: Thiotepa versus J Clin Oncol 1995; 13:1404.
Adriamycin versus cis-platinum in the intravesical 144. Brosman SA: Experience with bacillus Calmette-Guérin
prophylaxis of superficial bladder tumors. Eur Urol in patients with superficial bladder carcinoma. J Urol
1985; 11:73. 1982; 128:27.
336 Part IV Bladder

145. Martinez-Pineiro JA: BCG vaccine in superficial bladder silent but extensive intraprostatic involvement. Cancer
tumors: eight years later. Eur Urol 1989; 142:719. 1975; 36:514.
146. Lamm DL, Crissinan J, Blumenstein BA, et al: 165. Hardeman S, Soloway M: Transitional cell carcinoma of
Adriamycin versus BCG in superficial bladder cancer: a the prostate: diagnosis, staging and management. World
Southwest Oncology Group study. In Debruyne FMJ, J Urol 1988; 6:170.
van der Meijden APM (eds): BCG in Superficial Bladder 166. Hardeman S, Perry A, Soloway MS: Transitional cell
Cancer, p 123. New York, Alan R Liss, 1989. carcinoma of the prostate following intravesical therapy
147. Vegt PDJ, Witjes JA, Wities WPJ, et al: A randomized for transitional cell carcinoma of the bladder. J Urol
study of intravesical mitomycin C, bacillus Calmette- 1988; 140:289.
Guérin Tice and bacillus Calmette-Guérin RIVM 167. Herr H, Pinsky C, Whitmore W, et al: Long term effect
treatment in pTa-pT1 papillary carcinoma and of intravesical bacillus Calmette-Guérin on flat
carcinoma in situ of the bladder. J Urol 1995; 153:929. carcinoma-in-situ of the bladder. J Urol 1986; 135:265.
148. Cookson MS, Sarosdy MF: Management of stage T1 168. Bretton P, Herr H, Whitmore W, et al: Intravesical bacillus
superficial bladder cancer with intravesical bacillus Calmette-Guérin therapy for in-situ transitional cell
Calmette-Guérin therapy. J Urol 1992; 148:797. carcinoma of the prostatic urethra. J Urol 1989; 141:853.
149. Nadler RB, Catalona WJ, Hudson MA, et al: Durability 169. Hillyard R, Ladaga L, Schellhammer P: Superficial
of the tumor-free response for intravesical bacillus transitional cell carcinoma of the bladder associated with
Calrnette-Guérin therapy. J Urol 1994; 152:367. mucosal involvement of the prostatic urethra: results of
150. Lamm DL: Complications of bacillus Calmette-Guérin treatment with intravesical bacillus Calmette-Guérin.
immunotherapy. Urol Clin North Am 1992; 19:565. J Urol 1988; 139:290.
151. Nelson B, Borden E: Interferons: biological and clinical 170. Hardeman S, Soloway MS: Urethral recurrence
effects. Semin Surg Oncol 1989; 5:391. following radical cystectomy. J Urol 1990; 144:666.
152. Sargent ER, Williams RD: Immunotherapeutic 171. Schellhammer P, Bean M, Whitmore W: Prostatic
alternatives in superficial bladder cancer: interferon, involvement by transitional cell carcinoma: pathogenesis,
interleukin 2 and keyhole limpet hemocyanin. Urol Clin patterns, and prognosis. J Urol 1977; 118:399.
North Am 1992; 19(3):581. 172. Hudson M: When intravesical measures fail: indications
153. Torti F, Shortliffe L, Williams R, et al: Alpha-interferon for cystectomy in superficial disease. Urol Clin North
in superficial bladder cancer: a Northern California Am 1992; 19:601.
Oncology Group study. J Clin Oncol 1988; 6:476. 173. Soloway MS, Lopez A, Patel J, et al: Results of radical
154. Glashan RW: A randomized controlled study of cystectomy for transitional cell carcinoma of the bladder
intravesical et-2b-interferon in carcinoma in situ of the and the effect of chemotherapy. Cancer 1994; 73:1926.
bladder. J Urol 1990; 144:658. 174. Amling C, Thraser J, Frazier H, et al: Radical
155. Olsson D, Rao C, Menzoian J, et al: Immunologic cystectomy for stages Ta, Tis, & T1 transitional cell
unreactivity in bladder cancer patients. J Urol 1972; carcinoma of the bladder. J Urol 1994; 151:31.
107:607. 175. Bracken RB, McDonald WW, Johnson DE: Cystectomy
156. Jurinic C, Engelmann U, Gasch J: Immunotherapy in for superficial bladder cancer. Urology 1981; 18:459.
bladder cancer with keyhole-limpet hemocyanin: a 176. Malkowicz SB, Nicols P, Lieskovsky G, et al: The role of
randomized study. J Urol 1988; 139:723. radical cystectomy in the management of high grade
157. Wierenga. W: Antiviral and other bioactivities of superficial bladder cancer (pA, p1, pis, & p2). J Urol
pyrimidinones. Pharmacol Ther 1985; 30:67. 1990; 144:641.
158. Sarosdy MF, Lamm DL, Williams RD, et al: Phase I trial 177. Pagano F, Bassi P. Galetti TP, et al: Results of
of oral bropirimine in superficial bladder cancer. J Urol contemporary radical cystectomy for invasive bladder
1992; 147:31. cancer: a clinicopathological study with an emphasis on
159. Sarosdy MF, Lowe BA, Schellhammer PF, et al: the inadequacy of tumor, nodes and metastases
Bropirimine immunotherapy of bladder CIS: positive classification. J Urol 1991; 145:45.
phase 11 results of an oral interferon inducer. J Urol 178. Smith JA: Laser surgery for transitional cell carcinoma.
1994; 13:304A. Technique, advantages and limitations. Urol Clin North
160. Herr HW, Wartinger DD, Fair WR, et al: Bacillus Am 1992; 19(3):473.
Calmette-Guérin therapy for superficial bladder cancer: 179. Tarantino A, Aretz T, Libertino J, et al: Is the
a ten year follow-up. J Urol 1992; 147:1020. neodymium YAG laser effective therapy for invasive
161. Zinke H, Utz DC, Farrow GM: Review of Mayo Clinic bladder cancer? Urology 1991; 38:514.
experience with carcinoma-in-situ. Urology 1985; 180. Herr H: Outpatient flexible cystoscopy and fulguration
26(Suppl):39. of recurrent superficial bladder tumors. J Urol 1990;
162. Matzkin H, Soloway MS, Hardeman S: Transitional cell 144:1365.
carcinoma of the prostate. J Urol 1991; 146:1207. 181. Bianchi G, Nevelli P, Beltrami P, et al: Small urothelial
163. Wood D, Montie J, Pontes J, et al: Transitional cell carcinoma: diagnosis and treatment by cold cup forcep
carcinoma of the prostate in cystoprostatectomy specimens biopsy. J Urol 1990; 144:872.
removed for bladder cancer. J Urol 1989; 141:346. 182. Reading J, Hall RR, Parmar MKB: The application of
164. Seemayer TA, Knaack J, Thelmo W, et al: Further prognostic factor analysis for Ta/T1 bladder cancer in
observations on carcinoma-in-situ of the urinary bladder: routine urological practice. Br J Urol 1995; 75:604.
Chapter 18 Superficial Transitional Cell Carcinoma of the Bladder 337

183. Morris SB, Gordon EM, Shearer RJ, et al: Superficial 188. Miller EB, Eure GR, Schellharnmer PF, et al: Upper
bladder cancer: how long should a tumor-free patient tract transitional cell carcinoma following treatment of
have check cystoscopies? Br J Urol 1995; 75:193. superficial bladder cancer with BCG. Urology 1993;
184. Sheinfeld J, Reuter VE, Melamed MR, et al: Enhanced 42(l):26.
bladder cancer detection with the Lewis X antigen as a 189. O’Donnell MA, Krohn J, DeWolf WC: Salvage
marker of neoplastic transformation. J Urol 1990; intravesical therapy with interferon-α2b plus low dose
143:285. bacillus Calmette-Guérin is effective in patients with
185. Sarosdy MF, De Vere White RW, Soloway MS, et al: superficial bladder cancer in whom bacillus Calmette-
Results of a multicenter trial using the BTA test to Guérin alone previously failed. J Urol 2001;
monitor for and diagnose recurrent bladder cancer. 166:1300–1305.
J Urol 1995; 154:379. 190. Soloway MS, Sofer M, Vaidya A:. Contemporary
186. Korman HJ, Peabody JO, Cerny JC, et al: Autocrine management of stage T1 transitional cell carcinoma of
motility factor receptor as a possible urine marker for the bladder. J Urol 2002; 167:1573–1583.
transitional cell carcinoma of the bladder. J Urol 1996;
155:347.
187. Booth CM, Kellett MJ: Intravenous urography and the
follow-up of carcinoma of the bladder. Br J Urol 1981;
53:246.
C H A P T E R

19Prognosis and Management of


Invasive Transitional Cell Carcinoma
J. Matthew Hassan, MD, Sam S. Chang, MD,
Michael S. Cookson, MD, and Joseph A. Smith, Jr, MD

Fifty-four thousand new cases of bladder cancer are diag- muscle-invasive TCC, as well as current treatment rec-
nosed each year. It is the second most common genitouri- ommendations with regards to surgery, adjunctive sys-
nary cancer and results in over 12,000 cancer-related temic therapy, and alternative therapies.
deaths annually.1 Twenty percent to 25% of newly diag-
nosed bladder cancer consist of muscle-invasive disease,2
TUMOR EXTENT EVALUATION
with the majority of these invasive bladder cancers
Stage and Grade
demonstrating invasion at the time of diagnosis.3 While
tumor grade and stage do influence progression, only Transurethral resection (TUR) of a bladder tumor yields
approximately 15% of superficial tumors will eventually vital information to the urologist. The objective is to
develop the characteristic features of muscle-invasion.4–6 remove all visible tumors, if possible, and obtain tissue
For those that become muscle-invasive, however, the risk for pathologic evaluation. The two most commonly used
of metastasis and mortality unquestionably and dramati- prognostic indicators, clinical stage and histologic grade
cally rises.7 of the cancer, can be determined from this operative pro-
More than 90% of invasive and noninvasive bladder cedure. TUR under anesthesia is both diagnostic and
tumors are transitional cell carcinomas (TCCs). Invasive therapeutic, especially in cases of superficial disease.
bladder cancer, however, can be of squamous cell carci- Invasive bladder cancer consists of malignant transitional
noma (3%), adenocarcinoma (2%), or small cell epithelial cells that extend beyond the connective tissue
carcinoma (<1%) origin. TCCs may also contain focal of the lamina propria and into the muscularis propria
areas of squamous or glandular differentiation. These (T2) and surrounding perivesical fat and soft tissues
have been traditionally classified and managed as TCC, (T3,4). For this reason, the presence of muscle detrusor
but future investigation may demonstrate that these his- cells in the resected tissue is essential to provide patho-
tologic variants are in fact associated with different out- logic evidence of muscle invasion.
comes and should be treated as different pathologic Local muscular invasion classically has been described
entities. This chapter will deal specifically with the prog- by three mechanisms.8 The majority (60%) advance “en
nosis and management of invasive TCC of the bladder. bloc,” as a broad face below the primary epithelial lesion.
The management of muscle-invasive and metastatic Twenty-five percent spread with tentacle-like fronds.
TCC continues to evolve. The advent of multimodality The remainder (10% to 15%) spread by laterally
therapy has begun to challenge the traditional view of encroaching under normal adjacent mucosa. A more
radical cystectomy as the “gold standard” treatment for modern biochemical interpretation involves concepts of
muscle-invasive bladder cancer. A multitude of new stimulation of neovascularization, the regulation of cellu-
information about the cellular mechanisms and biology lar motility, and the avoidance of detection from cellular
of bladder cancer has led to trials ultimately designed to surveillance mechanisms.
improve patient outcome and survival. Herein, we will The presence and degree of local invasion has been
review the principles and concepts behind the biology of closely linked to poor outcomes both in time to metastasis

338
Chapter 19 Prognosis and Management of Invasive Transitional Cell Carcinoma 339

and time to cancer-related death. According to the frequent sites of metastasis are the pelvic lymph nodes.
National Cancer Data Base, the overall 5-year survival Among the pelvic lymph nodes, the obturator nodes and
rates for Ta, T1, T2, T3, and T4 tumors are 90%, 87%, the external iliac nodes are involved most commonly in
65%, 48%, and 23%, respectively.7 In other studies, the cases with positive nodes, 74% and 65% of the time,
median survival for T2 tumors is 85 months, whereas respectively. The presacral nodes (25%), common iliac
for T4 tumors, the median survival is 29 months.9 nodes (20%), and paravesical nodes (16%) are involved
Furthermore, a recent review of 1636 cases demonstrated with TCC less often.24 Other distant sites of metastasis
a significant variation with stage and survival following for TCC include bone, lung, skin, liver, and less com-
radical cystectomy; 63% of patients with T2 disease sur- monly, the brain and meninges.25 Few patients with
vived 5 years, while only 18% of those with T4 disease distant metastases survive 5 years.26 Clearly, the identifi-
survived as long.10 There is clear and persuasive evidence cation of metastasis affects the decision-making process
to suggest that the depth of infiltration correlates with for tumor thought to be locally invasive.
curability of TCC. In order to complete the pretreatment clinical staging,
While this review focusses on muscle-invasive bladder the urologist must survey the patient to detect the most
cancer, there are forms of non–muscle-invasive TCC common metastatic lesions. In addition to performing a
that have such a high propensity to invade that they merit full physical examination, including bimanual palpation
inclusion. Carcinoma in situ (CIS) is confined to the of the bladder before and after tumor resection,27 labora-
urothelium, but 20% of patients treated with cystectomy tory studies, such as renal and hepatic function panel,
for CIS are found to contain some elements of micro- should be performed. In addition, a chest radiograph is
scopic invasion.11 Between 40% and 83% of CIS tumors appropriate to document evidence of pulmonary nodules
will ultimately progress to muscle-invasion.5 Likewise, consistent with tumor metastasis.
T1 tumors have been shown to become invasive about Often, the initial test performed in evaluation of TCC
one-half of the time.12 In addition, one-third have is a computerized tomographic (CT) scan of the
involvement of the posterior urethra, which, as discussed abdomen and pelvis.28,29 While this is a simple test and
later, affects surgical treatment options.13 may assist in the detection of distant metastatic lesions,
Pathologic T1 tumors may require a different surgical bulky retroperitoneal adenopathy, or obstructive hydro-
management than other noninvasive bladder tumors ureteronephrosis, it has repeatedly been shown to be
because of their propensity for invasion.14 Recently inaccurate in assessing the stage of the primary cancer,
revised recommendations of management of high-grade especially with small volume pelvic nodal disease.30,31 In
(II and III) T1 cancers include transurethral re-resection addition to understaging tumors by not reliably detecting
of the previous tumor bed because incomplete resec- pelvic lymphadenopathy, CT also can overstage tumors
tion is closely linked with invasion15,16 and because of the because of the difficulty in distinguishing postbiopsy
risk of understaging.17 edema from residual tumor. Despite these shortcomings,
In addition to tumor depth, the grade, or the degree of CT scan remains the standard preoperative staging test.
differentiation, of the tumor cells is of critical prognostic Because CT scanning is inadequate to detect pelvic
significance.18 Grading has traditionally been applied to nodes, MRI has been studied to determine better the
superficial tumors to predict recurrence or progres- extent of tumor.32,33 MRI can be particularly beneficial in
sion.19,20 Patients with grade 1 and grade 2 tumors have patients who have renal insufficiency or a contrast allergy
approximately 10% risk of developing muscle-invasion, and cannot tolerate CT. The rate of detection of micro-
whereas one-third of patients with grade 3 tumors will scopic metastasis has compared similarly to CT,34 but
eventually have invasive disease.21 More recently, efforts some advocate its use in the staging of patients with
to grade muscle-invasive components have not demon- deeply invasive TCC.35 There is still a significant amount
strated any prognostic significance in T2 disease.22 of “mis-staging” (40%) of bladder tumors with MRI.36
Other less commonly used methods to detect invasive Combined with its high cost, these doubts reduce the fre-
disease and predict its course, such as the use of DNA quency of its use. Future advances in MRI, including
ploidy and the expression of various growth factors and endorectal coil enhancement37 and dynamic contrast
cell-cycle regulators, will be addressed in detail later. imaging,38 may make it more accurate, but they have not
been validated.
The role of PET scanning has also been investigated
Metastatic Evaluation
for use in invasive TCC.39–41 An intrinsic problem is that
Prior to proceeding with management of locally invasive fluoro-deoxy-glucose (FDG), the most commonly used
bladder cancer, the physician must determine the systemic radiopharmaceutical, is unsuitable for evaluation of blad-
extent of the tumor, specifically, if the cancer has metas- der cancer due to intense accumulation in the urine.42
tasized. A close correlation between muscle-invasion and Because of this inability to image the bladder, efforts have
distant metastasis has been well documented.23 The most been made to employ more specific tracers, which are not
340 Part IV Bladder

excreted in the urine. Early studies indicate that carbon-11 niques used in the investigation of TCC is cDNA
labeled choline may be helpful.43 Again, the cost of the microarrays. This is a high output method of studying the
study and current inaccuracies, however, make PET expression of thousands of genes on one tumor extract on
impractical for routine use. The most likely use of PET a single slide rapidly and reproducibly. Many groups are
scanning in invasive bladder cancer may be to assist in employing this technique to try and determine the molec-
detection of positive lymph nodes or distant metastatic ular profile of TCC.53–55 It is becoming apparent that
lesions. there may not be a uniform treatment for every patient
The most accurate method to stage regional lymph with muscle-invasive bladder cancer. Rather, clinically
nodes remains surgical, regional lymph node dissection. relevant subsets of bladder cancer patients may be identi-
Performed as a separate procedure, it can reliably distin- fied, and selective treatments may be required for patients
guish N+ from N0 patients and can be performed laparo- with specific tumor characteristics.56
scopically.44 While most believe there is an insignificant While many correlations have been made with tumor
risk of port site tumor recurrence,45 others have argued progression, the use of markers has not been accepted
that the risk may be greater in patients with bladder can- into standard practice yet.57 The molecular changes that
cer. One study shows a 4% risk of port site recurrence occur in the progression of invasive bladder cancer have
following pelvic node dissection for bladder cancer com- been examined as potential prognostic factors and can be
pared to a negligible risk following lymphadenectomy for organized into 3 categories: (1) chromosomal alterations;
prostate cancer.46 The reason for this discrepancy (2) cell adhesion molecules and angiogenesis; and (3) loss
between TCC and other genitourinary malignancies is of cell-cycle regulation involving tumor suppressor genes
unclear. The majority of time, however, regional lymph and oncogenes.
node dissection is performed at the time of radical cys-
tectomy at a single operative setting.
Chromosomal Abnormalities
As in many malignancies, the role of chromosomal
PROGNOSIS OF INVASIVE TRANSITIONAL CELL
abnormalities and DNA cytometry has been evaluated
CARCINOMA
for bladder cancer.58 Abnormal number of chromosomes
Even in organ-confined, muscle-invasive bladder cancer and chromosomes of abnormal morphology have been
(stage pT2N0M0) treated by radical cystectomy, approx- associated with an increased risk of bladder cancer recur-
imately 50% of patients ultimately develop recurrences. rence and progression.59,60 Several studies correlate
In one recent study of 594 patients with organ-confined polysomy, especially of chromosome 7, or aneuploidy
invasive cancer treated locally, 44% relapsed.47 Others with progression of superficial bladder cancer.61–64 There
have also shown 5-year survival rates following cystec- has been very little work, however, that can demonstrate
tomy for locally invasive bladder cancer to be 38% to the same prognostic significance for muscle-invasive
64%.7,48–51 Most patients with occult metastases develop transitional carcinoma. In invasive cancer, DNA ploidy
overt clinical evidence of distant metastasis within does not appear to statistically correlate with invasive
1 year.25 cancer in stage, tumor recurrence, lymph node metasta-
The above data imply that the current clinical staging sis, or survival.65–67 In addition, abnormal chromosomal
and histologic grading systems on which urologists rely number or configuration has not been correlated with
are insufficient. Except for the patients with extensive response to treatment. In patients with muscle-invasive
local disease or bulky lymphadenopathy, contemporary disease, DNA ploidy has not predicted outcome for those
preoperative evaluation continues to understage patients receiving either preoperative radiation therapy68,69 or
with invasive TCC. While significant thought has been neoadjuvant chemotherapy.70
devoted to which superficial tumors will progress to inva-
sion, equally intense research effort has been devoted to
Growth Events
predicting which invasive tumors will metastasize. What
has been shown is that there is a vast degree of biological The progression of TCC to invasion and metastasis
heterogeneity to urothelial cancer. By combing through requires a cascade of molecular events. Malignant cells
this cellular variability, it may be possible to identify par- must extend beyond their boundaries into the lamina
ticular genetic characteristics that will predict failure of propria and subsequently, the muscularis mucosa and
conventional therapy. The identification of any prognos- perivesical fat. This process relies on cellular motility,
tic factors for muscle-invasive bladder cancer would be of proteolysis, proliferation, and angiogenesis. There is
critical clinical utility. mounting evidence to suggest that alterations in the mol-
Advances in molecular genetics may provide the ecules that govern cell growth enable this progression. In
opportunity to better determine an invasive bladder addition, if one is able to detect at the molecular level
tumor’s biological potential.52 One of the newest tech- these processes, one may identify a real-time marker for
Chapter 19 Prognosis and Management of Invasive Transitional Cell Carcinoma 341

progression. The two most investigated of these growth metalloproteinases, that contribute to the destruction of
control markers as indicators of prognostic significance the basement membrane. Their role in the process of
in invasive TCC are cell adhesion molecules and angio- invasion of superficial tumors has been well described.82
genesis factors. Matrix metalloproteinases, however, have been suggested
to play a role in the progression of invasion to metastasis
as well.83 Their expression has been shown to be of prog-
Cell Adhesion Molecules
nostic significance in those patients who develop recur-
Various properties of neoplastic cells are altered, which rence of their TCC following what appeared to be
enables invasion. These involve the relationship between complete extirpation of their invasive cancers.84
cells, components of the extracellular matrix, and the
underlying basal lamina. The physical relationships
Angiogenic Factors
between cells are controlled by cell adhesion molecules
like cadherins. E-cadherin, in particular, has been inves- Like all solid tumors, TCC of the bladder requires angio-
tigated and has been demonstrated to be a critical factor genesis to promote progression and growth. The angio-
in facilitating the progression of bladder cancer.71 Not genic phenotype—the sum of the balance of stimulatory
only is loss of E-cadherin associated with an invasive phe- and inhibitory signals to the endothelium—not only
notype but also its normal expression decreases in direct determines the degree of neovascularity but also is evolv-
proportion to the tumor stage.72 Others have also shown ing into an important prognostic factor in the outcome of
abnormal E-cadherin expression to be an independent bladder cancer.85 Some of the variables, which have been
predictor of disease progression following cystectomy, studied, include vascular endothelial growth factor
demonstrating an inverse relationship with stage, grade, (VEGF), thrombospondin-1 (TSP-1), and microvascular
likelihood of lymph node metastasis, and even overall density.
survival.73 Several groups have looked at VEGF as a mediator of
Investigation is also being performed on the prognos- tumor angiogenesis. The VEGF serum level in patients
tic value of E-cadherin-related cytoplasmic molecules. with bladder cancer was significantly associated with
Cadherin function requires an intact network of catenins tumor stage, grade, vascular invasion, and metastasis.86
to network through the cytoskeleton. Abnormal expres- VEGF expression has also been identified by multivariate
sions of alpha-, beta-, and gamma-catenin have been analysis as a significant predictor of disease recurrence in
shown to significantly correlate with tumor stage, grade, patients who have undergone cystectomy with neoadju-
and survival in patients with invasive bladder cancer.74 vant chemotherapy.87
Other cellular adhesion molecules include the inte- Thrombospondin-1 (TSP-1), an important compo-
grins family, which are transmembrane proteins that link nent of the extracellular matrix, has also been identified
epithelial cells to each other and the basal membrane. as a potential prognostic angiogenic factor. It is a potent
Critical cell–cell interaction occurs via integrins and the inhibitor of neovascularity and a reduction in its gene
anchoring complex of desmosomes. In addition to con- expression has been associated with disease progression
tributing to the spread of CIS,75 abnormal expression of of superficial TCC.88 Other groups have examined TSP-
integrins have been linked to the proliferation of invasive 1 expression in invasive disease. TSP-1 was significantly
bladder cancer. It has been shown that loss of the coex- associated with disease recurrence and overall survival
pression of alpha-6-beta-4 integrin and collagen VII, a but not seen to behave independently of other tumor
component of this extracellular network, is prominent in markers like p53.89 This may indicate that TSP-1 may
progressively invasive urothelial cancer.76 have some inhibitory function but may be one mecha-
Malignant urothelium has also been associated with nism of action of p53.
abnormal expression of other cell-surface molecules like A different method of quantifying vascular develop-
epidermal growth factor receptors. The interaction of ment is to evaluate microvessel density (MVD). MVD
these signaling molecules induces a wide variety of has been shown to be independently influential to disease
changes within the cell nucleus, including mitosis. recurrence and survival in muscle-invasive bladder can-
Abnormal expression or production of these gene prod- cer.87,90–92. In patients with an elevated MVD, there is a
ucts may therefore upregulate growth and promote can- significantly higher rate of occult lymph node metastases
cer cell motility.77 Multiple groups have demonstrated in patients with clinically localized disease.93 Histologic
that the traditional signs of tumor aggressiveness, such as efforts to assess other shape-related morphometric char-
stage, grade, and survival, correlate with abnormal acteristics of the microvascularization as indicators of sig-
expression of epidermal growth factor.78–81 nificant neovascularity have been demonstrated in
Cellular motility through the extracellular matrix relies superficial disease. In muscle-invasive disease, however,
on an array of enzymes like collagenases, which provide MVD is the only significant histologic indicator of prog-
proteolysis. One such family of enzymes is the matrix nosis that has been identified.94
342 Part IV Bladder

While a majority of studies have found a positive cor- As suggested earlier, not all investigators have found a
relation between MVD and worsening prognosis, not all direct link between p53 expression and prognosis in
reports concur. Some have found MVD to be not signif- patients with muscle-invasive disease. One group found
icantly associated with lymph node status in patients with no correlation between mutation in the p53 and cancer-
high-stage, high-grade bladder cancer.66 As will be specific survival. Also, it has held no prognostic value in
addressed, many of the intergroup outcome variability patients with T3b or greater disease106 or node-positive
may be secondary to differing treatments. It remains to disease.107 These varying results may be due to multiple
be seen how treatment affects the upregulation of these reasons including differences in assays, technique, and
various markers of progressive invasion. tumor heterogeneity.

Cell-Cycle Regulators—p53 Cell-Cycle Regulators—Others


TCC is thought to develop from genetic changes, which Other molecules that regulate the cell cycle have been
affect cell growth and proliferation. Two such genes, associated with bladder cancer, and thus, have been
which can regulate the cell cycle, are tumor suppressor investigated for their potential prognostic significance
genes and proto-oncogenes. Both have been extensively when detected in abnormal levels. One such gene is the
studied in TCC.95 retinoblastoma gene. It appears on chromosome 13q and
The p53 is a tumor suppressor gene located on chro- when its protein product is phosphorylated by cyclin-
mosome 17p. It encodes a 5-kD protein that induces dependent kinases, it activates genes critical to DNA
apoptosis in cells with evidence of DNA damage. replication.
Approximately one-half of all human cancers have evi- The retinoblastoma gene appears mutated in approxi-
dence of mutations in the p53 gene.96 These mutations mately one-third of bladder tumors.108 Loss of normal
permit unabated cell growth by disrupting normal cell- retinoblastoma function has been shown to result in sta-
cycle checkpoints and apoptosis.97 There is evidence to tistically significant higher grade of TCC, along with a
support that p53 may also act by promoting other angio- higher likelihood of invasion.109 Others have shown that
genic factors already mentioned, such as VEGF and abnormal expression of retinoblastoma in patients who
TSP-1,98 although this is debated particularly in the have undergone radical cystectomy for muscle-invasive
angiogenesis of bladder cancer.99 disease has a significant higher rate of recurrence and
There is a vast amount of evidence to suggest that the decreased survival.110 Interestingly, in the same study,
dysregulation of p53 is critical in the development of those with both retinoblastoma dysregulation and p53
TCC.100 When p53 undergoes mutation, its half-life typ- mutation fared more poorly than either individually. This
ically increases, which causing accumulation of p53 in the may suggest a synergistic relationship between retinoblas-
cell nuclei. Advances in immunohistochemistry and toma and p53 in the promotion of tumor progression.
molecular genetics have allowed researchers to detect Similarly, any other genetic mutation that affects the
easily the cellular buildup of abnormal p53. cyclin-dependent kinase systems, which phosphorylate
p53 mutations occur in the pathogenesis of TCC rel- the retinoblastoma gene product, may also promote cel-
atively early, and researchers have attempted to corre- lular proliferation and malignant transformation.111 The
late varying expressions of abnormal p53 with patient p21 is a ras oncogene that transduces signals from the cell
outcome.101 While p53 has been associated with high membrane to the nucleus and is actually induced by wild-
rate of tumor progression in high-risk superficial dis- type p53.112 This particular kinase inhibitor, under
ease,102 there has been inconsistent evidence to suggest altered expression, has been shown to contribute to
the same for invasive bladder cancer. The nuclear over- tumor progression. Mutated p21 emerges in several stud-
expression of mutant p53 does appear to be more com- ies as an independent indicator of increased likelihood of
mon in muscle-invasive disease according to a large recurrence and of decreased disease-free survival in
meta-analysis from the International Study Initiative on patients undergoing radical cystectomy for muscle-inva-
Bladder Cancer. Of 1706 patients, p53 mutations sive disease.113,114 A deficiency of p21 expression has also
appeared in only 25% of superficial disease versus 48% been shown to be correlated with poor response follow-
of muscle-invasive disease.103 It has been shown that ing systemic chemotherapy115 and radiation therapy.116
nuclear overexpression in T2 or greater disease is asso- Another oncogene that has been suggested as a useful
ciated with a worse prognosis, resulting in higher stage prognostic index for bladder cancer is erb-B-2. This
and grade, earlier development of metastases, and oncogene codes for a growth factor receptor, which is
decreased survival.104,105 In other studies, p53 status was functionally related to epidermal growth factor receptor.
the only independent predictor of survival in those Its amplification and overexpression have been impli-
patients with clinically localized disease at the time of cated as prognostic markers for recurrent superficial
cystectomy.106 bladder tumors.117 Researchers, however, have been
Chapter 19 Prognosis and Management of Invasive Transitional Cell Carcinoma 343

unable to link protein overexpression to tumor stage, Investigators have evaluated the prognostic signifi-
grade, or disease progression in invasive bladder cancer cance of p53 alteration and overexpression with regards
patients.118,119. In terms of invasive TCC, erb-B-2 to radiation therapy as well. As compared to chemother-
appears to not possess much predictive value at the pres- apy, there is less evidence to suggest that there is a reli-
ent time. able correlation in treatment outcomes. Several groups
Transforming growth factor (TGF) is both a cellular have indicated no correlation of p53 status to local con-
regulator and a potent angiogenic factor. Serum levels of trol, delay to metastasis, or overall survival.126–128 Others
TGF have been shown to be more elevated in patients have suggested that p53 overexpression is associated with
with muscle-invasive cancer than with superficial can- radiosensitivity,129,130 while yet others have argued for a
cer.120 A recent work suggests that preoperative TGF- correlation between radioresistance and overexpres-
beta was an independent predictor of lymph-vascular sion.131 These varying reports indicate that p53 is not a
invasion, lymph node metastasis, disease recurrence, and reliable prognostic indicator for response to radiation
cancer-specific survival.121 therapy for invasive bladder cancer.
Another molecule to consider regarding prognosis of
treatment is P-glycoprotein, formerly known as the mul-
Genetic Determinants of Treatment Response
tidrug resistance (MDR) gene. Certain cytotoxic drugs
While the cellular components of TCC may someday have been demonstrated to be exported from tumor cells
elucidate its degree of aggressiveness, these markers may through a multidrug effusion pump. This pump activity
also serve to predict and determine tumor responsiveness is characterized by expression of P-glycoprotein, and can
to systemic chemotherapy. Theoretically, the molecules theoretically result in decreased efficacy of chemothera-
critical to cellular proliferation could modify tumor peutic agents. While the phenomenon of upregulation of
response by impairing apoptosis and altering cell-cycle P-glycoprotein has been well documented following sys-
kinetics. Thus, they may provide further prognostic temic chemotherapy for bladder cancer,132 the data con-
information for patient management. cerning drug resistance and P-glycoprotein expression
The p53 tumor suppressor gene, by preventing pro- have been variable.133
grammed cell death, can enable cells to undergo malig- Another molecule with possibly some prognostic sig-
nant transformation. Likewise, if certain drugs rely on nificance for therapy is the intracellular scavenger, glu-
p53-mediation to achieve apoptosis, subtle mutations in tathione (GSH). GSH has been shown to react with free
p53 may render that bladder cancer line more chemoresis- cisplatin, thus reducing the intracellular availability of
tant for those medications. For example, the overexpression the drug. The levels of GSH are upregulated in some
of p53 has been shown to be an independent prognostic fac- lines of TCC and it is possible that this contributes to the
tor for methotrexate, vinblastine, doxorubicin, and cisplatin mechanism of cisplatin resistance.134 In other cancer
(MVAC) failure and decreased survival in patients with T2 models, the administration of GSH inhibitors has been
or T3a bladder cancer.122 In fact, it has been suggested that shown to increase response to cisplatin,135,136 but this has
other agents like paclitaxel and gemcitabine be used in blad- not yet been applied to bladder cancer.
der cancer patients with p53 mutations.123
While some studies predict p53 as a marker for
Conclusion
chemoresistance for certain cytotoxic agents, there have
been equal data to suggest that p53 overexpression is The ultimate importance of the aforementioned data on
associated with good treatment response. Some have clinical decision-making is still unclear. TCCs are clearly
identified p53-mutated tumors to respond better to adju- variable and complex. The continued unveiling of the
vant cis-platinum-based chemotherapy prior to cystec- intricacies of the cancer’s cellular biology will likely one
tomy.56,124 Likewise, there have been several studies that day help determine prognosis and direct appropriate ther-
demonstrate no correlation between p53 expression and apy. For now, the knowledge produced from investiga-
response to chemotherapy. The p53 expression did not tions of the past will more specifically direct future clinical
affect response to chemotherapy and did not predict over- studies, thus, making them more clinically applicable.
all survival for patients with invasive bladder cancer.125
In summary, the p53 status and response rate are con-
RADICAL CYSTECTOMY
flicting regarding systemic chemotherapy and may
depend on the particular chemotherapeutic agent. One Radical cystectomy remains the gold standard therapeu-
particular reason for this may be the fact that p53 may be tic option for patients with muscle-invasive bladder car-
induced by exposure to chemotherapeutic agents. cinoma, and in the U.S., it is the most common
Therefore, uniform timing of the sampling and adminis- treatment option. Despite alternatives that may appear
tration of the chemotherapy are essential for completely appealing and may be appropriate for certain individuals,
evaluating this prognostic parameter. surgical removal still provides local control and chance
344 Part IV Bladder

for cure, to which other treatments attempt to compare 4.5%, 5%, and 8% of men with pTa/T1, pT2, and pT3,
themselves. respectively. At 1-year follow-up, 97% are fully continent
during the day and 95% are continent at night.
Additionally, 82% of patients maintained potency with
Preoperative Evaluation and Preparation
retrograde ejaculation due to the TUR, while 10% are
Patients with invasive bladder cancer often are elderly partially potent and 8.1% are impotent. Clearly, long-
with significant competing comorbidities who require term follow-up will be required to ensure continued
careful preoperative evaluation, and it is often the sur- excellent cancer control. As with other carcinomas, the
geon’s responsibility to ensure that an appropriate work- importance of achieving a negative surgical margin is
up has been performed. Recently, Miller et al.137 have paramount to obtaining local cancer control and should
demonstrated an association between comorbid illness not be compromised.145
and adverse pathological and survival outcome following A better understanding of the anatomic relationships
radical cystectomy. It is ultimately the surgeon’s respon- of the neurovascular bundles with respect to the prostate
sibility to try and optimize the patient’s health status combined with improved surgical technique has led
prior to cystectomy, and to determine if a patient is not a to major improvements in functional outcomes.146
candidate for major surgery. It is important to realize that Retaining potency in men following radical cystectomy is
the goal of the preoperative assessment is to assess risk also technically feasible and involves careful attention to
among these potential surgical candidates. Several the area of the prostatic pedicles, where nerve bundle
reports have now confirmed the procedure that it can be injury is most likely.147–149 Currently, it has been reported
performed safely with acceptable morbidity among the that potency is preserved in about 50% of men with
elderly, including those with high perioperative nerve-sparing techniques and there has been no apparent
risk.138–140 Despite more “conservative” approaches, such compromise in cancer control.149
as bladder sparing, age and even comorbidities should Urinary reconstruction following cystectomy involves
not be used as absolute deterrents when considering rad- the reconnection of the ureters to an intestinal segment
ical cystectomy as therapy for a potentially lethal cancer. that allows for the drainage of urine. There are essen-
Patients are seen preoperatively by an enterostomal tially three methods in which this can be accomplished:
nurse for counseling and optimal stoma site marking. an ileal conduit, a continent cutaneous reservoir, or an
This is recommended regardless of diversion type orthotopic urinary diversion. Historically, the majority of
planned in the event that an orthotopic diversion is not patients underwent an ileal conduit, an incontinent form
technically feasible or contraindicated. Prior to surgery, of urine drainage. Over the past decade, however, conti-
patients undergo a mechanical and antibiotic bowel nent urinary diversions, especially orthotopic bladder
preparation usually performed in the outpatient setting reconstruction, have evolved from experimental surgery
after 1 to 2 days of a clear liquid diet. Patients are rou- to become a commonly preformed method of urinary
tinely admitted on the day of surgery with an estimated diversion. The detection of cancer in younger patients,
length of stay of between 5 and 7 days. the desire to maintain a normal body image, and the real-
ization that quality of life can be improved by maintain-
ing near-normal function after radical surgery, and
Radical Cystectomy in Males
improved surgical techniques have all contributed to the
Radical cystectomy is the standard treatment in the sur- realization that continent urinary diversion is an option
gical management of male patients with muscle-invasive for a significant percentage of patients.150,151
bladder cancer.141 In a male, the prostate and seminal Patient selection criteria include both patient factors
vesicles are removed routinely along with the bladder and and cancer factors. Relative contraindications to ortho-
is the equivalent of an anterior exenteration. Reasons for topic urinary diversion include renal insufficiency (serum
routine removal of the prostate include direct extension creatinine > 2 ng/dl), hepatic dysfunction, and the inabil-
of the bladder cancer into the prostatic urethra, as well as ity to perform self-catheterization should the need arise.
prostatic ductal or stromal involvement. There is also a An absolute contraindication in a male patient is the
risk of secondary malignancy, namely, prostatic adeno- inability to achieve a negative cancer margin in the prox-
carcinomas, which has been detected in up to 40% of imal urethra (prostatic apex). Although controversial, age
specimens in historical series.142,143 alone is not a contraindication to orthotopic diversion
Recently, however, a series of 100 men who under- and thus can be performed judiciously in men over the
went prostate sparing at the time of radical cystectomy age of 70 years, and even in men with significant comor-
was reported.144 After careful patient selection, patients bidities.152 Prior pelvic radiation therapy is again a rela-
underwent TUR of the prostate with frozen section tive contraindication; however, there have been reports
analysis at the time of cystectomy. In the absence of can- of successful orthotopic diversion in the setting of salvage
cer in the TUR specimen, local recurrence occurred in cystectomy for failed radiation.153
Chapter 19 Prognosis and Management of Invasive Transitional Cell Carcinoma 345

Increasing experience has expanded the possible appli- sion, orthotopic urinary diversion has become increas-
cability of orthotopic diversion. The impact of local recur- ingly viable as an option. Stein et al.162 and others subse-
rence on ileal neobladder function and survival was recently quently have demonstrated the oncologic safety of
reported in a series of 357 men.154 Local recurrence devel- orthotopic reconstruction in properly selected female
oped in 43 patients (12%). Of the 43 patients with local patients. Exclusion criteria for orthotopic neobladder
recurrence at follow-up, 36 had local advanced cancer on reconstruction include tumor involving the bladder neck,
the final pathological evaluation (stage pT3a or node-posi- diffuse CIS, and a positive bladder neck margin at the
tive or greater), yet 40 maintained good neobladder func- time of radical cystectomy.162 In addition, females with
tion. The neobladder was removed only in 1 patient due to large, palpable tumors along the anterior vaginal wall are
a neobladder to intestinal fistula. These data imply that not appropriate candidates. In properly selected patients,
most patients may anticipate normal neobladder function local recurrence rates have been extremely low and func-
even in the presence of recurrent disease or until death. In tional outcomes have been comparable to those reported
addition, although it is unclear what the real likelihood is, among male patients.163–167
the retained urethra in any form of urinary diversion must The technique and outcomes of orthotopic diversion
be carefully followed as a possible site of recurrence. in females have been well described.164,165 These techni-
Orthotopic neobladder construction involves the cre- cal refinements include avoidance of overlapping suture
ation of an internal reservoir from some segment of bowel, lines, the interposition of a vascularized omental pedicle,
most commonly this is 45 to 60 cm of ileum, detubularized and preservation of the anterior vaginal wall.163 In
and fashioned in either a “W configuration” (Hautmann) patients with nonpalpable tumors, the plane between the
or “U shaped or J shaped” (Studer).150,155 While in the past posterior bladder wall and the anterior vaginal wall can
there was a fear that orthotopic diversion would be associ- be developed while ligating the posterior-lateral pedicles.
ated with significantly higher rate of complication as com- The plane is developed to the level of the bladder neck,
pared to an ileal conduit, recent reports suggest that in and the anterior and posterior dissections are connected
properly selected patients the early complications rates are with preservation of the bladder neck.
similarly acceptable.151,156 It was recently reported that there is a very low inci-
The functional results following orthotopic urinary dence of secondary gynecologic malignancies at the time
reconstruction have been excellent. Daytime urinary of cystectomy.168 In this series, only a single gynecologic
continence rates range from 80% to 95% with differ- malignancy was found and involvement of the uterus by
ences largely due to definition of continence, methods in direct extension of bladder cancer was discovered in only
which the data are obtained, and length of follow- 2 patients, both of whom had clinical suspicion based on
up.157–160 Nighttime continence is more problematic, bimanual examination or preoperative imaging. Thus, in
with rates of 65% to 85% reported. The incidence of most women, the risk of gynecologic involvement of uro-
hyper-continence or urinary retention requiring self- logic malignancy is small and can usually be determined
catheterization is approximately 3% to 5%. Men should either preoperatively or at the time of surgery. The
be counseled preoperatively that while most men are potential for improved functional outcomes and quality
continent in the day, they may experience some degree of of life through preservation of gynecological organs, par-
incontinence at night and a protective pad may be ticularly among young women with invasive bladder can-
required. Overall patient satisfaction with orthotopic cer, is currently an area of ongoing research, and these
neobladder substitution remains high with >95% of younger women are more likely to be concerned about
patients satisfied with their choice of diversion.161 preservation of fertility and continuation of normal hor-
monal status.169
In properly selected patients, functional outcomes
Radical Cystectomy in Females
have been comparable to those reported among male
In women, radical cystectomy for muscle-invasive blad- patients.163–167 Daytime continence rates range from
der cancer has historically been the equivalent of an ante- 70% to 95%, with high rates of overall satisfaction.
rior exenteration. This includes removal of the uterus, There may be a higher rate of urinary retention regard-
fallopian tubes, ovaries, bladder, urethra, and a segment ing intermittent catheterizations, and all patients
of anterior vaginal wall. This remains the gold standard. undergoing diversion should receive preoperative
However, early detection combined with a desire to counseling regarding this, as well as other possible
improve the functional outcomes, including sexual abili- complications.
ties and urinary control, has led surgeons to modify their
techniques in selected patients, where preservation of
Role of Lymphadenectomy
disease-free urethra is possible.
Although the majority of women still undergo ileal Although it has long been a standard part of radical cys-
conduit urinary diversion or continent cutaneous diver- tectomy, pelvic lymph node dissection has garnered more
346 Part IV Bladder

attention recently. This is derived in part from data can be achieved with excellent long-term survival
demonstrated long-term disease-free survival among afforded as well.
node-positive patients and benefit implied from the com-
pleteness of the lymph node dissection. It has been pro-
Complications of Radical Cystectomy
posed that the therapeutic benefit of lymphadenectomy
may be extended beyond the confines of the traditional Despite significant decrease in overall mortality and
pelvic template arguing in favor of a more extended node morbidity rates associated with cystectomy, complica-
dissection.47,170,171 Recent studies have proposed that the tions can occur as an exacerbation of the patients preex-
actual number of nodes harvested may play a role in isting comorbid conditions, arising from the bladder
patient outcomes, and the greater the lymph nodes removal and those arising from the use of an intestinal
removed the better the survival.172,173 segment. The mortality rate for radical cystectomy
The concept of lymph node density, defined as the remains 1% to 2%, with a an overall early complication
total number of positive lymph nodes divided by the total rate of about 25% to 30%.47,140,181–183 In one recent
number of lymph nodes removed, has recently been series of 304 patients, the overall major and minor com-
reported to be an important prognostic indicator among plication rates were 4.9% and 30.9%, respectively, within
patients with node-positive bladder cancer.174 The bene- the first 30 days.184 Postoperative ileus was the most
fit of an extended dissection may be derived from both common minor complication, affecting 18% of patients,
the removal of clinically apparent pathologic nodes which resolved with conservative management in all
and/or from undetected micrometastatic disease. Future cases. Despite these improved results, the serious life-
studies that carefully document the overall number and threatening major complications, such as pulmonary
location of lymph nodes removed (mapping) along with emboli, myocardial infarction, and stroke, do occur and
the number and location of positive nodes removed at the prompt recognition with appropriate intervention may
time of cystectomy using defined templates of dissection avoid mortality.
and possibly molecular staging will be important. Radical cystectomy has been associated with significant
blood loss and/or transfusion requirement, and compli-
cating the surgical treatment of these patients is the sig-
Outcomes of Radical Cystectomy
nificant number that have preoperative anemia. In one
The long-term efficacy of radical cystectomy among series, 45% of patients had anemia preoperatively.
patients with invasive bladder cancer has been demon- Median estimated blood loss was 600 ml (range 100 to
strated in terms of local control and disease-specific sur- 3000). Increased estimated blood loss was related to
vival. Numerous series have demonstrated excellent local patient age; American Society of Anesthesiologists (ASA)
control and excellent 5- and 10-year survivals for patients score longer operative time and paralytic ileus. Overall,
with invasive bladder cancer treated with cystec- transfusion was required in 30% of patients with a median
tomy.149,175–178 Long-term survival is best among those requirement of 2 units (range 1 to 10). The transfusion
with organ-confined disease, and consistently 5-year sur- rate in male and female patients was 26% and 40%,
vival is >70%.47,179,180 respectively.185 These data support the need for continued
The presence of locally advanced or lymph node refinement in surgical techniques designed to decrease
metastases is associated with a high rate of recurrence blood loss, as well as for strategies designed to lower the
and argues in favor of combined neoadjuvant or adjuvant need for blood transfusion during radical cystectomy.
chemotherapy. In the largest contemporary series to
date, investigators at the University of Southern
Role of Chemotherapy: Neoadjuvant Versus
California (USC) recently reported results in 1054
Adjuvant
patients with a median follow-up of 10.2 years.47 In this
series, the overall recurrence-free survival at 5- and 10-years The rationale for including chemotherapy regimens
was 68% and 60%, respectively. In patients with muscle- before or after radical cystectomy is based on the pre-
invasive disease (P2a and P3a), lymph node negative sumption that this therapy may increase patient survival.
tumors had 89% and 87%, and 78% and 76% 5- and 10-year Clearly, TCC is a chemosensitive malignancy and
recurrence-free survival, respectively. In patients with patients with metastatic disease can achieve a long-term
nonorgan-confined bladder cancer (P3b, P4), lymph cure.186–189 Even patients with large, bulky unresectable
node-negative cancer demonstrated a significantly higher disease that cannot be cured by surgery alone can survive
probability or recurrence with 5- and 10-year recur- with the addition of chemotherapy for their treatment.190
rence-free survival rates for P3b tumors were 62% and Neoadjuvant chemotherapy in patients with advanced
61% and for P4 tumors were 50% and 45%, respectively. disease is becoming increasingly touted as the possible
Clearly, these data support the aggressive surgical man- standard approach. This is based largely on two recent
agement of invasive bladder cancer. Durable local control trials that have reported a benefit. An intergroup trial
Chapter 19 Prognosis and Management of Invasive Transitional Cell Carcinoma 347

conducted by the Medical Reserve Crops (MRC) and the radical cystectomy in itself clearly has a negative impact
EORTC is the largest randomized trial (976 enrolled on patient pathology and survival.201–203 This negative
patients) examining neoadjuvant chemotherapy (CMV). impact on delay is true even in patients who undergo
The patients underwent some form of localized therapy some type of therapy, such as radiation therapy, as veri-
(cystectomy alone, radiation therapy alone, or cystec- fied by better survival in patients who undergo early cys-
tomy and radiation therapy) after 3 cycles of CMV. tectomy as opposed to late cystectomy.204
Although initially there was no survival difference, the
updated results did demonstrate a survival difference
BLADDER-SPARING THERAPY: A MULTIMODALITY
in those patients receiving neoadjuvant chemotherapy
PARADIGM
(p = 0.048).191 The second large trial that took almost
Rationale for Bladder Preservation
10 years to enroll its 307 patients was the SWOG-initiated
Intergroup 0080 trial.192 The results are compelling; but In certain patients, radical cystectomy may not be the
as in most trials, criticisms can be made of methodology, best option, and bladder preservation strategies for the
as well as interpretation of results. The SWOG study uti- treatment of invasive cancer have evolved for several rea-
lized a one-sided testing and when two-sided hypothesis sons. Advances in the treatments of other malignant
testing was performed, there was no statistical advantage processes, such as breast and esophageal cancer, have
in survival with neoadjuvant chemotherapy. Other stud- demonstrated the effectiveness of this strategy. In bladder
ies that in themselves had viable criticisms have not cancer, the majority of patients with recurrent and
demonstrated a survival benefit.193–195 metastatic disease die of disease that is distant in location
Arguments exist for both approaches when consider- as opposed to local. In addition, despite its accepted stan-
ing chemotherapy regimens. With adjuvant chemother- dard as the treatment of choice, radical cystectomy does
apy, there is better pathologic staging and assessment of not ultimately result in a cure in a large number of
true disease burden within each patient.196 Recently, patients and still represents a significant operative proce-
Herr et al.197 updated the Memorial Sloan-Kettering dure with associated complications. Many bladder cancer
experience with postchemotherapy surgery in patients patients are elderly, and although the number is decreas-
with unresectable or lymph node positive bladder can- ing with improvements in surgical techniques and peri-
cer. Of 207 patients with unresectable or regionally operative care, there are patients who are “too ill” to
metastatic bladder cancer, 80 (39%) underwent undergo radical cystectomy.
postchemotherapy surgery after treatment with a cis- Although this approach has its appeal it remains only
platin-based chemotherapy regimen. No viable cancer an option, and radical cystectomy continues to be the
was present at postchemotherapy surgery in 24 of the 80 standard treatment of choice. Survival data for bladder-
cases (30%), pathologically confirming a complete sparing approach presented in a certain light can first
response to chemotherapy. Of the 24 patients without appear promising, but it is difficult to compare directly
residual cancer, 14 (58%) survived 9 months to 5 years how patient survival rates differ between bladder-sparing
after salvage surgery. Residual viable cancer was com- regimens and initial radical cystectomy. This is especially
pletely resected in 49 patients (61%), resulting in a com- true when a significant number of patients do eventually
plete response to chemotherapy plus surgery and 20 require cystectomy, which is in fact only delayed by the
(41%) survived. bladder sparing approach. This strategy often requires a
Unfortunately, randomized trials with small numbers multitude of visits to different physicians requiring often-
have given mixed results. The two studies that did not invasive procedures to attempt to monitor closely the
show a benefit to adjuvant chemotherapy examined non- patient’s condition and tumor status. In addition, blad-
metastatic disease patients.198,199 Stockle et al.200 reported der-sparing protocols are not in themselves inherently
on 166 patients and demonstrated that those patients risk-free and have associated possible complications. To
most likely to gain benefit from immediate chemotherapy date, no prospective randomized trial has been per-
were those with small volume lymph node involvement. formed comparing a multimodality bladder-sparing
Trials are currently in progress, including an examination approach with radical cystectomy.
of the impact of p53 status. Other trials looking at more
advanced and node-positive patients are also accruing
Single Modality Approaches
patients, including an EORTC trial examining surveil-
lance versus immediate chemotherapy (M-VAC or gemc- The success of TUR as a single modality has been used
itabine plus cisplatin) after surgery. The CALGB 90104 for many years depends on clinical stage and proper
study treats all patients with extravesical and/or node- patient selection. Although an option, TUR alone can-
positive disease with a cis-platinum-based regimen. not adequately treat the majority of patients with inva-
As with any treatment modality, proper patient selec- sive carcinoma. Barnes et al.205 reported a 27% 5-year
tion is essential but in TCC it can be difficult. Delaying survival in patients with T2 bladder cancer. In highly
348 Part IV Bladder

selected series, 5-year survival rates have varied but


Multimodality Approach
have been as high as 67% in patients still with their
bladder.206–208 Single modality bladder-sparing therapy for muscle-inva-
Clearly, this approach is predicated on accurate evalu- sive bladder cancer, including TUR, partial cystectomy,
ation and staging of the true and limited extent of muscle systemic chemotherapy, or radiation therapy have been
invasion. Even after repeated resections, however, occult demonstrated to result in insufficient local control when
invasive disease may still be present that would not be compared to cystectomy.223 In an attempt to maximize
adequately treated by TUR alone.209 A single TUR, thus, the efficacy of less invasive approaches and overcome
would be an inadequate therapy. One of the major pitfalls their deficiencies, bladder-sparing approaches have com-
to only endoscopic management of these tumors is the bined different strategies to offer a comparable alterna-
continued limitations of clinical understaging, which is tive to radical cystectomy. This inclusion of more than
present in 30% to 50% of cases.17 This is also true in one approach seems to offer better oncologic cure rates
patients treated with radiation therapy or neoadjuvant and achieves bladder preservation at a higher rate than a
chemotherapy.196,210 In a selected few patients with lim- single treatment choice.193,224–228 TUR and radiation
ited disease of minimal depth and with no residual dis- therapy hopefully provide local control with chemother-
ease on repeat resection, this modality does have a role.15 apy providing a synergistic additive component and
By extending the boundaries of TUR, another blad- treating possible micrometastatic disease.
der-sparing therapy approach using surgery is partial cys- Multiple variations of treatment algorithms have been
tectomy. Once again, this approach could be advocated studied. This large number of studies makes comparisons
in a limited group of patients. Those patients who may difficult. In general, however, after patients are diagnosed
be reasonable candidates would include small, single with muscle-invasive bladder cancer, they undergo “com-
tumors, away from the trigone without related plete” resection of their tumor for local control, as well
CIS.211–213 Although bladder capacity may be enough to as attempting as accurate staging as possible. This is then
maintain normal voiding patterns, this remaining bladder followed by chemotherapy and external beam radiation.
tissue serves as an area for recurrent disease or previously Oftentimes, the chemotherapy includes a platinum-based
unrecognized disease. Localized recurrence in the form radiosensitizing chemotherapeutic agent (e.g., cisplatin).
of new tumors or recurrent tumors occur anywhere Patients are then carefully reevaluated by repeat cys-
between 15% and 75% of the time.211,212,214 This form of toscopy and TUR. Only those patients with a complete
therapy should not be accepted as a standard treatment response are candidates for continued bladder-sparing
choice. approach; those without complete response are recom-
Radiation therapy as a single modality is commonly mended to undergo radical cystectomy. If bladder spar-
employed in Europe and has been studied in the U.S. ing is viable, patients receive additional radiation and
Conventional radiation therapy has local control rates of chemotherapy with continued careful surveillance by cys-
30% to 50%.215–218 At M.D. Anderson, 135 patients were toscopy, physical exam, and radiographs.
treated with primary external beam radiation therapy,
with a 26% 5-year survival rate. Those patients with
Outcomes of Bladder-Sparing Approach
more advanced disease had a poor survival rate and less
than a third achieved long-term local control.219 Similar Approximately 40% of patients survive 5 years with a
survival rates have been demonstrated overseas.220,221 bladder-sparing approach.193,224–228 In one of the largest
Patients who receive monotherapy radiation treat- randomized trials evaluating the role of chemotherapy in
ment tend to be older with greater comorbidities. But multimodality bladder preservation, the Radiation
significant side effects, such as radiation cystitis, cannot Therapy Oncology Group (RTOG) reported an overall
be ignored.222 Unfortunately, as opposed to TUR or par- 49% 5-year survival with an intact functioning bladder in
tial cystectomy, any type of salvage surgery is difficult, in 38%.193 The local recurrence rate in this series was 28%,
one large series, <30% of patients who did not achieve a and the study was stopped prematurely due to severe
complete response to radiation underwent salvage cystec- complications from the chemotherapy, including
tomy.215 Radiation as the only primary therapy has not 3 deaths. Among these patients treated with bladder
been considered as equal treatment modality to cystec- preservation strategies, life-long surveillance is required
tomy. Similarly, the role of chemotherapy continues to not only to monitor for progressive disease but also for
evolve but in no way should it be considered as a single superficial recurrences that may culminate in cystectomy
treatment choice or as an acceptable alternative treat- in up to a third of these patients.229 Importantly, recur-
ment choice to cystectomy. It cannot consistently eradi- rence can occur after 5 years of disease-free survival.
cate the primary bladder tumor. This is the case even Also, induction treatment many times is not successful as
when there is an impressive response to disease outside of up to 40% have an incomplete response and are recom-
the bladder. mended to undergo cystectomy.193 One recent study of a
Chapter 19 Prognosis and Management of Invasive Transitional Cell Carcinoma 349

bladder-sparing approach, although not prospectively patients, quality of life may be as important as length of
randomizing patients, did find better overall survival in life or survival. Furthermore, among the long-term sur-
those patients who did at some point undergo cystectomy vivors of cancer therapy, the impact of cancer treatment
(65% versus 40%).230 on the HRQOL is even more profound, especially with
radical cystectomy, one of the most potentially stressful
and life-altering.233 One of the key reasons for bladder
Complications of Bladder-Sparing Approach
sparing approach is an attempt to avoid the morbidity
Bladder-sparing approaches may also be associated with and possible complications associated with loss of the
adverse therapy-related side effects that can result in sig- bladder. Patients oftentimes do maintain adequate blad-
nificant morbidity that negatively impacts on quality of der function after chemotherapy and/or radiation, but
life. Many patients cannot complete the recommended the different types of urinary diversion now available
protocol of chemotherapy because of toxicity. This make cystectomy and urinary diversion at least somewhat
include nausea, fatigue, neutropenia, and GI complaints, more acceptable if not desirable.231,234
such as diarrhea in up to 70% of patients.193 Also, there is Although more studies have been performed examin-
mortality associated with only the induction portion of ing the impact of cystectomy on quality of life, many of
this algorithm (4% in one series193). This is actually a these are inherently flawed.161,235–239 Most of these stud-
higher mortality rate than most cystectomy series. ies have compared quality of life in patients undergoing
Furthermore, the long-term effects of these multimodal- various types of urinary diversion and have been criti-
ity therapies may result in significant increased morbidity cized for flaws in methodology. In addition, most are ret-
among those patients who demonstrate recurrent or pro- rospective and so the actual quality of life comparison
gressive disease and ultimately require radical cystectomy. from baseline is not known. Also, as investigators have
One important point is that although a patient may toler- started realizing the value of validated instruments when
ate the chemotherapy and radiation changes to the blad- gauging outcome after treatment,233,240 it has become
der, symptoms that the patient may have caused by the clear that one does not yet exist for bladder cancer. It is
disease process and/or previous therapy (e.g., TURBTs, important, therefore, to keep in mind that despite the
intravesical therapies) are not at all altered or improved by perception that patients with neobladders enjoy a better
bladder sparing. Such bladder dysfunction may be better quality of life compared to those with ileal conduits, the
addressed by bladder removal and urinary diversion. In definitive study remains to be performed.228 Large,
addition, bowel symptoms and effect on small bowel and prospective longitudinal studies using both generic and
the rectum must also be considered. Moderate or severe disease-specific validated instruments are required.
symptoms can affect up to one-third of patients.231
In its present form, multimodality therapy aimed at
SUMMARY
bladder-preservation is best performed at select centers
that have a dedicated and cooperative multidisciplinary The diagnosis and treatment of muscle-invasive TCC
team that can provide these patients with close clinical fol- continues to evolve. With each patient, multiple issues
low-up. Patients should also be made aware of the selec- including patient selection, complexity of care, cost, sur-
tion criteria and specifics of the associated with the various vival outcomes, and quality of life measures must be care-
treatment protocols, including cost, before making an fully evaluated. An improved understanding of tumor
informed decision. Finally, the results of these various biology coupled with a multimodality approach treat-
strategies should be objectively balanced against the results ment strategy will hopefully provide continued improved
of contemporary radical cystectomy and orthotopic diver- results. Radical cystectomy currently remains the gold
sion. Attempting to spare the bladder requires complex standard for patients with invasive bladder cancer; and
and constant surveillance and assessment of patient status. modifications of surgery, urinary diversion, and perioper-
There has to be a coordinated effort between patient and ative care continue to translate into improved quality of
a number of disciplines, including surgery, radiation life for patients. Continued research to discover and val-
oncology, and oncology. This requires a dedicated effort idate accurate tumor markers will provide further benefit
by a team approach.225 Although not a primary concern, for these patients.
this approach is costly regardless of preservation of the
bladder or ultimately requiring cystectomy.232
REFERENCES
Quality of Life Concerns
1. Greenlee RT, Murray T, Bolden S, Wingo PA: Cancer
Increasingly, health-related quality of life (HRQOL) has statistics. CA Cancer J Clin 2000; 50:7.
been recognized as an important outcome measure fol- 2. Messing EM, Young TB, Hunt VB, et al: Comparison of
lowing the treatment of urologic malignancies. For many bladder cancer outcome in men undergoing hematuria
350 Part IV Bladder

home screening versus those with standard clinical 22. Jimenez RE, Gheiler E, Oskanian P, et al: Grading the
presentations. Urology 1995; 45:387. invasive component of urothelial carcinoma of the
3. Kaye KW, Lange PH: Mode of presentation of invasive bladder and its relationship with progression-free
bladder cancer: reassessment of the problem. J Urol survival. Am J Surg Pathol 2000; 24:980.
1982; 128:31. 23. Jewett HJ, strong GH: Infiltrating carcinoma of the
4. Kurth KH, Denis L, Bouffioux C, et al: Factors affecting bladder: relation of depth of penetration of the bladder
recurrence and progression in superficial bladder wall to incidence of local extension and metastasis.
tumours. Eur J Cancer 1995; 31A:1840. J Urol 1946; 55:366.
5. Althausen AF, Prout GR Jr, Daly JJ: Non-invasive 24. Smith JA Jr, Whitmore WF Jr: Regional lymph node
papillary carcinoma of the bladder associated with metastasis from bladder cancer. J Urol 1981; 126:591.
carcinoma in situ. J Urol 1976; 116:575. 25. Babaian RJ, Johnson DE, Llamas L, Ayala AG:
6. Lutzeyer W, Rubben H, Dahm H: Prognostic Metastases from transitional cell carcinoma of urinary
parameters in superficial bladder cancer: an analysis of bladder. Urology 1980; 16:142.
315 cases. J Urol 1982; 127:250. 26. Loehrer PJ Sr, Einhorn LH, Elson PJ, et al: A
7. Fleshner NE, Herr HW, Stewart AK, et al: The randomized comparison of cisplatin alone or in
National Cancer Data Base report on bladder carcinoma. combination with methotrexate, vinblastine, and
The American College of Surgeons Commission on doxorubicin in patients with metastatic urothelial
Cancer and the American Cancer Society. Cancer 1996; carcinoma: a cooperative group study. J Clin Oncol
78:1505. 1992; 10:1066.
8. Jewett HJ, Eversole SL, Jr: Carcinoma of the bladder: 27. Fossa SD, Ous S, Berner A: Clinical significance of the
characteristic modes of local invasion. J Urol 1960; “palpable mass” in patients with muscle-infiltrating
83:383. bladder cancer undergoing cystectomy after
9. Prout GR Jr: Bladder carcinoma and a TNM system of pre-operative radiotherapy. Br J Urol 1991; 67:54.
classification. J Urol 1977; 117:583. 28. Hodson NJ, Husband JE, MacDonald JS: The role of
10. Herr HW: Uncertainty, stage and outcome of invasive computed tomography in the staging of bladder cancer.
bladder cancer. J Urol 1994; 152:401. Clin Radiol 1979; 30:389.
11. Farrow GM, Utz DC, Rife CC: Morphological and 29. Kellett MJ, Oliver RT, Husband JE, Fry IK: Computed
clinical observations of patients with early bladder cancer tomography as an adjunct to bimanual examination for
treated with total cystectomy. Cancer Res 1976; 36:2495. staging bladder tumours. Br J Urol 1980; 52:101.
12. Cheng L, Bostwick DG: Progression of T1 bladder 30. Herr HW: Routine CT scan in cystectomy patients: does
tumors: better staging or better biology? reply. Cancer it change management? Urology 1996; 47:324.
1999; 86: 910. 31. Paik ML, Scolieri MJ, Brown SL, Spirnak JP, Resnick
13. Matzkin H, Soloway MS, Hardeman S: Transitional cell MI: Limitations of computerized tomography in staging
carcinoma of the prostate. J Urol 1991; 146:1207. invasive bladder cancer before radical cystectomy. J Urol
14. Holzbeierlein JM, Smith JA Jr: Surgical management of 2000; 163:1693.
noninvasive bladder cancer (stages Ta/T1/CIS). Urol 32. Barentsz JO, Jager GJ, Witjes JA, Ruijs JH: Primary
Clin North Am 2000; 27:15. staging of urinary bladder carcinoma: the role of MRI
15. Herr HW: The value of a second transurethral resection and a comparison with CT. Eur Radiol 1996; 6:129.
in evaluating patients with bladder tumors. J Urol 1999; 33. Tekes A, Kamel IR, Imam K, et al: MR imaging features
162:74. of transitional cell carcinoma of the urinary bladder. AJR
16. Brauers A, Buettner R, Jakse G: Second resection and Am J Roentgenol 2003; 180:771.
prognosis of primary high risk superficial bladder cancer: 34. Jager GJ, Barentsz JO, Oosterhof GO, Witjes JA, Ruijs
is cystectomy often too early? J Urol 2001; 165:808. SJ: Pelvic adenopathy in prostatic and urinary bladder
17. Dutta SC, Smith JA Jr, Shappell SB, et al: Clinical under carcinoma: MR imaging with a three-dimensional
staging of high risk nonmuscle invasive urothelial TI-weighted magnetization-prepared-rapid gradient-
carcinoma treated with radical cystectomy. J Urol 2001; echo sequence. Am J Roentgenol 1996; 167:1503.
166:490. 35. Robinson P, Collins CD, Ryder WD, et al: Relationship
18. Reuter VE: Pathology of bladder cancer: assessment of of MRI and clinical staging to outcome in invasive
prognostic variables and response to therapy. Semin bladder cancer treated by radiotherapy. Clin Radiol
Oncol 1990; 17:524. 2000; 55:301.
19. Jordan AM, Weingarten J, Murphy WM: Transitional 36. Buy JN, Moss AA, Guinet C, et al: MR staging of
cell neoplasms of the urinary bladder. Can biologic bladder carcinoma: correlation with pathologic findings.
potential be predicted from histologic grading? Cancer Radiology 1988; 169:695.
1987; 60:2766. 37. Hayashi N, Tochigi H, Shiraishi T, Takeda K, Kawamura
20. Kaubisch S, Lum BL, Reese J, Freiha F, Torti FM: Stage J: A new staging criterion for bladder carcinoma using
T1 bladder cancer: grade is the primary determinant for gadolinium-enhanced magnetic resonance imaging with
risk of muscle invasion. J Urol 1991; 146:28. an endorectal surface coil: a comparison with
21. Torti FM, Lum BL, Aston D, et al: Superficial bladder ultrasonography. BJU Int 2000; 85:32.
cancer: the primacy of grade in the development of 38. Lawler LP: MR imaging of the bladder. Radiol Clin
invasive disease. J Clin Oncol 1987; 5:125. North Am 2003; 41:161.
Chapter 19 Prognosis and Management of Invasive Transitional Cell Carcinoma 351

39. Harney JV, Wahl RL, Liebert M, et al: Uptake of 57. Stein JP, Grossfeld GD, Ginsberg DA, et al: Prognostic
2-deoxy, 2-(18F) fluoro-D-glucose in bladder cancer: markers in bladder cancer: a contemporary review of the
animal localization and initial patient positron emission literature. J Urol 1998; 160:645.
tomography. J Urol 1991; 145:279. 58. Wheeless LL, Badalament RA, de Vere White RW,
40. Letocha H, Ahlstrom H, Malmstrom PU, et al: Positron Fradet Y, Tribukait B: Consensus review of the clinical
emission tomography with L-methyl-11C-methionine in utility of DNA cytometry in bladder cancer. Report of
the monitoring of therapy response in muscle-invasive the DNA Cytometry Consensus Conference. Cytometry
transitional cell carcinoma of the urinary bladder. Br J 1993; 14:478.
Urol 1994; 74:767. 59. Sandberg AA: Chromosome changes in bladder cancer:
41. Heicappell R, Muller-Mattheis V, Reinhardt M, et al: clinical and other correlations. Cancer Genet Cytogenet
Staging of pelvic lymph nodes in neoplasms of the 1986; 19:163.
bladder and prostate by positron emission tomography 60. Falor WH, Ward-Skinner RM: The importance of
with 2-[(18)F]-2-deoxy-D-glucose. Eur Urol 1999; marker chromosomes in superficial transitional cell
36:582. carcinoma of the bladder: 50 patients followed up to 17
42. Kosuda S, Fisher S, Wahl RL: Animal studies on the years. J Urol 1988; 139:929.
reduction and/or dilution of 2-deoxy-2-[18F]fluoro- 61. Waldman FM, Carroll PR, Kerschmann R, et al:
D-glucose (FDG) activity in the urinary system. Ann Centromeric copy number of chromosome 7 is strongly
Nucl Med 1997; 11:213. correlated with tumor grade and labeling index in human
43. de Jong IJ, Pruim J, Elsinga PH, et al: Visualisation of bladder cancer. Cancer Res 1991; 51:3807.
bladder cancer using (11)C-choline PET: first clinical 62. Norming U, Tribukait B, Nyman CR, Nilsson B, Wang
experience. Eur J Nucl Med Mol Imaging 2002; 29:1283. N: Prognostic significance of mucosal aneuploidy in
44. Poulsen J, Krarup T: Pelvic lymphadenectomy (staging) stage Ta/T1 grade 3 carcinoma of the bladder. J Urol
in patients with bladder cancer laparoscopic versus open 1992; 148:1420.
approach. Scand J Urol Nephrol Suppl 1995; 172:19. 63. Richter J, Wagner U, Schraml P, et al: Chromosomal
45. Tsivian A, Sidi AA: Port site metastases in urological imbalances are associated with a high risk of progression
laparoscopic surgery. J Urol 2003; 169:1213. in early invasive (pT1) urinary bladder cancer. Cancer
46. Elbahnasy AM, Hoenig DM, Shalhav A, McDougall Res 1999; 59:5687.
EM, Clayman RV: Laparoscopic staging of bladder 64. Watters AD, Going JJ, Grigor KM, Bartlett JM:
tumor: concerns about port site metastases. J Endourol Progression to detrusor-muscle invasion in bladder
1998; 12:55. carcinoma is associated with polysomy of chromosomes 1
47. Stein JP, Lieskovsky G, Cote R, et al: Radical and 8 in recurrent pTa/pT1 tumours. Eur J Cancer 2002;
cystectomy in the treatment of invasive bladder cancer: 38:1593.
long-term results in 1054 patients. J Clin Oncol 2001; 65. Hosaka Y: DNA ploidy pattern of flow cytometry as
19:666. indicator of malignancy and prognosis in bladder cancer.
48. Richie JP, Skinner DG, Kaufman JJ: Carcinoma of the Nippon Rinsho 1992; 50:2426.
bladder: treatment by radical cystectomy. J Surg Res 66. Lianes P, Charytonowicz E, Cordon-Cardo C, et al:
1975; 18:271. Biomarker study of primary nonmetastatic versus
49. Prout GR Jr: The surgical management of bladder metastatic invasive bladder cancer. National Cancer
carcinoma. Urol Clin North Am 1976; 3:149. Institute Bladder Tumor Marker Network. Clin Cancer
50. Pearse HD, Reed RR, Hodges CV: Radical cystectomy Res 1998; 4:1267.
for bladder cancer. J Urol 1978; 119:216. 67. Desgrippes A, Izadifar V, Assailly J, Fontaine E, Beurton
51. Raghavan D, Shipley WU, Garnick MB, Russell PJ, D: Diagnosis and prediction of recurrence and
Richie JP: Biology and management of bladder cancer. progression in superficial bladder cancers with DNA
N Engl J Med 1990; 322:1129. image cytometry and urinary cytology. BJU Int 2000;
52. Raghavan D: Molecular targeting and 85:434.
pharmacogenomics in the management of advanced 68. Jacobsen AB, Pettersen EO, Amellem O, et al: The
bladder cancer. Cancer 2003; 97:2083. prognostic significance of deoxyribonucleic acid flow
53. Sanchez-Carbayo M, Socci ND, Charytonowicz E, et al: cytometry in muscle invasive bladder carcinoma treated
Molecular profiling of bladder cancer using cDNA with preoperative irradiation and cystectomy. J Urol
microarrays: defining histogenesis and biological 1992; 147:34.
phenotypes. Cancer Res 2002; 62:6973. 69. Granfors T, Duchek M, Tomic R, Roos G, Ljungberg B:
54. Dyrskjot L, Thykjaer T, Kruhoffer M, et al: Identifying Predictive value of DNA ploidy in bladder cancer treated
distinct classes of bladder carcinoma using microarrays. with preoperative radiation therapy and cystectomy.
Nat Genet 2003; 33:90. Scand J Urol Nephrol 1996; 30:281.
55. Sanchez-Carbayo M: Use of high-throughput DNA 70. Turkolmez K, Baltaci S, Beduk Y, Muftuoglu YZ, Gogus
microarrays to identify biomarkers for bladder cancer. O: DNA ploidy and S-phase fraction as predictive factors
Clin Chem 2003; 49:23. of response and outcome following neoadjuvant
56. Tiguert R, Lessard A, So A, Fradet Y: Prognostic methotrexate, vinblastine, epirubicin and cisplatin
markers in muscle invasive bladder cancer. World J Urol (M-VEC) chemotherapy for invasive bladder cancer.
2002; 20:190. Scand J Urol Nephrol 2002; 36:46.
352 Part IV Bladder

71. Imao T, Koshida K, Endo Y, et al: Dominant role of prognostic factor in bladder cancer. J Urol 2001;
E-cadherin in the progression of bladder cancer. J Urol 166:1275.
1999; 161:692. 87. Inoue K, Slaton JW, Karashima T, et al: The prognostic
72. Sun W, Herrera GA: E-cadherin expression in urothelial value of angiogenesis factor expression for predicting
carcinoma in situ, superficial papillary transitional cell recurrence and metastasis of bladder cancer after
carcinoma, and invasive transitional cell carcinoma. Hum neoadjuvant chemotherapy and radical cystectomy. Clin
Pathol 2002; 33:996. Cancer Res 2000; 6:4866.
73. Bringuier PP, Umbas R, Schaafsma HE, et al: Decreased 88. Goddard JC, Sutton CD, Jones JL, O’Byrne KJ,
E-cadherin immunoreactivity correlates with poor Kockelbergh RC: Reduced thrombospondin-1 at
survival in patients with bladder tumors. Cancer Res presentation predicts disease progression in superficial
1993; 53:3241. bladder cancer. Eur Urol 2002; 42:464.
74. Shimazui T, Schalken JA, Giroldi LA, et al: Prognostic 89. Grossfeld GD, Ginsberg DA, Stein JP, et al:
value of cadherin-associated molecules (alpha-, beta-, Thrombospondin-1 expression in bladder cancer:
and gamma-catenins and p120cas) in bladder tumors. association with p53 alterations, tumor angiogenesis, and
Cancer Res 1996; 56:4154. tumor progression. J Natl Cancer Inst 1997; 89:219.
75. Harabayashi T, Kanai Y, Yamada T, et al: Reduction of 90. Dickinson AJ, Fox SB, Persad RA, et al: Quantification
integrin beta 4 and enhanced migration on laminin in of angiogenesis as an independent predictor of prognosis
association with intraepithelial spreading of urinary in invasive bladder carcinomas. Br J Urol 1994; 74:762.
bladder carcinomas. J Urol 1999; 161:1364. 91. Bochner BH, Cote RJ, Weidner N, et al: Angiogenesis in
76. Liebert M, Washington R, Wedemeyer G, Carey TE, bladder cancer: relationship between microvessel density
Grossman HB: Loss of co-localization of alpha 6 beta and tumor prognosis. J Natl Cancer Inst 1995; 87:1603.
4 integrin and collagen VII in bladder cancer. Am J 92. Chaudhary R, Bromley M, Clarke NW, et al: Prognostic
Pathol 1994; 144:787. relevance of micro-vessel density in cancer of the urinary
77. Theodorescu D, Laderoute KR, Gulding KM: bladder. Anticancer Res 1999; 19:3479.
Epidermal growth factor receptor-regulated human 93. Jaeger TM, Weidner N, Chew K, et al: Tumor
bladder cancer motility is in part a phosphatidylinositol angiogenesis correlates with lymph node metastases in
3-kinase-mediated process. Cell Growth Differ 1998; invasive bladder cancer. J Urol 1995; 154:69.
9:919. 94. Korkolopoulou P, Konstantinidou AE, Kavantzas N,
78. Messing EM: Clinical implications of the expression of et al: Morphometric microvascular characteristics predict
epidermal growth factor receptors in human transitional prognosis in superficial and invasive bladder cancer.
cell carcinoma. Cancer Res 1990; 50:2530. Virchows Arch 2001; 438:603.
79. Neal DE, Sharples L, Smith K, et al: The epidermal 95. Sidransky D, Messing E: Molecular genetics and
growth factor receptor and the prognosis of bladder biochemical mechanisms in bladder cancer. Oncogenes,
cancer. Cancer 1990; 65:1619. tumor suppressor genes, and growth factors. Urol Clin
80. Messing EM: Growth factors and bladder cancer: clinical North Am 1992; 19:629.
implications of the interactions between growth factors 96. Levine AJ: p53, the cellular gatekeeper for growth and
and their urothelial receptors. Semin Surg Oncol 1992; division. Cell 1997; 88:323.
8:285. 97. Barbacid M: Oncogenes in human cancers and in
81. Mellon K, Wright C, Kelly P, Horne CH, Neal DE: chemically induced animal tumors. Prog Med Virol
Long-term outcome related to epidermal growth 1985; 32:86.
factor receptor status in bladder cancer. J Urol 1995; 98. Bouck N: p53 and angiogenesis. Biochim Biophys Acta
153:919. 1996; 1287:63.
82. Ozdemir E, Kakehi Y, Okuno H, Yoshida O: Role of 99. Reiher FK, Ivanovich M, Huang H, et al: The role of
matrix metalloproteinase-9 in the basement membrane hypoxia and p53 in the regulation of angiogenesis in
destruction of superficial urothelial carcinomas. J Urol bladder cancer. J Urol 2001; 165:2075.
1999; 161:1359. 100. Smith ND, Rubenstein JN, Eggener SE, Kozlowski JM:
83. Ocharoenrat P, Modjtahedi H, Rhys-Evans P, et al: The p53 tumor suppressor gene and nuclear protein:
Epidermal growth factor-like ligands differentially up- basic science review and relevance in the management of
regulate matrix metalloproteinase 9 in head and neck bladder cancer. J Urol 2003; 169:1219.
squamous carcinoma cells. Cancer Res 2000; 60:1121. 101. Grossfeld GD, Muscheck M, Stein JP, et al: Cellular
84. Gohji K, Fujimoto N, Fujii A, et al: Prognostic proliferation and cell-cell cycle regulatory proteins as
significance of circulating matrix metalloproteinase-2 to prognostic markers for transitional cell carcinoma of the
tissue inhibitor of metalloproteinases-2 ratio in bladder. Adv Exp Med Biol 1999; 462:425.
recurrence of urothelial cancer after complete resection. 102. Lacombe L, Dalbagni G, Zhang ZF, et al:
Cancer Res 1996; 56:3196. Overexpression of p53 protein in a high-risk population
85. Jayet C, Deperthes D, Leisinger HJ: Angiogenesis and of patients with superficial bladder cancer before and
bladder cancer: trendy prognostic factor or new after bacillus Calmette-Guerin therapy: correlation to
therapeutic target? Ann Urol (Paris) 2002; 36:258. clinical outcome. J Clin Oncol 1996; 14:2646.
86. Bernardini S, Fauconnet S, Chabannes E, et al: Serum 103. Schmitz-Drager BJ, Goebell PJ, Ebert T, Fradet Y: p53
levels of vascular endothelial growth factor as a immunohistochemistry as a prognostic marker in bladder
Chapter 19 Prognosis and Management of Invasive Transitional Cell Carcinoma 353

cancer. Playground for urology scientists? Eur Urol carcinoma using competitive PCR technique. J Urol
2000; 38:691. 1996; 156:2089.
104. Esrig D, Elmajian D, Groshen S, et al: Accumulation of 120. Eder IE, Stenzl A, Hobisch A, et al: Transforming
nuclear p53 and tumor progression in bladder cancer. growth factors-beta 1 and beta 2 in serum and urine
N Engl J Med 1994; 331:1259. from patients with bladder carcinoma. J Urol 1996;
105. Hemal AK, Khaitan A, Dinda AK, et al: Prognostic 156:953.
significance of p53 nuclear overexpression in patients of 121. Shariat SF, Kim JH, Andrews B, et al: Preoperative
muscle invasive urinary bladder carcinoma treated with plasma levels of transforming growth factor beta(1)
cystectomy. Urol Int 2003; 70:42. strongly predict clinical outcome in patients with bladder
106. Herr HW, Bajorin DF, Scher HI, Cordon-Cardo C, carcinoma. Cancer 2001; 92:2985.
Reuter VE: Can p53 help select patients with invasive 122. Sarkis AS, Bajorin DF, Reuter VE, et al: Prognostic value
bladder cancer for bladder preservation? J Urol 1999; of p53 nuclear overexpression in patients with invasive
161:20. bladder cancer treated with neoadjuvant MVAC. J Clin
107. Fleshner N, Kapusta L, Ezer D, Herschorn S, Klotz L: Oncol 1995; 13:1384.
p53 nuclear accumulation is not associated with 123. Kielb SJ, Shah NL, Rubin MA, Sanda MG: Functional
decreased disease-free survival in patients with node p53 mutation as a molecular determinant of paclitaxel
positive transitional cell carcinoma of the bladder. J Urol and gemcitabine susceptibility in human bladder cancer.
2000; 164:1177. J Urol 2001; 166:482.
108. Horowitz JM, Park SH, Bogenmann E, et al: Frequent 124. Cote RJ, Esrig D, Groshen S, Jones PA, Skinner DG:
inactivation of the retinoblastoma anti-oncogene is p53 and treatment of bladder cancer. Nature 1997;
restricted to a subset of human tumor cells. Proc Natl 385:123.
Acad Sci U S A 1990; 87:2775. 125. Qureshi KN, Griffiths TR, Robinson MC, et al: TP53
109. Cairns P, Proctor AJ, Knowles MA: Loss of accumulation predicts improved survival in patients
heterozygosity at the RB locus is frequent and correlates resistant to systemic cisplatin-based chemotherapy for
with muscle invasion in bladder carcinoma. Oncogene muscle-invasive bladder cancer. Clin Cancer Res 1999;
1991; 6:2305. 5:3500.
110. Cote RJ, Dunn MD, Chatterjee SJ, et al: Elevated and 126. Jahnson S, Risberg B, Karlsson MG, et al: p53 and Rb
absent pRb expression is associated with bladder cancer immunostaining in locally advanced bladder cancer:
progression and has cooperative effects with p53. Cancer relation to prognostic variables and predictive value for
Res 1998; 58:1090. the local response to radical radiotherapy. Eur Urol
111. Williams SG, Buscarini M, Stein JP: Molecular markers 1995; 28:135.
for diagnosis, staging, and prognosis of bladder cancer. 127. Ogura K, Habuchi T, Yamada H, Ogawa O, Yoshida O:
Oncology (Huntingt) 2001; 15:1461. Immunohistochemical analysis of p53 and proliferating
112. Cordon-Cardo C: Mutations of cell cycle regulators. cell nuclear antigen (PCNA) in bladder cancer: positive
Biological and clinical implications for human neoplasia. immunostaining and radiosensitivity. Int J Urol 1995;
Am J Pathol 1995; 147:545. 2:302.
113. Stein JP, Ginsberg DA, Grossfeld GD, et al: Effect of 128. Wu CS, Pollack A, Czerniak B, et al: Prognostic value of
p21WAF1/CIP1 expression on tumor progression in p53 in muscle-invasive bladder cancer treated with
bladder cancer. J Natl Cancer Inst 1998; 90:1072. preoperative radiotherapy. Urology 1996; 47:305.
114. Korkolopoulou P, Konstantinidou AE, Thomas-Tsagli E, 129. Ribeiro JC, Barnetson AR, Fisher RJ, Mameghan H,
et al: WAF1/p21 protein expression is an independent Russell PJ: Relationship between radiation response and
prognostic indicator in superficial and invasive bladder p53 status in human bladder cancer cells. Int J Radiat
cancer. Appl Immunohistochem Mol Morphol 2000; Biol 1997; 2:11.
8:285. 130. Rotterud R, Berner A, Holm R, Skovlund E, Fossa SD:
115. Jankevicius F, Goebell P, Kushima M, et al: p21 and p53 p53, p21 and mdm2 expression vs the response to
immunostaining and survival following systemic radiotherapy in transitional cell carcinoma of the
chemotherapy for urothelial cancer. Urol Int 2002; 69:174. bladder. BJU Int 2001; 88:202.
116. Qureshi KN, Griffiths TR, Robinson MC, et al: 131. Ong F, Moonen LM, Gallee MP, et al: Prognostic factors
Combined p21WAF1/CIP1 and p53 overexpression in transitional cell cancer of the bladder: an emerging
predict improved survival in muscle-invasive bladder role for Bcl-2 and p53. Radiother Oncol 2001; 61:169.
cancer treated by radical radiotherapy. Int J Radiat Oncol 132. Petrylak DP, Scher HI, Reuter V, O’Brien JP, Cordon-
Biol Phys 2001; 51:1234. Cardo C: P-glycoprotein expression in primary and
117. Sauter G, Moch H, Moore D, et al: Heterogeneity of metastatic transitional cell carcinoma of the bladder. Ann
erbB-2 gene amplification in bladder cancer. Cancer Res Oncol 1994; 5:835.
1993; 53:2199. 133. Shinohara N, Liebert M, Wedemeyer G, Chang JH,
118. Underwood M, Bartlett J, Reeves J, et al: C-erbB-2 gene Grossman HB: Evaluation of multiple drug resistance in
amplification: a molecular marker in recurrent bladder human bladder cancer cell lines. J Urol 1993; 150:505.
tumors? Cancer Res 1995; 55:2422. 134. Pendyala L, Perez R, Weinstein A, Zdanowicz J,
119. Orlando C, Sestini R, Vona G, et al: Detection of Creaven PJ: Effect of glutathione depletion on the
c-erbB-2 amplification in transitional cell bladder cytotoxicity of cisplatin and iproplatin in a human
354 Part IV Bladder

melanoma cell line. Cancer Chemother Pharmacol 1997; 151. Parekh DJ, Gilbert WB, Koch MO, Smith JA Jr:
40:38. Continent urinary reconstruction versus ileal conduit: a
135. Hospers GA, Mulder NH, de Jong B, et al: contemporary single-institution comparison of
Characterization of a human small cell lung carcinoma perioperative morbidity and mortality. Urology 2000;
cell line with acquired resistance to cis- 55:852.
diamminedichloroplatinum(II) in vitro. Cancer Res 1988; 152. Parekh DJ, Clark T, O’Connor J, et al: Orthotopic
48:6803. neobladder following radical cystectomy in patients with
136. Liebmann JE, Hahn SM, Cook JA, et al: Glutathione high perioperative risk and co-morbid medical
depletion by L-buthionine sulfoximine antagonizes Taxol conditions. J Urol 2002; 168:2454.
cytotoxicity. Cancer Res 1993; 53:2066. 153. Bochner BH, Figueroa AJ, Skinner EC, et al: Salvage
137. Miller DC, Taub DA, Dunn RL, Montie JE, Wei JT: radical cystoprostatectomy and orthotopic urinary
The impact of co-morbid disease on cancer control and diversion following radiation failure. J Urol 1998;
survival following radical cystectomy. J Urol 2003; 160:29.
169:105. 154. Hautmann RE, Simon J: Ileal neobladder and local
138. Chang SS, Alberts G, Cookson MS, Smith JA Jr: Radical recurrence of bladder cancer: patterns of failure and
cystectomy is safe in elderly patients at high risk. J Urol impact on function in men. J Urol 1999; 162:1963.
2001; 166:938. 155. Studer UE, Zingg EJ: Ileal orthotopic bladder
139. Stroumbakis N, Herr HW, Cookson MS, Fair WR: substitutes. What we have learned from 12 years’
Radical cystectomy in the octogenarian. J Urol 1997; experience with 200 patients. Urol Clin North Am 1997;
158:2113. 24:781.
140. Soulie M, Straub M, Game X, et al: A multicenter study 156. Gburek BM, Lieber MM, Blute ML: Comparison of
of the morbidity of radical cystectomy in select elderly Studer ileal neobladder and ileal conduit urinary
patients with bladder cancer. J Urol 2002; 167:1325. diversion with respect to perioperative outcome and late
141. Skinner DG, Stein JP, Lieskovsky G, et al: 25-year complications. J Urol 1998; 160:721.
experience in the management of invasive bladder cancer 157. Parekh DJ, Gilbert WB, Smith JA Jr: Functional lower
by radical cystectomy. Eur Urol 1998; 33(Suppl 4):25. urinary tract voiding outcomes after cystectomy and
142. Nixon RG, Chang SS, Lafleur BJ, Smith JJ, Cookson orthotopic neobladder. J Urol 2000; 163:56.
MS: Carcinoma in situ and tumor multifocality predict 158. Hautmann RE, de Petriconi R, Gottfried HW, et al: The
the risk of prostatic urethral involvement at radical ileal neobladder: complications and functional results in
cystectomy in men with transitional cell carcinoma of the 363 patients after 11 years of followup. J Urol 1999;
bladder. J Urol 2002; 167:502. 161:422.
143. Wood DP Jr, Montie JE, Pontes JE, VanderBrug 159. Madersbacher S, Mohrle K, Burkhard F, Studer UE:
Medendorp S, Levin HS: Transitional cell carcinoma of Long-term voiding pattern of patients with ileal
the prostate in cystoprostatectomy specimens removed orthotopic bladder substitutes. J Urol 2002;
for bladder cancer. J Urol 1989; 141:346. 167:2052.
144. Vallancien G, Abou El Fettouh H, Cathelineau X, et al: 160. Stein JP, Lieskovsky G, Ginsberg DA, Bochner BH,
Cystectomy with prostate sparing for bladder cancer Skinner DG: The T pouch: an orthotopic ileal
in 100 patients: 10-year experience. J Urol 2002; neobladder incorporating a serosal lined ileal antireflux
168:2413. technique. J Urol 1998; 159:1836.
145. Herr HW: Extent of surgery and pathology evaluation 161. Dutta SC, Chang SC, Coffey CS, et al: Health related
has an impact on bladder cancer outcomes after radical quality of life assessment after radical cystectomy:
cystectomy. Urology 2003; 61:105. comparison of ileal conduit with continent orthotopic
146. Marshall FF, Mostwin JL, Radebaugh LC, Walsh PC, neobladder. J Urol 2002; 168:164.
Brendler CB: Ileocolic neobladder post-cystectomy: 162. Stein JP, Esrig D, Freeman JA, et al: Prospective
continence and potency. J Urol 1991; 145:502. pathologic analysis of female cystectomy specimens: risk
147. Pritchett TR, Schiff WM, Klatt E, Lieskovsky G, factors for orthotopic diversion in women. Urology
Skinner DG: The potency-sparing radical cystectomy: 1998; 51:951.
does it compromise the completeness of the cancer 163. Chang SS, Cole E, Cookson MS, Peterson M, Smith JA
resection? J Urol 1988; 140:1400. Jr: Preservation of the anterior vaginal wall during
148. Brendler CB, Steinberg GD, Marshall FF, Mostwin JL, female radical cystectomy with orthotopic urinary
Walsh PC: Local recurrence and survival following diversion: technique and results. J Urol 2002; 168:1442.
nerve-sparing radical cystoprostatectomy. J Urol 1990; 164. Ali-el-Dein B, el-Sobky E, Hohenfellner M, Ghoneim
144:1137. MA: Orthotopic bladder substitution in women:
149. Schoenberg MP, Walsh PC, Breazeale DR, et al: Local functional evaluation. J Urol 1999; 161:1875.
recurrence and survival following nerve sparing radical 165. Blute ML, Gburek BM: Continent orthotopic urinary
cystoprostatectomy for bladder cancer: 10-year followup. diversion in female patients: early Mayo Clinic
J Urol 1996; 155:490. experience. Mayo Clin Proc 1998; 73:501.
150. Hautmann RE: 15 years experience with the ileal 166. Mills RD, Studer UE: Female orthotopic bladder
neobladder. What have we learned? Urologe A 2001; substitution: a good operation in the right circumstances.
40:360. J Urol 2000; 163:1501.
Chapter 19 Prognosis and Management of Invasive Transitional Cell Carcinoma 355

167. Ghoneim MA: Orthotopic bladder substitution in retrospective review of 675 cases in 2 decades. J Urol
women following cystectomy for bladder cancer. Urol 1992; 148:1401.
Clin North Am 1997; 24:225. 184. Chang SS, Baumgartner RG, Wells N, Cookson MS,
168. Chang SS, Cole E, Smith JA Jr, Cookson MS: Smith JA Jr: Causes of increased hospital stay after
Pathological findings of gynecologic organs obtained at radical cystectomy in a clinical pathway setting. J Urol
female radical cystectomy. J Urol 2002; 168:147. 2002; 167:208.
169. Stenzl A, Draxl H, Posch B, et al: The risk of urethral 185. Chang SS, Smith JA Jr, Wells N, et al: Estimated blood
tumors in female bladder cancer: can the urethra be used loss and transfusion requirements of radical cystectomy.
for orthotopic reconstruction of the lower urinary tract? J Urol 2001; 166:2151.
J Urol 1995; 153:950. 186. Sternberg CN, Calabro F: Neo-adjuvant chemotherapy
170. Poulsen AL, Horn T, Steven K: Radical cystectomy: in invasive bladder cancer. World J Urol 2001; 19:94.
extending the limits of pelvic lymph node dissection 187. Bajorin DF: Plenary debate of randomized phase III trial
improves survival for patients with bladder cancer of neoadjuvant MVAC plus cystectomy versus
confined to the bladder wall. J Urol 1998; 160:2015. cystectomy alone in patients with locally advanced
171. Leissner J, Hohenfellner R, Thuroff JW, Wolf HK: bladder cancer. J Clin Oncol 2001; 19:17S.
Lymphadenectomy in patients with transitional cell 188. Sternberg CN: Current perspectives in muscle invasive
carcinoma of the urinary bladder; significance for staging bladder cancer. Eur J Cancer 2002; 38:460.
and prognosis. BJU Int 2000; 85:817. 189. von der Maase H, Hansen SW, Roberts JT, et al:
172. Konety BR, Joslyn SA, O’Donnell MA: Extent of pelvic Gemcitabine and cisplatin versus methotrexate,
lymphadenectomy and its impact on outcome in patients vinblastine, doxorubicin, and cisplatin in advanced or
diagnosed with bladder cancer: analysis of data from the metastatic bladder cancer: results of a large, randomized,
surveillance, epidemiology and end results program data multinational, multicenter, phase III study. J Clin Oncol
base. J Urol 2003; 169:946. 2000; 18:3068.
173. Herr HW, Bochner BH, Dalbagni G, et al: Impact of the 190. Donat SM, Herr HW, Bajorin DF, et al: Methotrexate,
number of lymph nodes retrieved on outcome in patients vinblastine, doxorubicin and cisplatin chemotherapy and
with muscle invasive bladder cancer. J Urol 2002; cystectomy for unresectable bladder cancer. J Urol 1996;
167:1295. 156:368.
174. Stein JP: Editorial: contemporary concepts of radical 191. Hall R, Updated results of a randomised controlled trial
cystectomy and the treatment of bladder cancer. J Urol of neoadjuvant cisplatin©, methotrexate (M), and
2003; 169:116. vinblastine (V) chemotherapy for muscle-invasive
175. Wishnow KI, Levinson AK, Johnson DE, et al: Stage B bladder cancer (Abstract 710). Proc Am Soc Clin Oncol
(P2/3A/N0) transitional cell carcinoma of bladder highly 2002; 21:178A.
curable by radical cystectomy. Urology 1992; 39:12. 192. Natale RB, Grossman HB, Blumenstein B, et al:
176. Ghoneim MA, el-Mekresh MM, el-Baz MA, el-Attar IA, SWOG 8710 (INT-0080): randomized phase III trial
Ashamallah A: Radical cystectomy for carcinoma of the of neoadjuvant MVAC + cystectomy versus
bladder: critical evaluation of the results in 1026 cases. cystectomy alone in patients with locally advanced
J Urol 1997; 158:393. bladder cancer (Abstract 3). Proc Am Soc Clin Oncol
177. Gschwend JE, Dahm P, Fair WR: Disease specific survival 2001; 20:21.
as endpoint of outcome for bladder cancer patients 193. Shipley WU, Winter KA, Kaufman DS, et al: Phase III
following radical cystectomy. Eur Urol 2002; 41:440. trial of neoadjuvant chemotherapy in patients
178. Dalbagni G, Genega E, Hashibe M, et al: Cystectomy with invasive bladder cancer treated with selective
for bladder cancer: a contemporary series. J Urol 2001; bladder preservation by combined radiation therapy
165:1111. and chemotherapy: initial results of Radiation
179. Frazier HA, Robertson JE, Dodge RK, Paulson DF: The Therapy Oncology Group 89-03. J Clin Oncol 1998;
value of pathologic factors in predicting cancer-specific 16:3576.
survival among patients treated with radical cystectomy 194. Wallace DM, Raghavan D, Kelly KA, et al: Neo-
for transitional cell carcinoma of the bladder and adjuvant (pre-emptive) cisplatin therapy in invasive
prostate. Cancer 1993; 71:3993. transitional cell carcinoma of the bladder. Br J Urol
180. Pagano F, Bassi P, Galetti TP, et al: Results of 1991; 67:608.
contemporary radical cystectomy for invasive bladder 195. Bassi P, Pappagallo G, Cosciani S, et al: Neoadjuvant
cancer: a clinicopathological study with an emphasis on M-VAC of invasive bladder cancer: G. U. O. N. E.
the inadequacy of the tumor, nodes and metastases multicenter phase III trial. Eur Urol 1998; 33:142.
classification. J Urol 1991; 145:45. 196. Dodd PM, McCaffrey JA, Herr H, et al: Outcome of
181. Montie JE, Wood DP Jr: The risk of radical cystectomy. postchemotherapy surgery after treatment with
Br J Urol 1989; 63:483. methotrexate, vinblastine, doxorubicin, and cisplatin in
182. Rosario DJ, Becker M, Anderson JB: The changing patients with unresectable or metastatic transitional cell
pattern of mortality and morbidity from radical carcinoma. J Clin Oncol 1999; 17:2546.
cystectomy. BJU Int 2000; 85:427. 197. Herr HW, Donat SM, Bajorin DF: Post-chemotherapy
183. Frazier HA, Robertson JE, Paulson DF: Complications surgery in patients with unresectable or regionally
of radical cystectomy and urinary diversion: a metastatic bladder cancer. J Urol 2001; 165:811.
356 Part IV Bladder

198. Bono AV, Benvenuti C, Reali L, et al: Adjuvant 215. Jenkins BJ, Caulfield MJ, Fowler CG, et al: Reappraisal
chemotherapy in advanced bladder cancer: Italian of the role of radical radiotherapy and salvage
Uro-Oncologic Cooperative Group. Prog Clin Biol Res cystectomy in the treatment of invasive (T2/T3) bladder
1989; 303:533. cancer. Br J Urol 1988; 62:343.
199. Studer UE, Bacchi M, Biedermann C, et al: Adjuvant 216. Mameghan H, Fisher RJ, Watt WH, et al: The
cisplatin chemotherapy following cystectomy for bladder management of invasive transitional cell carcinoma of
cancer: results of a prospective randomized trial. J Urol the bladder. Results of definitive and preoperative
1994; 152:81. radiation therapy in 390 patients treated at the Prince of
200. Stockle M, Wellek S, Meyenburg W, et al: Radical Wales Hospital, Sydney, Australia. Cancer 1992;
cystectomy with or without adjuvant polychemotherapy 69:2771.
for non-organ-confined transitional cell carcinoma of the 217. Fossa SD, Waehre H, Aass N, et al:: Bladder cancer
urinary bladder: prognostic impact of lymph node definitive radiation therapy of muscle-invasive bladder
involvement. Urology 1996; 48:868. cancer. A retrospective analysis of 317 patients. Cancer
201. Chang SS, Hassan JM, Cookson M, Wells N, Smith J: 1993; 72:3036.
Delaying radical cystectomy for muscle invasive bladder 218. Hayter CR, Paszat LF, Groome PA, et al: A population-
cancer results in worse pathologic stage. J Urol 2003; based study of the use and outcome of radical
170:1085. radiotherapy for invasive bladder cancer. Int J Radiat
202. Gschwend JE, Fair WR, Vieweg J: Radical cystectomy Oncol Biol Phys 1999; 45:1239.
for invasive bladder cancer: contemporary results and 219. Pollack A, Zagars GK, Swanson DA: Muscle-invasive
remaining controversies. Eur Urol 2000; 38:121. bladder cancer treated with external beam radiotherapy:
203. Hautmann RE: Complications and results after prognostic factors. Int J Radiat Oncol Biol Phys 1994;
cystectomy in male and female patients with locally 30:267.
invasive bladder cancer. Eur Urol 1998; 33(Suppl 4):23. 220. Gospodarowicz MK, Hawkins NV, Rawlings GA, et al:
204. Abratt RP, Wilson JA, Pontin AR, Barnes RD: Salvage Radical radiotherapy for muscle invasive transitional cell
cystectomy after radical irradiation for bladder cancer- carcinoma of the bladder: failure analysis. J Urol 1989;
prognostic factors and complications. Br J Urol 1993; 142:1448.
72:756. 221. Duncan W, Quilty PM: The results of a series of 963
205. Barnes RW, Bergman RT, Hadley HT, et al: Survival patients with transitional cell carcinoma of the urinary
following transurethral resection of bladder carcinoma. bladder primarily treated by radical megavoltage x-ray
Cancer Res 1977; 37:2895. therapy. Radiother Oncol 1986; 7:299.
206. Herr H: Conservative management of muscle-infiltrating 222. Feneley MR, Schoenberg MP: Bladder-sparing strategies
bladder cancer: prospective experience. J Urol 1987; for transitional cell carcinoma. Urology 2000; 56:549.
138:1162. 223. Kuczyk M, Machtens S, Bokemeyer C, et al: Surgical
207. Solsona E, Iborra I, Ricos JV, et al: Feasibility of bladder preserving strategies in the treatment of
transurethral resection for muscle infiltrating carcinoma muscle-invasive bladder cancer. World J Urol 2002;
of the bladder: long-term followup of a prospective 20:183.
study. J Urol 1998; 159:95. 224. Kaufman DS, Shipley WU, Griffin PP, et al: Selective
208. Henry K, Miller J, Mori M, et al: Comparison of bladder preservation by combination treatment of
transurethral resection to radical therapies for stage B invasive bladder cancer. N Engl J Med 1993; 329:1377.
bladder tumours. J Urol 1988; 140:964. 225. Shipley WU, Kaufman DS, Heney NM, Althausen AF,
209. Malkowicz SB, Nichols P, Lieskovsky G, et al: The Zietman AL: An update of selective bladder preservation
role of radical cystectomy in the management of by combined modality therapy for invasive bladder
high-grade superficial bladder cancer. J Urol 1990; cancer. Eur Urol 1998; 33(Suppl 4):32.
44:641. 226. Dunst J, Rodel C, Zietman A, et al: Bladder preservation
210. Whitmore WF Jr, Batata MA, Ghoneim MA, Grabstald H, in muscle-invasive bladder cancer by conservative
Unal A: Radical cystectomy with or without prior surgery and radiochemotherapy. Semin Surg Oncol
irradiation in the treatment of bladder cancer. Trans Am 2001; 20:24.
Assoc Genitourin Surg 1977; 69:100. 227. Zietman AL, Shipley WU, Kaufman DS:
211. Novick AC, Stewart BH: Partial cystectomy in the Organ-conserving approaches to muscle-invasive bladder
treatment of primary and secondary carcinoma of the cancer: future alternatives to radical cystectomy. Ann
bladder. J Urol 1976; 116:570. Med 2000; 32:34.
212. Dandekar NP, Tongaonkar HB, Dalal AV, Kulkarni JN, 228. Bradley BA, Wajsman Z: The role of chemotherapy
Kamat MR: Partial cystectomy for invasive bladder and radiation in organ-preservation strategies for
cancer. J Surg Oncol 1995; 60:24. muscle-invasive bladder cancer. World J Urol 2002;
213. Schoborg TW, Sapolsky JL, Lewis CW Jr: Carcinoma of 20:167.
the bladder treated by segmental resection. J Urol 1979; 229. Zietman AL, Grocela J, Zehr E, et al: Selective bladder
122:473. conservation using transurethral resection,
214. Resnick MI, O’Connor VJ Jr: Segmental resection for chemotherapy, and radiation: management and
carcinoma of the bladder: review of 102 patients. J Urol consequences of Ta, T1, and Tis recurrence within the
1973; 109:1007. retained bladder. Urology 2001; 58:380.
Chapter 19 Prognosis and Management of Invasive Transitional Cell Carcinoma 357

230. Given RW, Parsons JT, McCarley D, Wajsman Z: 235. Hardt J, Filipas D, Hohenfellner R, Egle UT: Quality
Bladder-sparing multimodality treatment of of life in patients with bladder carcinoma after
muscle-invasive bladder cancer: a five-year follow-up. cystectomy: first results of a prospective study. Qual
Urology 1995; 46:499. Life Res 2000; 9:1.
231. Caffo O, Fellin G, Graffer U, Luciani L: Assessment of 236. Hart S, Skinner EC, Meyerowitz BE, et al: Quality of
quality of life after cystectomy or conservative therapy life after radical cystectomy for bladder cancer in
for patients with infiltrating bladder carcinoma. A survey patients with an ileal conduit, cutaneous or urethral
by a self-administered questionnaire. Cancer 1996; Kock pouch. J Urol 1999; 162:77.
78:1089. 237. Kitamura H, Miyao N, Yanase M, et al: Quality of life in
232. Zietman AL, Shipley WU, Kaufman DS, et al: A phase patients having an ileal conduit, continent reservoir or
I/II trial of transurethral surgery combined with orthotopic neobladder after cystectomy for bladder
concurrent cisplatin, 5-Fluorouracil and twice daily carcinoma. Int J Urol 1999; 6:393.
radiation followed by selective bladder preservation in 238. Skinner EC: Quality of life with reconstruction. Semin
operable patients with muscle invading bladder cancer. Urol Oncol 2001; 19:56.
J Urol 1998; 160:1673. 239. Hobisch A, Tosun K, Kinzl J, et al: Life after cystectomy
233. Mansson A, Mansson W: When the bladder is gone: and orthotopic neobladder versus ileal conduit urinary
quality of life following different types of urinary diversion. Semin Urol Oncol 2001; 19:18.
diversion. World J Urol 1999; 17:211. 240. van der Veen JH, van Andel G, Kurth KH: Quality-of-life
234. Lynch WJ, Jenkins BJ, Fowler CG, Hope-Stone HF, assessment in bladder cancer. World J Urol 1999; 17:219.
Blandy JP: The quality of life after radical radiotherapy
for bladder cancer. Br J Urol 1992; 70:519.
C H A P T E R

20 Transurethral Surgery of Bladder


Tumors
Rabi Tiguert, M.D., Louis Lacombe, M.D., F.R.C.S.C.,
and Yves Fradet, M.D.

Cancers originating from the urothelium are responsible Laser therapy, either as thermocoagulation of bladder
for 8% of all diagnosed tumors in men and 4% in women. tumors or as photodynamic therapy, is an alternative that
In the United States alone, this corresponds to more than has become part of the treatment armamentarium. Recent
56,000 new cases yearly,1 transitional cell carcinoma advances in fluorescent cystoscopy may influence visualiza-
(TCC) accounting for approximately 90% of all cases.2 At tion of tumors and enhance the completeness of resection.
the time of diagnosis, about 80% of bladder tumors are This chapter will review the indications, techniques, and
superficial and limited to the mucosa or submucosa (Ta, outcomes of TUR, LASER thermocoagulation, and pho-
T1), while 20% invade the muscular layer of the bladder todynamic therapy in the management of bladder cancer.
wall. In recent cohort studies of patients newly diagnosed
with Ta, T1 tumors, a 55% recurrence rate was observed
TRANSURETHRAL RESECTION
within 3 years,3,4 but previous studies had reported up to
Preparation
70% recurrence within 5 years of the initial diagnosis.5
The majority of these recurrent tumors will remain super- Before TUR of the first tumor, it is mandatory to study
ficial and progression to muscle invasive or metastatic can- the upper urinary tract either by retrograde pyelogram or
cer will occur in <10% of recurrent cases. These statistics intravenous pyelogram (IVP) to rule out any associated
underscore the overall importance of transurethral surgery tumor of the renal pelvis and/or ureters and to identify
as the primary and often only treatment modality for blad- any hydroureteronephrosis, which is most often the
der cancer. Obviously, intravesical immunotherapy and result of tumor invasiveness. The IVP is usually normal
chemotherapy have become important therapeutic modal- in the presence of superficial bladder tumor with the
ities to prevent recurrence and progression of superficial exception of tumors directly overlying the ureteral orifice
bladder tumors. Nevertheless, the quality of the and causing obstruction. About 3% of patients with blad-
transurethral surgery has an important influence on the der cancer will concurrently or subsequently have an
risk of tumor recurrence, since up to 41% of recurrent upper tract urothelial neoplasm. The presence of ureteral
tumors were reported to occur at the site of the original obstruction, particularly in the absence of intravesical
resection in the National Bladder Cancer Collaborative involvement of the ureteral orifices, suggests a high-
Group study.6 Recent EORTC studies on variability of stage, muscle-invasive, and metastatic tumor in more
recurrence were linked to the experience of the surgeon. than 90% of the patients. If parameters of the preopera-
Endoscopic resection or fulguration of bladder tumor is tive work-up are suspicious for advanced stage disease or
usually effective, has low morbidity, and is associated with a muscle invasive disease, then computed tomography
rapid postoperative recovery. Transurethral resection (CT) scan of the abdomen and pelvis with injection may
(TUR) is the standard method for the initial diagnosis and be a suitable alternative to IVP if thin cuts through the
staging of bladder cancer, as well as for the eradication of kidney and ureter are obtained. CT scan has the advan-
low-stage bladder tumor. Some infiltrating bladder cancers tage of determining size of the bladder tumor and, to
can also be treated primarily by TUR, with or without some extent, presence of local invasion or regional lym-
adjuvant radiation therapy or systemic chemotherapy. phadenopathy. Ureteral obstruction is a significant risk

358
Chapter 20 Transurethral Surgery of Bladder Tumors 359

factor of poor prognosis and has been associated with sig- bladder cancer. It is performed with the patient under
nificantly lower, overall, and stage-specific, survival rates, anesthesia before tumor resection and should be repeated
despite radical surgery. More than 90% of patients with after the endoscopic procedure is completed. In the male
bilateral obstruction had disease with extravesical exten- patient, the urologist puts a finger in the rectum with the
sion. One-third of patients with unilateral hydronephro- other hand placed suprapubically and palpates for any
sis had disease confined to the bladder, of whom 42% had mass (Figure 20-1). In the female patient, bimanual exam-
tumor confined to the mucosa with no muscle invasion ination is performed with two fingers in the vagina and
(p0-p1). These findings suggest that a significant propor- the other hand placed suprapubically. The presence of a
tion of patients with unilateral obstruction may have mass should be noted, along with the evidence of indura-
good outcome with cystectomy alone.7 tions or fixation to the adjacent organs or the pelvis walls.
The disappearance of a movable mass after TUR is more
likely consistent with a superficial tumor. When the mass
Anesthesia
is fixed or indurated or persists after TUR, it is more
Following the diagnosis of a bladder tumor, which is usu- likely to be muscle-invasive tumor. Bimanual examination
ally made in an office setting, the formal endoscopic eval- cannot distinguish benign from malignant bladder fixa-
uation, including biopsy-resection, is performed in the tion in patients who underwent previous radiation ther-
operating room and requires that the patient have an apy or had pelvic inflammatory disease.
adequate anesthesia for the pelvic floor and bladder
relaxation and thus a prior consultation with the anesthe-
Instrumentation and Equipment
siologist is required to provide a successful outcome.
Anesthesia could be attempted either with spinal or gen- Several instruments are potentially useful for the surgical
eral anesthesia. Other methods of anesthesia have been removal of bladder tumors. At the time of the operative
reported for removal of small tumors or performing procedure, the urologist should have sufficient lenses,
cold-cup biopsies, including local anesthesia with intrav- which consist of either a 12-degree or 30-degree lens, and
esical instillations of lignocaine or with electromotive a 70-degree lens. On the other hand, several loops may be
drug administration system (EMDA) that enhances blad- available, including the 90-degree loop, which is usually
der wall penetration of lidocaine. This approach has not used for resecting a tumor along the floor of the bladder,
been widely accepted.8,9 the trigone, or near the bladder neck. The angled loop is
preferable to resect tumors located along the lateral walls,
posterior wall, or dome. The roller or ball electrode is
Bimanual Examination
helpful to achieve hemostasis at the end of tumor resec-
The bimanual examination is an integral part of the oper- tion and to cauterize small noninvasive papillary tumors
ative procedure for patients undergoing treatment for or areas of diffuse carcinoma in situ (CIS).

Figure 20-1 Bimanual examination.


360 Part IV Bladder

Video Endoscopy
the bladder wall.10 Some cup biopsy forceps have an
The new generation of endoscopic video equipment is attachment for electrocautery that allows immediate
very attractive and its use is quickly becoming routine in coagulation of the bladder wall. A Bugbee electrode or
private and teaching hospitals as instruction during endo- resectoscope or roller may be used to cauterize bleeding
scopic procedures takes on a new dimension. The advan- vessels.11 Suspicious erythematous areas for CIS can be
tage of the endoscopic video system is that all of the staff sampled by cold biopsy. There is almost no risk of blad-
in the operating room feels that they are taking part in der perforation, and the absence of electrocautery during
the procedure, which no longer isolates the urologist removal helps preserve the architecture of the urothe-
from the other members of the team. The likelihood of lium for pathology evaluation. The role of a cold-cup
contamination of body fluids is negligible with endo- biopsy in normal appearing areas of the bladder is con-
scopic video procedures. troversial.

Flexible Cystoscope Technique for Transurethral Resection


Flexible cystoscope is often used for the initial diagnosis The first step of TUR is to visualize the whole bladder
and follow-up of patients with bladder tumors in the mucosa, the trigone, ureteral orifices, bladder neck, and
office setting. It has less discomfort than rigid endoscopy prostatic urethra (Figure 20-2). Sometimes it is difficult
especially in men. The entire urinary bladder mucosa can to visualize the dome especially in obese patient. In such
be examined with a single instrument, as well as bladder circumstances, suprapubic manual pressure toward the
neck, by retrograde view. Small laser fibers or Bugbee resectoscope should be applied, the mucosa in the begin-
electrode devices can be inserted through a flexible ning of bladder filling is to be observed. The second step
cystoscope to allow destruction of small, low-grade, non- includes the report of any abnormality, such as erythe-
invasive papillary tumors in the office setting. matous areas, that could correspond to CIS or dysplasia.
In our institution, we used a diagram to report the loca-
tion, appearance (papillary or sessile), and size of all
Cold-Cup Biopsy
tumors, which is a part of the patient’s record. The sites
Cold-cup biopsy is used primarily to remove papillary of mucosal biopsies are also recorded on the diagram, as
tumors or to sample multiple areas of the bladder well as all the observed abnormalities during cystoscopy
mucosa. The small size of the cup limits its indication to were also noted. Photographs of initial tumors and
tumors generally <0.5 cm. Biopsy forceps exist for flexi- abnormal areas can be taken in order to be compared
ble cystoscope but most often a rigid cystoscope is used. with the recurrent one. Following endoscopy, a resecto-
The cup is closed over a papillary tumor, and with a scope is used to remove all visible tumors. A continuous
twisting and pulling motion, the tumor is avulsed from flow resectoscope has the advantage to maintain the

Figure 20-2 Transurethral resection of a bladder tumor.


Chapter 20 Transurethral Surgery of Bladder Tumors 361

bladder at a reduced capacity and thus minimize the risk ent techniques, a better exposure of these tumors could
of bladder perforation or stimulation of the obturator be obtained. By pushing down the anterior lower wall of
nerve. A 70-degree lens is used prior and after tumor the abdomen with the opposite hand, these tumors are
resection to review the bladder, since there are locations usually brought to a better view. An assistant can carry
in the bladder that are difficult to visualize with the out this manipulation if the surgeon prefers the two-
12-degree or 30-degree lens used with a resectoscope. After handed instrument. However, even with the use of a
the resectoscope has been inserted, the resecting loop long resectoscope, such tumors may remain inaccessible
should be readily visible and the instrument should be in a morbidly obese patient. In this uncommon setting,
checked for full excursion of the loop. The resecting loop the resectoscope can be inserted by a perineal urethros-
should be passed beyond the tumor and then pulled back tomy performed in the bulbar urethra over a metal
toward the viewing eye while applying the electrocautery. sound to circumvent the suspensory ligament of the
At some point, it is necessary to maintain the resecto- penis. After the TUR a Foley catheter is inserted into
scope loop in an extended fixed position and to move the the penile urethra or through the perineal urethrostomy,
resectoscope downward with a rocking motion parallel to which heals within 24 hours of removal of the catheter.
the base of the tumor, similar to scraping a wall. Constant Another method to remove these anterior bladder wall
orientation is necessary during these maneuvers. With tumors has been described, which consists to perform a
papillary tumors, the resecting loop is often used to ele- 2-cm incision in the skin that is deepened by blunt dis-
vate the tumor off the bladder wall before application of section to the fascial level. A nasal speculum is used to
the electric current. The base of bladder tumors with refract the fat and allow an incision to be made on the
small pedicle can often be resected in a whole piece. In fascia. A finger is inserted to localize the bowel. A peanut
larger tumors, however, early attempt to resect its base or empty sponge stick holder is introduced through the
will result in a nonfixed tumor rendering the resection opening and the TUR began with control of the tumor
difficult. Resection of bladder tumors should ideally be position. This technique has been used in 9 patients by
performed in two steps: resection of the tumor and its the author who reported this technique. It is mandatory
base, which must include muscle wall. Care should be to carry out the TUR with usual precautions to avoid
taken to limit coagulation artifact on the specimen for a bladder perforation.13
better pathologic analysis. In patients with possible mus- Deep resection in the bladder neck area must be care-
cle invasive tumors or suspicion of CIS, a TUR of the fully performed. In females, TUR in such areas can cre-
prostatic urethra should be performed to rule out CIS ate a vesicovaginal fistula. In men, it may result in
or intraductal neoplasia in the prostate. Such information trigonal undermining hampering further resection and
is important for counseling the patient in planning even bladder catheterization. If an undermining trigone
intravesical therapy, or cystectomy with orthotopic is apparent at the end of surgery, the catheter should be
neobladder. Prostatic urethral sampling could be inserted with the help of a catheter guide.
attempted either with superficial resection immediately Tumors involving the ureteral orifice occur in approx-
proximal to the verumontanum on both sides at 5 and imately 10% to 15% of cases. Noninvasive tumors should
7 o’clock, or by resection of the whole prostatic urethra be resected aggressively and resection may be pursued to
for a complete sampling if cystectomy with prostate spar- remove the whole intramural ureter if a papillary tumor
ing is indicated.12 is found to extend in this area. Complete resection of a
noninvasive papillary tumor will leave a bulging ureteral
mucosa. To prevent stenosis of the orifice, fulguration
Irrigation Fluid
and coagulation should be minimized in this area. If
The choice of irrigation fluid is important during a TUR extensive coagulation is required or the ureteral mucosa
of a bladder tumor, because of problems with electrical does not appear to protrude satisfactorily, a ureteral
conduction, saline solution should not be used. The catheter should be placed for a few days to allow initial
amino acid solutions used for TUR of the prostate are an healing and prevent ureteral obstruction. Resection of a
option but are expensive and unnecessary, since the ureteral orifice and the intramural portion of a ureter will
absorption of large amounts of irrigation fluids is rarely result in vesicoureteral reflux. While reflux may increase
encountered with bladder tumor resection. Sterile water the risk of upper urinary tract seeding, it also allows pas-
or isotonic irrigation fluid are mostly used. sage of endovesical immunotherapy or chemotherapy
and ensuing preventive effect.
TUR of tumors arising in an acquired bladder diver-
Management of Intraoperative Problems
ticulum is limited, since the absence of an underlying
Some tumors may be difficult to resect because of their muscular component will result in a perforation. Only
location, such as anterior bladder wall tumors, especially small superficial tumors should be resected or fulgurated
those located close to the bladder neck. By using differ- in a diverticulum with a wide mouth. Otherwise, we can
362 Part IV Bladder

rely on laser therapy or intravesical immunotherapy or cer, radical TUR is an acceptable bladder-sparing treat-
chemotherapy. If there is a doubt or a persistent tumor, a ment strategy. TUR alone is justified only when the blad-
diverticulectomy by an extravesical approach should be der tumor is clinically limited to the superficial muscle
performed after proper control of any present tumor. layer and when re-resection biopsies of the periphery and
The goal of TUR treatment for superficial bladder deep muscle in the tumor bed are negative for invasive
tumors should be complete eradication of the tumors. If carcinoma. Patients who have any evidence of residual
there are numerous tumors or bad visibility hampered by tumor invasion are more likely to undergo incomplete
uncontrolled bleeding, surgery should be aborted and resection and be understaged, which predisposes them to
resumed 1 or 2 weeks after. Laser therapy and endovesi- early local relapse or metastatic disease and higher risk of
cal therapy may be an option for residual superficial blad- developing new invasive cancers in the bladder. Lifelong
der tumors. surveillance cystoscopy seems necessary to detect recur-
Sometimes, concurrent benign prostatic hyperplasia rent invasive tumors that can be successfully treated with
may be too large or protruding into the bladder and thus salvage cystectomy. Even with close follow-up, patients
impeding TUR of bladder tumors. Ideally, the bladder who elect treatment by TUR alone assume some risk (at
tumor should be resected first and allow the healing least 8% and possibly as high as 16%) that a recurrent
before the prostate is resected in order to prevent tumor invasive tumor in the retained bladder may eventually
seeding in the prostatic fossa. In rare instances, the resec- cause death from TCC.
tion of the prostatic lobe is performed prior to the blad-
der tumor. There is a lack of increase of transitional cell
Postoperative Management
tumors seeding of the prostatic fossa in previous reports
when concomitant TUR of bladder tumor and prostate Resection of smaller bladder tumors with very minimal
were performed. Mitomycin C has been shown to bleeding may not require postoperative catheterization
decrease the likelihood of bladder tumors seeding and it unless postoperative administration of mitomycin C is
may be safe to administer a single instillation of 40 mg indicated. Furthermore, most endoscopic resections
after the surgery. require a short period of bladder catheterization with a
Foley catheter. The catheter can be removed when the
hematuria has resolved and this is routinely observed on
Second Look Resection
postoperative day one. Catheterization helps quantify both
On occasion, the pathologist will have difficulty deter- urine output and degree of hematuria. With a catheter in
mining whether the tumor has invaded the muscularis place, the bladder does not have to contract to empty, and
propria. As this is important to dictate therapy, these this may beneficially reduce the amount of postoperative
patients need to undergo a re-resection of the base of the bleeding. On the other hand, bladder spasms may occur
tumor. In some instances, a second look resection is per- with the Foley catheter in place responding to anticholin-
formed to resect any residual tumor left behind in 37% ergic therapy. Bleeding should be controlled at the time of
of cases.13 This maneuver will decrease the rate of blad- surgery. When bleeding control is adequate, there is gen-
der tumor recurrences and increase the number of blad- erally no need for three-way bladder irrigation. Catheter
der spared. We perform a second look resection usually drainage may be indicated for longer periods of time if
4 weeks after the initial one in the presence of T1G3 perforation has occurred or if tumor resection extends up
tumor invading more than 50% of the depth of the to the limit of the bladder wall. Antibiotics are not required
muscularis mucosa.14 postoperatively; however, many urologists prescribe prophy-
lactic oral antibiotics for a period of 3 to 5 days.
TUR as Primary Treatment of Muscle Invasive
Tumors Complications
TUR has been mainly reserved to treat superficial blad- The most frequent encountered complications following
der tumors. However, some select patients with muscle TUR of bladder tumors are intra- and immediate post-
invasive bladder cancer could be treated with TUR alone operative bleeding with an incidence ranging from 2% to
or associated with chemotherapy and radiation therapy. 13%. In a recent study, the complication rate was 5.1%
Several studies show that in selected patients, TUR in a series of 2821 patients.18
achieves 5-year survival rates comparable to those
achieved with radical cystectomy,15–17 and approximately
Hemorrhage
10% of cystectomy patients have no residual tumor (p0)
found in the cystectomy specimen.16 This suggests that The incidence of hemorrhage following TUR of bladder
TUR can control some muscle-invasive bladder cancers. tumors ranges between 2% and 13%.18–20 This variability
For patients with primary muscle-invasive bladder can- of incidence is due to the differences of the definition of
Chapter 20 Transurethral Surgery of Bladder Tumors 363

hemorrhage. The best way to prevent this bleeding is to voir site of the germs and during the TUR, the germs are
perform a careful coagulation in the beginning of bladder dislocated and may be the source of infection.21
filling after TUR to avoid the intravesical high pressure However, this observation was not confirmed by others
that stops temporarily the bleeding by compression of who held the endoscopy maneuver responsible for uri-
the vessels. Nevertheless, it is not uncommon to have an nary infection by inoculating germs into the bladder.22,23
important postoperative bleeding in patients with crys-
tal-clear irrigation fluid at the end of surgery. The risk of
Urethral Stricture
hemorrhage exists until the third week postoperative fol-
lowing scare sloughing. These secondary hemorrhages The incidence of urethral stricture was 15% in males and
are seen when the coagulation of the tumor is extensive 4% in females but less than one-third were symptomatic.
leading to a necrosis of the bladder wall. The strictures were found distally in the urethra in 83% of
the patients and could be treated with optical urethrotomy.
The risk factors of urethral stenosis include longer duration
Bladder Perforation
of postoperative catheterization and use of larger size resec-
Bladder perforation was encountered in 0.9% to 5% of toscope. Furthermore, repeated transurethral instrumenta-
patients undergoing TUR of bladder tumors. It is prob- tions may increase the risk of stricture development.24
ably underestimated since it is less reported in the litera-
ture. Many factors have been implicated including deep
Obturator Nerve Reflex
TUR, bad vision of the site to be resected, tumor located
in a diverticulum, stimulation of the obturator nerve by Obturator nerve reflex may be evoked while resecting
an electrical current, and overdistension of the bladder. lesions on the lower lateral wall and neck of the bladder,
Thin bladder wall secondary to multiple TUR of bladder particularly in thin patients. This reflex will result in
tumors is also at risk for perforation. Perforation of the adductor spasms of the leg, as well as inward movement
bladder should be recognized whenever there is difficulty of the bladder wall during resection that could result in
to distend the bladder; the quantity of fluid instilled in its perforation and to some extent in trauma to major
the bladder is greater than the recuperated one and also pelvic vessels. The obturator reflex may be prevented by
in the presence of an abdominal distension associated lowering the cutting current and avoiding distension of
with a tachycardia. When there is a doubt a cystogram the bladder during resection of this area. If these maneu-
under fluoroscopy could be performed during the TUR. vers are not successful, d-tubocurarine and succinyl-
Perforations of the bladder are divided into two groups, choline can be used during general anesthesia, or to block
intraperitoneal and extraperitoneal; the treatment is differ- the obturator nerve as it passes through the obturator
ent in the two categories. Management of extraperitoneal canal with infiltration by a local anesthetic as described
bladder perforation is conservative, consisting of drainage previously by Augspurger.25
of the bladder by a urethral catheter and administration of
large spectrum antibiotics. Surgical or percutaneous
Ureteral Orifice Injury
drainage is rarely indicated and is dictated when there is an
important urine leakage or a pelvic abscess. Intraperitoneal Ureteral orifices could be damaged when the tumor is
bladder perforation is classically managed surgically. The localized inside the orifice or close to it, raising the risk
indication of exploratory surgery relies on the importance of stricture and reflux vesicoureteral. Excretory urogra-
of the perforation, presence of a bowel traumatism, and phy should be performed within a few weeks after a TUR
the risk of infection. In some selected cases, it is possible to that has involved the periureteral area to detect
manage it conservatively in the absence of bowel perfora- hydronephrosis due to ureteral injury. Management of
tion. Draining the bladder with a urethral catheter during ureteral stricture post-TUR consists of antegrade or ret-
a week together with antibiotic administration is sufficient. rograde intraluminal dilatation with a risk of reflux. The
However, if there is no improvement with the conservative risk of tumor seeding is 22-fold greater when there is a
treatment, a surgical or laparoscopic exploration is manda- vesicoureteral reflux. No consensus has been reached to
tory to repair the bladder as a whole, as well as any con- treat the reflux in patients with history of bladder tumors,
comitant bowel perforation. but it seems reasonable to treat patients with reflux grade
III, or having multiple recurrences of bladder tumors or
tumors located near the orifices.26–28
Urinary Infection
The reported incidence of urinary infection following
Reabsorption Syndrome
TUR of bladder tumor (TURBT) ranged from 2% to
39%. The etiology of urinary infection is controversial. It This is a rarely encountered complication when non-
has been suggested that the bladder tumor is the reser- saline irrigating fluid was used. This appears usually fol-
364 Part IV Bladder

lowing intraperitoneal bladder perforation. An extracel- noncontact fiber with a 5-degree to 15-degree angle of
lular hyper hydration was observed leading sometimes to divergence (Table 20-1).
severe hyponatremia causing cerebral edema with nau-
sea, vomiting, and visual disturbances. The treatment
Surgical Techniques
consists of water restriction and diuretics.29
Laser fibers could be inserted through standard cysto-
scopes either flexible or rigid. For rigid cystoscope a
Bladder Explosion
modified Albarran apparatus is available. Energy emis-
Intravesical explosion during endoscopic resection are sion is usually controlled with a foot pedal; 35 to 40 W
rare but may be devastating. The formation of explosive is ample for necrosis. After visualization of the tumor,
gas, essentially an air–hydrogen mixture, results from the laser fiber is introduced into the cystoscope and the
carbonization of tissues (particularly with the coagula- tip is positioned 2 to 3 mm from the surface of the
tion current) and the introduction of air into the blad- tumor. Often, the best way to appreciate the distance
der during manipulation of the resectoscope. The between the fiber tip and the tumor is to start approxi-
nature of the bladder infusion liquid does not appear to mately 1 cm away and slowly move closer to the tumor
play an important role. Prevention implies the use of a until the thermal changes become apparent. The end
coagulation current of moderate power, the avoidance point of treatment is determined by visible changes in
of air entering the bladder accidentally, and the contin- the tumor, which is characterized by a white discol-
uous or repeated evacuation of the air bubble under the oration (Figure 20-3).
bladder vault.30–32
Advantages and Disadvantages of Laser Use
LASER SURGERY
One of the most important advantages of laser treatment
LASER is an acronym that stands for “light amplification is the lack of bleeding during surgery and in the postop-
by the stimulated emission of radiation.” It describes the erative period. Whether energy is applied directly to the
physical process by which light energy is produced. A center of the tumor initially or to the periphery, bleeding
laser is a device that generates an intense beam of light simply does not occur unless there is trauma to the treated
that falls between the longest and the shortest wave- area from the tip of the cystoscope or fiber. Catheter
lengths, in the infrared, visible, and the ultraviolet por- insertion is not required. Patients treated with Nd:YAG
tions of the electromagnetic spectrum. The lasers are laser can immediately resume their daily activity with an
named by the laser medium (solid, liquid dye, or gas extremely low risk of bleeding. Laser treatment appears to
component), which when stimulated produces photons be less painful than electrocautery resection. Patients are
(laser light). During the last two decades, when the FDA able to perceive the laser energy and often describe it as a
approved the use of the Neodymium:YAG (Nd:YAG) burning discomfort. Laser energy does not stimulate the
(1984), Argon (1987), KTP (1988), and in 1992 the obturator nerve. Therefore, contraction of the adductor
Holmium:YAG (Ho:YAG) lasers in urology, these surgi- muscles of the thigh is avoided. This helps to prevent
cal tools have been used to treat various urologic diseases, bladder perforation.
including superficial bladder tumors. The first reported Laser treatment can be effective for tumors located in
study using lasers to treat superficial bladder tumors bladder diverticulum by delivering energy with either a
occurred more than 20 years ago.33, 34 The most of the flexible or rigid cystoscope. Even though the penetration
clinical experience in the treatment of bladder cancer has depth of the laser energy exceeds the thickness of the
been obtained using an Nd:YAG laser with an end-fire diverticulum wall, free perforation rarely occurs.

Table 20-1 Different Laser Types Used in Urology Practice


Laser Type Wavelength (nm) Energy Delivery Endoscopic Uses Tissue Penetration

KTP 532 Continuous wave Yes 1.0 mm

Diode 800–1.1 Continuous wave or pulsed Yes Varies with wavelength

Nd:Yag 1064 Continuous wave Yes 5.0 mm

Ho:YAG 2100 Pulsed Yes <0.5 mm

CO2 10,600 Continuous wave No 0.1 mm


Chapter 20 Transurethral Surgery of Bladder Tumors 365

Figure 20-3 Nd:YAG laser-induced coagulation necrosis of a


bladder tumor. (Courtesy of Dr. Guy Drouin.)

Disadvantages of laser fulguration include lack of


histopathologic specimens for diagnosis. It is recom-
mended to obtain tissue with TUR for proper grading
and staging of the tumor. It requires some training and
some manipulation with safety applications concern for
the patient and the surgeon. The most feared complica-
tion of laser treatment is forward scatter of the energy
with perforation of an adjacent organ even in the absence
of bladder perforation.

Bladder Tumor Recurrence Rate


The clinical results achieved with laser treatment of
superficial bladder cancer are comparable with the Figure 20-4 A, Superficial TCC of the bladder on white light
results of electrocautery resection. In a prospective cystoscopy. B, Same tumors seen by fluorescence
study of 122 patients randomized for Nd:YAG laser (n = cystoscopy.
62) or TUR (n = 60), new tumors in nontreated areas
occurred in 12 patients in the laser group and in 13
patients in the TUR group. Thus, no difference in the increase production of porphyrin is the use of
recurrence rate was observed for laser and electro- 5-aminolevulinic acid (5-ALA), which could be adminis-
cautery treated patients.34 trated intravesically without any major side effects. The
5-ALA is a well-known precursor in the synthesis of pro-
toporphyrin IX, the latter being the immediate precur-
Fluoroscopic Cystoscopy
sor of heme. Accumulation of protoporphyrin IX is
The porphyrins are intermediate in the synthesis of found in urothelial tumor tissue compared with unaf-
heme,35 which all mammalian nucleotide cells have the fected urothelium (ratio 17:1). Thus, tumors fluoresce
ability to synthesize as HAEME-containing enzymes red under excitation with violet light at wavelength of
that are required for energy metabolism. The derivative 375 to 440 nm36 (Figure 20-4). The neoplastic cells can
of hematoporphyrins, like Photofrin, a derivative puri- then be used as an endogenous photosensitizer under
fied form of hematoporphyrin, can be used for photode- excitation with blue light, which produces red fluores-
tection of malignant cells, as well as photodynamic cence. The use of porphyrin fluorescence has been stud-
therapy. However, the latter product needs to be taken ied to improve the diagnosis of superficial bladder
by mouth several days before treatment and one poten- cancer.37–39 Results from these publications have shown
tial side effect of such medication is a skin photosensiti- that fluorescence cystoscopy improved the detection
zation, which can last for 6 to 8 weeks. Another way to rate of all malignant/dysplastic lesions by approximately
366 Part IV Bladder

18% to 76%. In cases of lesion of dysplasia/CIS, detec- 15. Herr HW: Conservative management of muscle-
tion rate was improved from 50% to 140%.37–39 infiltrating bladder cancer: prospective experience. Urol
Furthermore, three recent studies have prospectively 1987; 138:1162–1163.
shown a decrease risk of residual tumor after TUR and 16. Solsona E, Iborra I, Ricos JV, Monros JL, Dumont R:
a significantly improved recurrence-free survival at fol- Feasibility of transurethral resection for muscle-
low-up.40–42 Larger multicenter studies are ongoing to infiltrating carcinoma of the bladder: prospective study.
further evaluate the exact role of fluorescence cys- J Urol 1992; 147:1513–1515.
17. Herr HW: Transurethral resection of muscle-invasive
toscopy in superficial bladder tumors.
bladder cancer: 10-year outcome. J Clin Oncol 2001;
19:89–93.
18. Collado A, Chechile GE, Salvador J, Vicente J: Early
REFERENCES complications of endoscopic treatment for superficial
bladder tumors. J Urol 2000; 164:1529–1532.
1. Jemal A, Thomas A, Murray T, et al: Cancer statistics. 19. Dick A, Barnes R, Hadley H, Bergman RT, Ninan CA:
CA Cancer J Clin 2002; 52:23–47. Complications of transurethral resection of bladder
2. Weiss MA, Mills SE: Atlas of Genitourinary Tract tumors: prevention, recognition and treatment. J Urol
Disorders, pp 12.5–12.10. Singapore, Gower Medical 1980; 124(6):810–811.
Publishing, 1989. 20. Kondas J, Szentgyorgyi E: Transurethral resection of 1250
3. Allard P, Fradet Y, Têtu B, et al: Tumor-associated bladder tumours. Int Urol Nephrol 1992; 24:35–42.
antigens as prognostic factors for recurrence in 382 21. Appell RA, Flynn JT, Paris AM, Blandy JP: Occult
patients with primary transitional cell carcinoma of the bacterial colonization of bladder tumors. J Urol 1980;
bladder. Clin Cancer Res 1995; 1:1195. 124:345–346.
4. Kiemeney LA, Witjes JA, Heijbroek RP, et al: Predictability 22. Goldwasser B, Bogokowsky B, Nativ O, et al: Urinary
of recurrent and progressive disease in individual patients infections following transurethral resection of bladder
with primary superficial bladder cancer. J Urol 1993; 150:60. tumors—rate and source. J Urol 1983; 129:1123–1124.
5. Malmstrom PU, Busch C, Norlen BJ: Recurrence, 23. Badenoch DF, Murdoch DA, Tiptaft RC: Microbiological
progression and survival in bladder cancer: a retrospective study of bladder tumors, their histology and infective
analysis of 232 patients with greater or equal to 5-year complications. Urology 1990; 35:5–8.
follow-up. Scand J Urol Nephrol 1987; 21:185. 24. Nielsen KT, Christensen MM, Olesen S: Urethral
6. National Bladder Cancer Collaborative Group: strictures after transurethral bladder tumor resection.
Surveillance, initial assessment, and subsequent progress Scand J Urol Nephrol 1989; 23(2):81–83.
of patients with superficial bladder cancer in a 25. Augspurger RR, Donohue RE: Prevention of obturator
prospective longitudinal study. Cancer Res 1977; nerve stimulation during transurethral surgery. J Urol
37:2907–2910. 1980; 123:170–172.
7. Haleblian GE, Skinner EC, Dickinson MG, et al: 26. Mukamel E, Nissenkorn I, Glanz I, Vilcovsky E, Servadio
Hydronephrosis as a prognostic indicator in bladder C: Upper tract tumours in patients with vesicoureteral
cancer patients. J Urol 1998; 160:2011–2014. reflux and recurrent bladder tumours. Eur Urol 1985;
8. Holmang S, Aldenborg F, Hedelin H: Extirpation and 11:6–8.
fulguration of multiple superficial bladder tumor 27. De Torres Mateos JA, Banus Gassol JM, Palou Redorta J,
recurrences under intravesical lignocaine anesthesia. Br J Morote Robles J: Vesicorenal reflux and upper urinary
Urol 1994; 73:177–180. tract transitional cell carcinoma after transurethral
9. Jewett MA, Valiquette L, Sampson HA, et al: resection of recurrent superficial bladder carcinoma.
Electromotive drug administration of lidocaine as an J Urol 1987; 138:49–51.
alternative anesthesia for transurethral surgery. J Urol 28. Amar AD, Das S: Upper urinary tract transitional
1999; 161:482–485. cell carcinoma in patients with bladder carcinoma
10. Beaghler M, Grasso M III: Flexible cystoscopic bladder and associated vesicoureteral reflux. J Urol 1985;
biopsies: a technique for outpatient evaluation of the lower 133:468–471.
urinary tract urothelium. Urology 1994; 44:756–759. 29. Hahn RG: Transurethral resection syndrome after
11. Tauber R: Biopsy-coagulation forceps to simplify biopsy transurethral resection of bladder tumours. Can J Anaesth
of the bladder. J Urol 1985; 133:783–785. 1995; 42:69–72.
12. Vallancien G, Abou El Fettouh H, Cathelineau X, et al: 30. Viville C, de Petriconi R, Bietho L: Intravesical explosion
Cystectomy with prostate sparing for bladder cancer in during endoscopic resection. Apropos of a case. J Urol
100 patients: 10-year experience. J Urol 2002; (Paris) 1984; 90:361–363.
168:2413–2417. 31. Hansen RI, Iversen P: Bladder explosion during
13. Horan AH: Urologists at work. Method permitting easier uninterrupted transurethral resection of the prostate.
resection of bladder tumors in the anterior midline. J Urol A case report and an experimental model. Scand J Urol
2000; 164(1):106. Nephrol 1979; 13:211–212.
14. Rigaud J, Karam G, Braud G, et al: T1 bladder tumors: 32. Ning TC Jr, Atkins DM, Murphy RC: Bladder
value of a second endoscopic resection. Prog Urol 2002; explosions during transurethral surgery. J Urol 1975;
12(1):27–30. 114:536–539.
Chapter 20 Transurethral Surgery of Bladder Tumors 367

33. Hofstetter A, Frank F, Keiditsch E, Bowering R: 39. Jichlinski P, Forrer M, Mizeret J, et al: Clinical evaluation
Endoscopic neodymium-YAG laser application of a method for detecting superficial transitional cell
for destroying bladder tumors. Eur Urol 1981; carcinoma of the bladder by light-induced fluorescence of
7:278–282. protoporphyrin IX following topical application of
34. Beisland HO, Seland P: A prospective randomized study 5-aminolevulinic acid: preliminary results. Laser Surg
on neodymium-YAG laser irradiation versus TUR in the Med 1997; 20:402–408.
treatment of urinary bladder cancer. Scand J Urol 40. Kriegmair M, Zaak D, Rothenberger KH, et al:
Nephrol 1986; 20(3):209–212. Transurethral resection for bladder cancer using
35. Moore MR, Disler PB: Biochemical diagnosis of the 5-aminolevulinic acid induced fluorescence endoscopy
porphyrias. Clin Dermatol 1985; 3:24–40. versus white light endoscopy. J Urol 2002; 168:475–478.
36. Koenig F, McGovern FJ: Fluorescence detection of 41. Riedi CR, Daniltchenko D, Koenig F, et al: Fluorescence
bladder carcinoma. Urology 1997; 50:778–779. endoscopy with 5-aminolevulinic acid reduces early
37. Koenig F, McGovern FJ, Larne R, et al: Diagnosis of recurrence rate in superficial bladder cancer. J Urol 2001;
bladder carcinoma using protoporphyrin IX fluorescence 165:1121–1123.
induced by 5-aminolevulinic acid. BJU Int 1999; 42. Filbeck T, Pichhmeier U, Knuechel R, et al: Clinically
83(1):129–135. relevant improvement of recurrence-free survival with
38. Kriegmair M, Baumgartner R, Knuchel R, et al: Detection 5-aminolevulinic acid induced fluorescence diagnosis in
of early bladder cancer by 5-aminolevulinic acid induced patients with superficial bladder tumors. J Urol 2002;
porphyrin fluorescence. J Urol 1996; 155:105–109. 168:67–71.
C H A P T E R

21Partial and Radical Cystectomy


John P. Stein, MD, and Donald G. Skinner, MD

Radical cystectomy has traditionally been considered the A dedicated effort has been made to improve the tech-
standard of therapy for high-grade, invasive bladder can- nique of radical cystectomy and provide an acceptable
cer with the best survival results and lowest local recur- form of urinary diversion without compromise of a sound
rence rates reported to date.1,2 Radical cystectomy cancer operation.8–10 Certain technical issues regarding
provides accurate pathologic staging of the primary blad- the surgical procedure of a radical cystectomy are critical
der tumor and the regional lymph nodes that may then in order to minimize local recurrence and positive surgi-
influence the decision for adjuvant chemotherapy based cal margins, and maximize cancer-specific survival. In
on clear pathologic criteria.3 Furthermore, advances in addition, attention to surgical details is important in opti-
urinary diversion and lower urinary tract reconstruction mizing the successful outcomes of orthotopic diversion:
to the urethra (orthotopic neobladder) have provided maintaining the rhabdosphincter mechanism and urinary
patients a more socially acceptable means to store urine, continence in these patients.10
allowing volitional voiding per urethra without the need
for a urostomy appliance, cutaneous stoma, or the need
PARTIAL CYSTECTOMY
for catheterization in most instances. Currently, most
men and women undergoing radical cystectomy are Although radical cystectomy is the standard therapy for
appropriate candidates for this form of urinary diversion.4 high-grade, invasive bladder cancer, partial cystectomy
Improvements over the past several decades in med- may be an option in a few, very carefully selected indi-
ical, surgical, and anesthetic techniques have dramatically viduals. Partial cystectomy has been considered an alter-
reduced the morbidity and mortality associated with rad- native surgical procedure to radical cystectomy in an
ical cystectomy. Prior to 1970, the perioperative compli- attempt to preserve the bladder and maintain sexual
cation rate of radical cystectomy was reportedly close to function. Many of these quality-of-life issues have been
35%, with a mortality rate of nearly 20%. This has dra- significantly reduced with the development of continent
matically diminished to less than a 10% perioperative urinary diversion (orthotopic reconstruction) and nerve-
complication rate and 2% mortality rate reported in con- sparing approaches to radical cystectomy.4 Furthermore,
temporary cystectomy series.1,2 In addition, radical cys- when one considers the fact that bladder cancer is gener-
tectomy with en bloc pelvic lymphadenectomy provides ally considered a panurothelial disease and rarely an iso-
optimal local control of the bladder tumor. Pelvic recur- lated phenomena, this makes partial cystectomy an even
rence rates in patients following radical cystectomy are less desirable surgical option.
less than 10% for patients with node-negative bladder Despite the potential limitations of partial cystec-
tumors, and 10% to 20% for patients with resected pelvic tomy, certain patients may not be suitable candidates
nodal metastases.1,2,5 Furthermore, transitional cell carci- for radical cystectomy, or simply refuse this form of
noma (TCC) is generally resistant to radiation therapy therapy. Partial cystectomy may be considered in indi-
even at high doses. To date, chemotherapy alone or as viduals with the following selection criteria: a unifocal
adjuvant therapy, coupled with bladder-sparing surgery, tumor without evidence of atypia, or carcinoma in situ
has yet to demonstrate equivalent recurrence and long- (cis) in the bladder; the ability to obtain at least a 2-cm
term survival rates compared to radical cystectomy circumferential margin around the tumor; a bladder
alone.6,7 tumor in a diverticulum; no previous history of bladder

368
Chapter 21 Partial and Radical Cystectomy 369

cancer. Relative contraindications to a partial cystectomy,


En Bloc Radical Cystectomy and Pelvic-Iliac
multiple tumors, include: multiple tumors, cis of the pro-
Lymphadenectomy: Surgical Technique
static urethra or ureters, and tumors involving the trigone
Preoperative Preparation
and bladder neck or those with potential extravesical
tumor extension. Patients with a small bladder capacity Patients undergoing radical cystectomy are admitted the
are also not ideal candidates for a partial cystectomy. morning prior to surgery. All patients receive a mechan-
Considering the aforementioned selection criteria, ical and antibacterial bowel preparation the day prior to
only a small percent of patients with bladder cancer are surgery. Intravenous hydration must be considered in
appropriate candidates for a partial cystectomy. Most these patients to prevent dehydration on arrival to the
reported studies of partial cystectomy are those operating room. In addition, all patients should be eval-
reported prior to the era of orthotopic lower urinary uated and counseled by the enterostomal therapy nurse
tract reconstruction. In a 20-year experience at the prior to surgery. A clear liquid diet may be consumed
Mayo clinic, only 199 of 3454 patients (6%) were con- until midnight, at which time the patient takes nothing
sidered appropriate candidates.11 Similarly, in a 10-year per mouth. A standard modified Nichols bowel prepara-
experience at the Cleveland Clinic, only 50 of 2000 tion22 is initiated the morning of admission: 120 ml of
patients (2.5%) underwent partial cystectomy for inva- Neoloid per mouth at 9:00 a.m.; 1 g of neomycin per
sive bladder cancer.12 Although the role of a pelvic lym- mouth at 10:00 a.m., 11:00 am, 12:00 noon, 1:00 p.m.,
phadenectomy is controversial in partial cystectomy, it 4:00 p.m., 8:00 p.m., and 12:00 midnight; and 1 g of
is becoming apparent that a meticulous, extended lym- erythromycin base per mouth at 12:00 noon, 4:00 p.m.,
phadenectomy is important in patient outcome follow- 8:00 p.m., and 12:00 midnight. This regimen is generally
ing radical cystectomy.13–16 An additional consideration well tolerated, obviates the need for enemas and main-
in patients with bladder tumors undergoing partial cys- tains nutritional and hydrational support. Intravenous
tectomy is the potential for significant tumor recur- crystalloid fluid hydration is begun in the evening prior
rence,17 and tumor spill and wound implantation to surgery in those patients admitted to the hospital the
occurring in at least 10% to 20% of patients without day prior to surgery and maintained to ensure an ade-
preoperative radiation therapy.18 This may be prevented quate circulating volume as the patient enters the operat-
with a high-dose, short course of preoperative radiation ing room. This may be particularly important in the
therapy.19 elderly or frail patient with associated comorbidities.
Patients over 50 years of age routinely undergo pro-
phylactic digitalization prior to cystectomy unless a spe-
RADICAL CYSTECTOMY
cific contraindication exists. Patients younger than 50
Preoperative Evaluation
years of age are not routinely digitalized. Digoxin is given
Complete clinical staging for bladder cancer should eval- orally; 0.5 mg at 12:00 noon, 0.25 mg at 4:00 p.m., and
uate the retroperitoneum and pelvis along with the most 0.125 mg at 8:00 p.m. Our experience with preoperative
common metastatic sites, including the lungs, liver, and digitalization in patients undergoing cystectomy has been
bone. A chest x-ray, liver function tests, and serum alka- positive and there is evidence suggesting that preopera-
line phosphatase should be obtained routinely. Patients tive digitalization may decrease the risk of perioperative
with an elevated serum alkaline phosphatase or with/ dysrhythmias and congestive heart failure in the elderly
without complaints of bone pain should undergo a bone patient undergoing an extensive operative procedure.23,24
scan. A CT scan of the chest is obtained when pulmonary Attention to fluid management is important in these eld-
metastases are suspected by history, or because of an erly patients particularly on postoperative days 3 and 4
abnormal chest x-ray. A CT scan of the abdomen and when mobilization of third-space fluid is highest, subse-
pelvis is routinely performed to evaluate the pelvis and quently necessitating liberal use of diuretics. In addition,
retroperitoneum for any significant lymphadenopathy or intravenous broad-spectrum antibiotics are administered
local contiguous spread. This radiographic study should en route to the operating room, providing adequate tis-
also be performed in patients with suspected metastases, sue and circulating levels at the time of incision.
elevated liver functions tests, a bladder tumor associated Preoperative evaluation and counseling by the
with hydronephrosis, or in patients with an extensive pri- enterostomal therapy nurse is a critical component to the
mary bladder tumor that is either nonmobile or fixed; the successful care of all patients undergoing cystectomy and
results of which may impact on the decision for neoadju- urinary diversion. Patients determined to be appropriate
vant therapy. However, CT scan of the primary bladder candidates for orthotopic reconstruction are instructed
is neither sensitive nor specific enough to evaluate the how to catheterize per urethra should it be necessary
degree of bladder wall tumor invasion, or to accurately postoperatively. All patients are site marked for a cuta-
determine pelvic lymph node involvement with neous stoma, instructed in the care of a cutaneous diver-
tumor.20,21 sion (continent or incontinent form), and instructed in
370 Part IV Bladder

proper catheterization techniques should medical, tech- The anterior rectus fascia is incised, the rectus muscles
nical, or oncologic factors preclude orthotopic recon- retracted laterally, and the posterior rectus sheath and
struction. The ideal cutaneous stoma site is determined peritoneum entered in the superior aspect of the incision.
only after the patient is examined in the supine, sitting, As the peritoneum and posterior fascia are incised inferi-
and standing position. Proper stoma site selection is orly to the level of the umbilicus, the urachal remnant
important to patient acceptance, and to the technical suc- (median umbilical ligament) is identified, circumscribed,
cess of lower urinary tract reconstruction should some and removed en bloc with the cystectomy specimen
form of cutaneous diversion be necessary. Incontinent (Figure 21-2). This maneuver prevents early entry into a
stoma sites are best located higher on the abdominal wall, high-riding bladder and ensures complete removal of all
while stoma sites for continent diversions can be posi- bladder remnant tissue. Care is taken to remain medial
tioned lower on the abdomen (hidden below the belt and avoid injury to the inferior epigastric vessels (lateral
line) since they do not require an external collecting umbilical ligaments), which course posterior to the rectus
device. The use of the umbilicus as the site for catheter- muscles. If the patient has had a previous cystotomy or
ization may be employed with excellent functional and segmental cystectomy, the cystotomy tract and cutaneous
cosmetic results. incision should be circumscribed full-thickness and
excised en bloc with the bladder specimen. The medial
insertion of the rectus muscles attached to the pubic
Patient Positioning
symphysis can be slightly incised, maximizing pelvic
The patient is placed in the hyperextended supine posi- exposure throughout the operation.
tion with the superior iliac crest located at the fulcrum of
the operating table (Figure 21-1). The legs are slightly
Abdominal Exploration
abducted so that the heels are positioned near the corners
of the foot of the table. In the female patient considering A careful, systematic intraabdominal exploration is per-
orthotopic diversion, the modified frogleg or lithotomy formed to determine the extent of disease, and to evalu-
position is employed allowing access to the vagina. Care ate for any hepatic metastases, or gross retroperitoneal
should be taken to ensure that all pressure points are well lymphadenopathy. The abdominal viscera are palpated to
padded. Reverse Trendelenburg position levels the detect any concomitant unrelated disease. If no con-
abdomen parallel with the floor and helps to keep the traindication exists at this time, all adhesions should be
small bowel contents in the epigastrium. A nasogastric incised and freed.
tube is placed, and the patient is prepped from nipples to
mid-thigh. In the female patient the vagina is fully
Bowel Mobilization
prepped. After the patient is draped, a 20-F Foley
catheter is placed in the bladder and left to gravity The bowel is mobilized beginning with the ascending
drainage. A right-handed surgeon stands on the patient’s colon. A large right angle Richardson retractor elevates
left-hand side of the operating table. the right abdominal wall. The cecum and ascending
colon are reflected medially to allow incision of the lat-
eral peritoneal reflection along the avascular/white line
Incision
of Toldt. The mesentery to the small bowel is then
A vertical midline incision is made extending from the mobilized off its retroperitoneal attachments cephalad
pubic symphysis to the cephalad aspect of the epigas- (toward the ligament of Treitz) until the retroperitoneal
trium. The incision should be carried lateral to the portion of the duodenum is exposed. This mobilization
umbilicus on the contralateral side of the marked cuta- facilitates a tension-free ureteroenteric anastomosis if
neous stoma site. When considering the umbilicus as the orthotopic diversion is performed. Combined sharp and
site for a catheterizable stoma, the incision should be blunt dissection facilitates mobilization of this mesen-
directed 2 to 3 cm lateral to the umbilicus at this location. tery along a characteristic avascular fibroareolar plane.

Figure 21-1 Proper patient positioning for cystectomy in the male patient. Note, the iliac
crest is located at the break of the table.
Chapter 21 Partial and Radical Cystectomy 371

Figure 21-2 Wide excision of the urachal remnant and medial umbilical ligaments en bloc
with the cystectomy specimen.

Conceptually, the mobilized mesentery forms an the operation. This sigmoid mobilization also facilitates
inverted right triangle; the base formed by the third and retraction of the sigmoid mesentery while performing
fourth portions of the duodenum, the right edge repre- the lymph node dissection. Care should be taken to dis-
sented by the white line of Toldt along the ascending sect along the base of the mesentery and avoid injury to
colon; the left edge represented by the medial portion of the inferior mesenteric artery and blood supply to the
the sigmoid and descending colonic mesentery; and the sigmoid colon.
apex represented by the ileocecal region (Figure 21-3). Following mobilization of the bowel, a self-retaining
This mobilization is critical in setting up the operative retractor is placed. The right colon and small intestine
field and facilitates proper packing of the intraabdominal are carefully packed into the epigastrium with three
contents into the epigastrium. moist lap pads, followed by a moistened towel rolled to
The left colon and sigmoid mesentery are then mobi- the width of the abdomen. The descending and sigmoid
lized to the region of the lower pole of the left kidney by colon are not packed, and they remain as free as possible,
incising the peritoneum lateral to the colon along the providing the necessary mobility required for the ureteral
avascular/white line of Toldt. The sigmoid mesentery is and pelvic lymph node dissection.
then elevated off the sacrum, iliac vessels, and distal aorta Successful packing of the intestinal contents is an art
in a cephalad direction up to the origin of the inferior and prevents their annoying spillage into the operative
mesenteric artery (Figure 21-4). This maneuver provides field. Packing begins by sweeping the right colon and
a wide mesenteric window through which the left ureter small bowel under the surgeon’s left hand along the right
will pass (without angulation or tension) for the sidewall gutter. A moist open lap pad is then swept with
ureteroenteric anastomosis at the terminal portions of the right hand along the palm of the left hand, under the
372 Part IV Bladder

Figure 21-3 View of the pelvis from overhead; after the ascending colon and peritoneal
attachments of the small bowel mesentery have been mobilized up to the level of the
duodenum. This mobilization allows the bowel to be properly packed in the epigastrium and
exposes the area of the aortic bifurcation, which is the starting point of the lymph node
dissection.

viscera along the retroperitoneum, and sidewall gutter. In two large hemoclips. A section of the proximal cut
similar fashion, the left sidewall gutter is packed ensuring ureteral segment (distal to the proximal hemoclip) is sent
not to incorporate the descending or sigmoid colon. The for frozen section analysis to ensure the absence of carci-
central portion of the small bowel is packed with a third noma in situ or overt tumor. The ureter is then slightly
lap pad. A moist rolled towel is then positioned horizon- mobilized in a cephalad direction and tucked under the
tally below the lap pads but cephalad to the bifurcation of rolled towel to prevent inadvertent injury. Frequently, an
the aorta. Occasionally, prior to placement of the first arterial branch from the common iliac artery or the aorta
moist lap pad, a mobile greater omental apron can be needs to be divided to provide adequate ureteral mobi-
used to facilitate packing of the intestinal viscera in a sim- lization. In addition, the rich vascular supply emanating
ilar fashion to the lap pad. After the bowel has been laterally from the gonadal vessels should remain intact
packed, a wide Deaver retractor is placed with gentle and undisturbed. These attachments are an important
traction on the previous packing to provide cephalad blood supply to the ureter, which ensure an adequate vas-
exposure. cular supply for the ureteroenteric anastomosis at the
time of diversion. This is particularly important in irra-
diated patients. Leaving the proximal hemoclip on the
Ureteral Dissection
divided ureter during the exenteration allows for hydro-
The ureters are most easily identified in the retroperi- static ureteral dilation and facilitates the ureteroenteric
toneum just cephalad to the common iliac vessels. They anastomosis. In women, the infundibulopelvic ligaments
are carefully dissected into the deep pelvis (several cen- are ligated and divided at the level of the common iliac
timeters beyond the iliac vessels) and divided between vessels.
Chapter 21 Partial and Radical Cystectomy 373

Figure 21-4 View of the pelvis from overhead; after the ascending colon and small bowel
have been packed in the epigastrium. Note that the sigmoid mesentery is mobilized off the
sacral promontory and distal aorta up to the origin of the inferior mesenteric artery.

Pelvic Lymphadenectomy
The cephalad portion (2 cm above the aortic bifurca-
A meticulous pelvic lymph node dissection is routinely tion) of the lymphatics are ligated with hemoclips to pre-
performed en bloc with radical cystectomy. The extent of vent lymphatic leak, while the caudal (specimen) side is
the lymphadenectomy may vary depending on the ligated only when a blood vessel is encountered.
patient and surgeon preference. An accumulating body of Frequently, small anterior tributary veins originate from
evidence suggests that an extended lymphadenectomy the vena cava just above the bifurcation, which should be
may be beneficial in patients undergoing cystectomy for clipped and divided. In men, the spermatic vessels are
high-grade, invasive bladder cancer.13–16 When perform- retracted laterally and spared. In women, the infundibu-
ing a salvage procedure following definitive radiation lopelvic ligament along with the corresponding ovarian
treatment (>5000 rads), a pelvic lymphadenectomy is vessels has been previously ligated and divided at the
usually not performed because of the significant risk of pelvic brim as previously described.
iliac vessel and obturator nerve injury.25 All fibroareolar and lymphatic tissues are dissected
For a combined common and pelvic iliac lymphadenec- caudally off the aorta, vena cava, and common iliac vessels
tomy, the lymph node dissection is initiated 2 cm above over the sacral promontory into the deep pelvis. The ini-
the aortic bifurcation (superior limits of dissection), and tial dissection along the common iliac vessels is performed
the dissection extends laterally over the inferior vena cava over the arteries, skeletonizing them. As the common iliac
to the genitofemoral nerve, representing the lateral limits veins are dissected medially, care is taken to control small
of dissection. Distally, the lymph node dissection extends arterial and venous branches coursing along the anterior
to the lymph node of Cloquet medially (on Cooper’s liga- surface of the sacrum. Electrocautery is helpful at this
ment) and the circumflex iliac vein laterally. location, which allows the adherent fibroareolar tissue to
374 Part IV Bladder

be swept off the sacral promontory down into the deep attached only at the base within the obturator fossa. The
pelvis with the use of a small gauze sponge. Significant lateral lymphatic compartment (freed medially from the
bleeding from these presacral vessels can occur if not vessels and laterally from the psoas) is bluntly swept into
properly controlled. Hemoclips are discouraged in this the obturator fossa by retracting the iliac vessels medially
location as they can be easily dislodged from the anterior and passing a small gauze sponge lateral to the vessels
surface of the sacrum, resulting in troublesome bleeding. along the psoas and pelvic sidewall (Figure 21-5B). This
Once the proximal portion of the lymph node dissec- sponge should be passed anterior and distal to the
tion is completed, a finger is passed from the proximal hypogastric vein, directed caudally into the obturator
aspect of dissection under the pelvic peritoneum (ante- fossa. The external iliac vessels are then elevated and
rior to the iliac vessels), distally toward the femoral canal. retracted laterally, and the gauze sponge is carefully with-
The opposite hand can be used to strip the peritoneum drawn from the obturator fossa with gentle traction using
from the undersurface of the transversalis fascia and to the left hand (Figure 21-6A). This maneuver effectively
connect with the proximal dissection from above. This sweeps all lymphatic tissue into the obturator fossa and
maneuver elevates the peritoneum and defines the lateral facilitates identification of the obturator nerve deep to the
limit of peritoneum to be incised and removed with the external iliac vein. The obturator nerve is best identified
specimen. The peritoneum is divided medial to the sper- proximally and carefully dissected free from all lymphat-
matic vessels in men and lateral to the infundibulopelvic ics. The obturator nerve is then retracted laterally along
ligament in female patients. The only structure encoun- with the iliac vessels (Figure 21-6B). At this point, the
tered is the vas deferens in the male or round ligament in obturator artery and vein should be carefully entrapped
females; these structures are clipped and divided. between the index finger (medial to the obturator nerve)
A large right-angled rake retractor (e.g., Israel) is used to laterally and the middle finger medially with the left hand.
elevate the lower abdominal wall, including the spermatic This isolates the obturator vessels exiting the obturator
cord or remnant of the round ligament, to provide distal canal along the pelvic floor. These vessels are then care-
exposure in the area of the femoral canal. Tension on the fully clipped and divided ensuring to stay medial to the
retractor is directed vertically toward the ceiling, with care obturator nerve. The obturator lymph node packet is then
taken to avoid injury to the inferior epigastric vessels. This swept medially toward the sidewall of the bladder, ligating
provides excellent exposure to the distal external iliac ves- small tributary vessels and lymphatics from the pelvic
sels. The distal limits of the dissection are then identified: sidewall. The nodal packet will be removed en bloc with
the circumflex iliac vein crossing anterior to the external the cystectomy specimen.
iliac artery distally, the genitofemoral nerve laterally, and
Cooper’s ligament medially. The lymphatics draining the
Ligation of the Lateral Vascular Pedicle
ipsilateral leg, particularly medial to the external iliac vein,
to the Bladder
are carefully clipped and divided to prevent lymphatic leak-
age. This includes the lymph node of Cloquet (also known Following dissection of the obturator fossa and dividing
as Rosenmüller), which represents the distal limit of the the obturator vessels, the lateral vascular pedicle to the
lymphatic dissection at this location. The distal external bladder is isolated and divided. Developing this plane iso-
iliac artery and vein are then circumferentially dissected lates the lateral vascular pedicle to the bladder; a critical
and skeletonized with care taken to ligate an accessory maneuver in performing a safe cystectomy with proper
obturator vein (present in 40% of patients) originating vascular control. Isolation of the lateral vascular pedicle is
from the inferomedial aspect of the external iliac vein. performed with the left hand. The bladder is retracted
Following completion of the distal limits of dissection, toward the pelvis, placing traction and isolating the ante-
the proximal and distal dissections are joined. The proxi- rior branches of the hypogastric artery. The left index fin-
mal external iliac artery and vein are skeletonized circum- ger is passed medial to the hypogastric artery, posterior to
ferentially to the origin of the hypogastric artery (Figure the anterior visceral branches, and lateral to the previ-
21-5A). Care should be taken to clip and divide a com- ously transected ureter. The index finger is directed cau-
monly encountered vessel arising from the lateral aspect dally toward the endopelvic fascia, parallel to the sweep of
of the proximal external iliac vessels coursing to the psoas the sacrum. This maneuver defines the two major vascu-
muscle. The external iliac vessels (artery and vein) are lar pedicles to the anterior pelvic organs: the lateral pedi-
then retracted medially, and the fascia overlying the psoas cle—anterior to the index finger, composed of the visceral
muscle is incised medial to the genitofemoral nerve. On branches of the anterior hypogastric vessel; the posterior
the left side, branches of the genitofemoral nerve often pedicle—posterior to the index finger, composed of the
pursue a more medial course and may be intimately visceral branches between the bladder and rectum.
related to the iliac vessels, in which case they are excised. With the lateral pedicle entrapped between the left
At this point, the lymphatic tissue surrounding the iliac index and middle fingers, firm traction is applied verti-
vessels are composed of a medial and lateral components cally and caudally. This facilitates identification and
Chapter 21 Partial and Radical Cystectomy 375

Figure 21-5 A, Technique of skeletonizing the external iliac artery and vein. Note, the
vessels are completely dissected free up to the level of the origin of the hypogastric artery.
This allows for the vessels to be carefully retracted medially and the psoas fascia incised to
allow passage of a gauze sponge. B, Technique of passing a small gauze sponge lateral to
the external iliac vessels and medial to the psoas muscle.

allowing individual branches off the anterior portion of is positioned as far apart as possible to ensure that 0.5 to
the hypogastric artery to be isolated (Figure 21-7). The 1 cm of tissue projects beyond each clip when the pedicle
posterior division of the hypogastric artery, including the is divided. This prevents the hemoclips from being dis-
superior gluteal, iliolumbar, and lateral sacral arteries, is lodged resulting in unnecessary bleeding. Occasionally,
preserved to avoid gluteal claudication. Distal to this pos- in patients with an abundance of pelvic fat, the lateral
terior division, the hypogastric artery may be ligated for pedicle may be thick and require division into two man-
vascular control but should not be divided since the lat- ageable pedicles. The inferior vesicle vein serves as an
eral pedicle is easier to dissect if left in continuity. The excellent landmark as the endopelvic fascia is just distal to
largest and most consistent anterior branch to the blad- this structure. The endopelvic fascia just lateral to the
der, the superior vesical artery, is usually isolated and prostate may then be incised, which helps to identify the
individually ligated and divided easily. The remaining distal limit of the lateral pedicle.
anterior branches of the lateral pedicle are then isolated
and divided between hemoclips down to the endopelvic
Ligation of the Posterior Pedicle to the Bladder
fascia, or as far as is technically possible. With blunt dis-
section the index finger of the left hand helps identify this Following division of the lateral pedicles, the bladder spec-
lateral pedicle and protects the rectum as it is pushed imen is retracted anteriorly exposing the cul-de-sac (pouch
medially. Right angle hemoclip appliers are ideally suited of Douglas). The surgeon elevates the bladder with a small
for proper placement of the clips. Each pair of hemoclips gauze sponge under the left hand, while the assistant
376 Part IV Bladder

Figure 21-6 A, Technique of withdrawing the gauze sponge with the left hand. This aids in
dissecting and clearing the obturator fossa, sweeping all fibroareolar and lymphatic tissue
toward the bladder. B, Obturator fossa cleaned. This allows proper identification of the
obturator nerve passing deep to the external iliac vein.

retracts on the peritoneum of the rectosigmoid colon in a the prostate and seminal vesicles anterior to the rectum
cephalad direction. This provides excellent exposure to the posterior. The plane between the prostate and seminal
recess of the cul-de-sac and places the peritoneal reflection vesicles, and the anterior sheath of Denonvilliers’ will not
on traction facilitating the proper division. The peri- develop easily. However, the plane between the rectum
toneum lateral to the rectum is incised and extended ante- and the posterior sheath of Denonvilliers’ (Denonvilliers’
riorly and medially across the cul-de-sac to join the space) should develop easily with blunt and sharp dissec-
incision on the contralateral side (Figure 21-8). tion. Therefore, the peritoneal incision in the cul-de-sac
An understanding of the fascial layers is critical for the must be made slightly on the rectal side rather than the
appropriate dissection of this plane. The anterior and bladder side (see Figure 21-9, small arrow). This allows
posterior peritoneal reflections converge in the cul-de- proper and safe entry and development of Denonvilliers’
sac to form Denonvilliers’ fascia, which extends caudally space between the anterior rectal wall and the posterior
to the urogenital diaphragm (Figure 21-9, large arrow). sheath of Denonvilliers’ fascia (Figure 21-10).
This important anatomic boundary in the male separates Employing a posterior sweeping motion of the fingers,
Chapter 21 Partial and Radical Cystectomy 377

Figure 21-7 Isolation of the lateral vascular pedicle. The left hand is used to define the right
lateral pedicle, extending from the bladder to the hypogastric artery. This plane is developed
by the index finger (medial) and the middle finger (lateral), exposing the anterior branches of
the hypogastric artery. This vascular pedicle is clipped and divided down to the endopelvic
fascia. Traction with the left hand defines the pedicle, allows direct visualization, and
protects the rectum from injury.

the rectum can be carefully swept off the seminal vesicles, making the posterior dissection difficult. To prevent
prostate, and bladder in men and off the posterior vagi- injury to the rectum in these situations, only sharp dis-
nal wall in women. This sweeping motion, when section should be performed under direct vision. To pre-
extended laterally, helps to thin and develop the posterior vent a rectal injury is to avoid blunt dissection with the
pedicle, which appears like a collar emanating from the finger in areas where normal tissue planes have been
lateral aspect of the rectum. Care should be taken as one obliterated by previous surgery or radiation. Sharp dis-
develops this posterior plane more caudally as the ante- section under direct vision will dramatically reduce the
rior rectal fibers often are adherent to the specimen and potential for rectal injury. If a rectotomy occurs, a 2- or
can be difficult to bluntly dissect. In this region, just 3-layer closure is recommended. A diverting proximal
cephalad (proximal) to the urogenital diaphragm, sharp colostomy is not routinely required unless gross contam-
dissection may be required to dissect the anterior rectal ination occurs, or if the patient has received previous
fibers off the apex of the prostate in order to prevent rec- pelvic radiation therapy. If orthotopic diversion or vaginal
tal injury at this location. reconstruction is planned, an omental interposition is rec-
Particular mention should be made concerning several ommended to prevent fistulization between suture lines.
situations that may impede the proper development of Once the posterior pedicles have been defined, they
this posterior plane. Most commonly, when the incision are clipped and divided to the endopelvic fascia in the
in the cul-de-sac is made too far anteriorly, proper entry male patient. The endopelvic fascia is then incised adja-
into Denonvilliers’ space is prevented. Improper entry cent to the prostate, medial to the levator ani muscles (if
can occur in between the two layers of Denonvilliers’ fas- not done previously), to facilitate the apical dissection. In
cia, or even anterior to this, making the posterior dissec- the female patient, the posterior pedicles, including the
tion difficult, increasing the risk of rectal injury. cardinal ligaments, are divided 4 to 5 cm beyond the
Furthermore, posterior tumor infiltration, or previous cervix. With cephalad pressure on a previously placed
high-dose pelvic irradiation can obliterate this plane vaginal sponge stick, the apex of the vagina can be iden-
378 Part IV Bladder

Figure 21-8 The peritoneum lateral to the rectum is incised down into the cul-de-sac and
carried anteriorly over the rectum to join the opposite side. Note that the incision should be
made precisely so the proper plane behind Denonvilliers’ fascia can be developed safely.

tified and incised posteriorly just distal to the cervix. The the Foley catheter balloon. At this point, the specimen
vagina is then circumscribed anteriorly with the cervix remains attached only at the apex in men and vesi-
attached to the cystectomy specimen. If there is concern courethral junction in women.
about an adequate surgical margin at the posterior or
base of the bladder, then the anterior vaginal wall should
Anterior Apical Dissection in the Male Patient
be removed en bloc with the bladder specimen; subse-
quently requiring vaginal reconstruction if sexual func- Once the cystectomy specimen is completely freed and
tion is desired. It is our preference to spare the anterior mobile posteriorly, attention is directed anteriorly to the
vaginal wall if orthotopic diversion is planned. This elim- pelvic floor and urethra. All fibroareolar connections
inates the need for vaginal reconstruction, and helps between the anterior bladder wall, prostate, and under-
maintain the complex musculofascial support system and surface of the pubic symphysis are divided. The
helps prevent injury to the pudendal innervation to the endopelvic fascia is incised adjacent to the prostate and
rhabdosphincter proximal urethra, both important com- the levator muscles are carefully swept off the lateral and
ponents to the continence mechanism in women. The apical portions of the prostate. It must be emphasized
anterior vaginal wall is then sharply dissected off the pos- that minimal dissection is to be performed along the
terior bladder down to the region of the bladder neck pelvic floor. The innervation to the rhabdosphincter and
(vesicourethral junction), which is identified by palpating continence mechanism arises from the pudendal innervation
Chapter 21 Partial and Radical Cystectomy 379

Figure 21-9 Illustration of the formation of Denonvilliers’ fascia. Note that it is derived from a
fusion of the anterior and posterior peritoneal reflections. Denonvilliers’ space lies behind the
fascia. To successfully enter this space and facilitate mobilization of the anterior rectal wall
off Denonvilliers’ fascia, the incision in the cul-de-sac is made close to the peritoneal fusion
on the anterior rectal wall side, and not on the bladder side.

and courses along the pelvic floor.10 Therefore, any slightly divided just beneath the pubis, lateral to the dor-
unnecessary dissection may disrupt this delicate innerva- sal venous complex that courses between these ligaments.
tion and may compromise the continence mechanism in Care should be taken in avoiding any extensive dissection
patients undergoing orthotopic reconstruction. in this region along the pelvic floor. The puboprostatic
The superficial dorsal vein is identified, ligated, and ligaments need only to be incised enough to allow for a
divided. With tension placed posteriorly on the prostate, proper apical dissection of the prostate. The apex of the
the puboprostatic ligaments are identified, and only prostate and membranous urethra now becomes palpable.
380 Part IV Bladder

ments between the dorsal venous complex and rhab-


dosphincter, which could potentially injure these struc-
tures and compromise the continence mechanism. After
the complex has been ligated, it can be sharply divided
with excellent exposure to the anterior surface of the ure-
thra. Once the venous complex has been severed, the
suture can be used to further secure the complex. The
suture is then used to suspend the venous complex ante-
riorly to the periosteum to help reestablish anterior fixa-
tion of the dorsal venous complex and puboprostatic
ligaments (Figure 21-14). This may enhance continence
recovery. The anterior urethra is now exposed.
Regardless of the aforementioned technique to control
the dorsal venous complex, the urethra is then incised 270
degrees just beyond the apex of the prostate. A series of 2-
0 polyglycolic acid sutures are placed in the anterior ure-
thra, carefully incorporating only the mucosa and
submucosa of the striated urethral sphincter muscle ante-
riorly. The catheter is clamped and divided, allowing two
posterior sutures to be placed in the urethra, which should
incorporate the rectourethralis muscle or the caudal extent
of Denonvilliers’ fascia posteriorly. Following this, the
posterior urethra is divided and the specimen removed.
Alternatively, the dorsal venous complex can be
sharply transected without securing vascular control of
the dorsal venous complex. Cephalad traction on the
prostate elongates the proximal and membranous ure-
thra, and allows the urethra to be skeletonized laterally
by dividing the so-called “lateral pillars,” extensions of
Figure 21-10 After the peritoneum of the cul-de-sac has the rhabdosphincter. The anterior two-third of the ure-
been incised, the anterior rectal wall can be swept off the
thra is divided, exposing the urethral catheter. The ure-
posterior surface of the Denonvilliers’ fascia. This effectively
defines the posterior pedicle that extends from the bladder to
thral sutures are then placed. Six 2-0 polyglycolic acid
the lateral aspect of the rectum on either side. sutures are placed, equally spaced, into the urethral
mucosa and lumen anteriorly. The rhabdosphincter, the
edge of which acts as a hood overlying the dorsal venous
Several methods can be performed to properly control complex, is included in these sutures if the dorsal venous
the dorsal venous plexus. One may carefully pass an complex was sharply incised. This maneuver compresses
angled clamp beneath the dorsal venous complex, ante- the dorsal vein complex against the urethra for hemosta-
rior to the urethra (Figure 21-11). The venous complex tic purposes. The urethral catheter is then drawn
can then be ligated with a 2-0 absorbable suture and through the urethrotomy, clamped on the bladder side,
divided close to the apex of the prostate. If any bleeding and divided. Cephalad traction on the bladder side with
occurs from the transected venous complex, it can be the clamped catheter occludes the bladder neck, prevents
oversewn with an absorbable (2-0 polyglycolic acid) tumor spill from the bladder, and provides exposure to
suture. In a slightly different fashion, the dorsal venous the posterior urethra. Two additional sutures are placed
complex may be gathered at the apex of the prostate with in the posterior urethra, again incorporating the rec-
a long Allis clamp (Figure 21-12). This may help better tourethralis muscle or distal Denonvilliers’ fascia. The
define the plane between the dorsal venous complex and posterior urethra is then divided and the specimen is
anterior urethra. A figure-of-eight 2-0 absorbable suture removed. Bleeding from the dorsal vein is usually mini-
can then be placed under direct vision anterior to the mal at this point. If additional hemostasis is required, 1 or
urethra (distal to the apex of the prostate) around the 2 anterior urethral sutures can be tied to stop the bleed-
gathered venous complex (Figure 21-13). This suture is ing. Regardless of the technique, frozen section analysis
best placed with the surgeon facing the head of the table of the distal urethral margin of the cystectomy specimen
and holding the needle driver perpendicular to the is then performed to exclude tumor involvement.
patient. The suture is then tagged with a hemostat. This If a cutaneous form of urinary diversion is planned,
maneuver avoids the unnecessary passage of any instru- urethral preparation is slightly modified. Once the dorsal
Chapter 21 Partial and Radical Cystectomy 381

Figure 21-11 Control of the dorsal venous complex. A right-angled clamp can be passed
posterior to the venous complex and anterior to the urethra. An absorbable suture can be
passed to ligate the complex distal to the apex of the prostate.

Figure 21-12 The dorsal venous complex is gathered with Figure 21-13 An absorbable suture is carefully passed in a
an Allis clamp distal to the apex of the prostate. This figure-of-eight fashion anterior to the urethra around the
maneuver will define the plane between the dorsal venous gathered dorsal venous complex to control the vascular
complex and urethra. structure.
382 Part IV Bladder

anterior vaginal wall and the bladder specimen. Careful


dissection of the proper plane will prevent entry into the
posterior bladder and also reduce the amount of bleeding
in this vascular area (Figure 21-16). Development of this
posterior plane and vascular pedicle is best performed
sharply and carried just distal to the vesicourethral junc-
tion (Figure 21-17). Palpation of the Foley catheter bal-
loon assists in identifying this region. This dissection
effectively maintains a functional vagina.
In the case of a deeply invasive posterior bladder
tumor in women, with concern of an adequate surgical
margin, the anterior vaginal wall should be removed en
bloc with the cystectomy specimen. After dividing the
posterior vaginal apex, the lateral vaginal wall subse-
quently serves as the posterior pedicle and is divided dis-
tally. This leaves the anterior vaginal wall attached to the
posterior bladder specimen. The Foley catheter balloon
Figure 21-14 The dorsal venous complex is completely again facilitates identification of the vesicourethral junc-
divided. The previously placed suture is then used to further tion. The surgical plane between the vesicourethral
secure the venous complex. The complex is then fixed
junction and the anterior vaginal wall is then developed
anteriorly to the periosteum.
distally at this location. A 1-cm length of proximal ure-
thra is mobilized, while the remaining distal urethra is
venous complex is secured and divided, the anterior ure- left intact with the anterior vaginal wall. Vaginal recon-
thra is identified. The urethra is mobilized from above as struction by a clam shell (horizontal) or side-to-side (ver-
far distally as possible into the pelvic diaphragm. With tical) technique is required. Other means of vaginal
cephalad traction, the urethra is stretched above the uro- reconstruction may include a rectus myocutaneous flap,
genital diaphragm, a curved clamp is placed as distal on the detubularized cylinder of ileum, a peritoneal flap, or an
urethra as feasible and divided distal to the clamp. Care omental flap.
must be taken avoid rectal injury with this clamp. This is It is emphasized that no dissection to be performed
prevented by placing gentle posterior traction with the left anterior to the urethra along the pelvic floor. The
hand or index finger on the rectum and ensuring that the endopelvic fascia should remain undisturbed and not to
clamp is passed anterior. The specimen is then removed. be opened in women considering orthotopic diversion.
This mobilization of the urethra will facilitate a late ure- This prevents injury to the rhabdosphincter region and
threctomy. The levator musculature can then be reapprox- corresponding innervation, which is critical in maintain-
imated along the pelvic floor to facilitate hemostasis. ing the continence mechanism. Anatomic studies have
demonstrated that the innervation to this rhabdosphinc-
ter region in women arises from branches off the puden-
Anterior Dissection in The Female
dal nerve that course along the pelvic floor posterior to
The wide female pelvis allows for better anterior expo- the levator muscles.26,27 Any dissection performed ante-
sure in women, particularly at the vesicourethral junc- riorly may injure these nerves and compromise the con-
tion. However, urologists may be less familiar with pelvic tinence status. This surgical principle similarly applies to
surgery in women than in men. In addition, paravaginal the male patients as previously described.
vascular control may be troublesome in women, and the When the posterior dissection is completed (ensuring
venous plexus anterior to the urethra is less well defined to dissect just distal to the vesicourethral junction), a
in women. When considering orthotopic diversion in Satinsky vascular clamp is placed across the bladder neck.
female patients undergoing cystectomy, several technical The Satinsky vascular clamp placed across the catheter at
issues are critical to the procedure in order to maintain the bladder neck prevents any tumor spill from the blad-
the continence mechanism in these women.10 der. With gentle traction the proximal urethra is com-
When developing the posterior pedicles in women, pletely divided anteriorly, distal to the bladder neck and
the posterior vagina is incised at the apex just distal to the clamp. The female urethra is situated more anteriorly
cervix (Figure 21-15). This incision is carried anteriorly than in men, and the urethral sutures can be placed easily
along the lateral and anterior vaginal wall forming a cir- after the specimen is completely removed (Figure 21-18).
cumferential incision. The anterolateral vaginal wall is A total of 10 to 12 sutures are placed. Frozen section
then grasped with curved Kocher clamps. This provides analysis is performed on the distal urethral margin of the
countertraction and facilitates dissection between the cystectomy specimen to exclude tumor. Once hemostasis
Chapter 21 Partial and Radical Cystectomy 383

Figure 21-15 In women, the vagina is incised distal to the cervix. Note that cephalad
traction on the posterior aspect of the vagina facilitates the incision of the anterior vaginal
wall. Slight dissection of the posterior vaginal wall off the rectum provides mobility to the
vaginal cuff.

is obtained, the vaginal cuff may be closed in 2 layers with previously described and suspended. Alternatively, a per-
absorbable sutures. The vaginal cuff is then anchored ineal approach is to be used for this dissection with com-
via a colposacralpexy using a strut of Marlex mesh to plete removal of the entire urethra.
the sacral promontory. This fixates the vagina without Following removal of the cystectomy specimen, the
angulation or undo tension. Note, at the terminal por- pelvis is irrigated with warm sterile water. The presacral
tions of the operation, a well-vascularized omental nodal tissue previously swept off the common iliac vessels
pedicle graft is placed between the reconstructed and sacral promontory into the deep pelvis is collected,
vagina and neobladder, and secured to the levator ani and sent separately for pathologic evaluation. Nodal tis-
muscles to separate the suture lines and prevent fis- sue in the presciatic notch, anterior to the sciatic nerve,
tulization (Figure 21-19). is also sent for histologic analysis. Hemostasis is obtained
If a cutaneous diversion is planned in the female and the pelvis is packed with a lap pad, while attention is
patient, the posterior pedicles are developed as previ- directed to the urinary diversion.
ously mentioned. Attention is then directed anteriorly The use of various tubes and drains postoperatively is
and the pubourethral ligaments are divided. A curved important. The pelvis is drained for urine or lymph leak
clamp is placed across the urethra, and the anterior vagi- with a 1-in. Penrose drain for 3 weeks, and a large suc-
nal wall is opened distally and incised circumferentially tion Hemovac drain for the evacuation of blood for
around the urethral meatus. The vaginal cuff is closed as 24 hours. A gastrostomy tube with an 18 French Foley
384 Part IV Bladder

Figure 21-16 Dissection of the anterior vaginal wall off of the bladder. Note caudal traction
of the cystectomy specimen with countertraction applied to the vagina in a cephalad
direction.

catheter is routinely placed utilizing a modified Stamm dous and deceiving. Patients with compromised cardiac
technique, which incorporates a small portion of omen- function or pulmonary function may require invasive car-
tum (near the greater curvature of the stomach) inter- diac monitoring with a pulmonary artery catheter placed
posed between the stomach and the abdominal wall.28 prior to surgery to precisely ascertain the cardiac
This provides a simple means to drain the stomach and response to fluid shifts. A combination of crystalloid and
prevents the need for an uncomfortable nasogastric tube, colloid fluid replacement is given on the night of surgery,
while the postoperative ileus resolves. and converted to crystalloid on postoperative day 1.
Prophylaxis against stress ulcer is initiated with an H2
blocker. Intravenous broad-spectrum antibiotics are con-
POSTOPERATIVE CARE
tinued in all patients and subsequently converted to oral
A meticulous, team-oriented approach to the care of antibiotics as the diet progresses. Pulmonary toilet is
these generally elderly patients undergoing radical cys- encouraged with incentive spirometry, deep breathing,
tectomy helps reduce perioperative morbidity and mor- and coughing.
tality. Patients are best monitored in the surgical Prophylaxis against deep vein thrombosis is impor-
intensive care unit (ICU) for at least 24 hours or until sta- tant in these patients undergoing extensive pelvic oper-
ble. Careful attention to fluid management is imperative ations for malignancies. The anticoagulation is initiated
as third space fluid loss in these patients can be tremen- in the recovery room with 10 mg of sodium warfarin via
Chapter 21 Partial and Radical Cystectomy 385

Figure 21-17 Dissection continues only slightly distal to the vesicourethral junction. This can
be identified by palpation of the Foley balloon in the bladder.

a nasogastric or the gastrostomy tube. The daily dose is later, in which situation the patient may become farther
adjusted to maintain a prothrombin time in the range of behind nutritionally.
18 to 22 seconds. If the prothrombin time exceeds 22
seconds, 2.5 mg of vitamin K is administered intramus-
DISCUSSION
cularly to prevent bleeding. No systemic anticoagula-
tion is used. Pain control by a patient-controlled Improvements in medical, surgical, and anesthetic ther-
analgesic system provides comfort and enhances deep apy have reduced the morbidity and mortality associated
breathing, and early ambulation. If digoxin was given with radical cystectomy. An anticipated perioperative
preoperatively, it is continued until discharge. The gas- mortality rate following cystectomy is 1% to 3%.1,2 The
trostomy tube is removed on postoperative day 7, or administration of preoperative therapy (radiation and/or
later if bowel function is delayed. The catheter and chemotherapy) and the form of urinary diversion per-
drain management is specific to the form of urinary formed (continent or incontinent) do not appear to alter
diversion. Some patients may develop a prolonged ileus the mortality rate or the perioperative complication
or some other complication that delays the quick return rate.1 Strict attention to perioperative details, meticulous
of oral intake. In such circumstances, total parenteral surgery, and a team-oriented surgical and postoperative
nutrition (TPN) is wisely instituted earlier rather than approach is the key to minimize morbidity and mortality,
386 Part IV Bladder

Figure 21-18 View of the female pelvis from above with open vaginal cuff and urethral
suture placement.

Figure 21-19 Sagittal section of the female pelvis. Not a vascularized omental pedicle graft
is situated between the reconstructed vagina/vaginal cuff and the neobladder. The graft is
secured to the pelvic floor to prevent fistulization.
Chapter 21 Partial and Radical Cystectomy 387

and to ensure the best clinical outcomes following radical advances in lower urinary tract reconstruction have
cystectomy. allowed a reasonable alternative for patients following
The development of orthotopic lower urinary tract removal of the bladder and have improved the quality of
reconstruction has dramatically lessened the impact of life of these patients requiring removal of their bladder.
cystectomy on the quality of life of patients following
removal of their bladder.7 Orthotopic diversion has elimi-
nated the need for a cutaneous stoma, urostomy appliance, REFERENCES
and the need for intermittent catheterization. Continence
rates following orthotopic diversion are excellent, provid- 1. Stein JP, Lieskovsky G, Cote R, et al: Radical cystectomy
ing patients a more natural voiding pattern per urethra. in the treatment of invasive bladder cancer: long-term
Currently, orthotopic diversion should be considered the results in 1054 patients. J Clin Oncol 2001; 19:666–675.
diversion of choice in all cystectomy patients and the urol- 2. Ghoneim MA, El-Mekresh MM, El-Baz MA, El-Attar IA,
ogist should have a specific reason why an orthotopic Ashamallah A: Radical cystectomy for carcinoma of the
diversion is not performed. Patient factors, such as frail bladder: critical evaluation of the results in 1026 cases.
J Urol 1997; 158:393–399.
general health, motivation, associated comorbidity, and
3. Skinner DG, Daniels J, Russell C, et al: The role of
the oncologic issue of a positive urethral margin, will dis- adjuvant chemotherapy following cystectomy for invasive
qualify some patients. Nevertheless, the option of lower bladder cancer: a prospective comparative trial. J Urol
urinary tract reconstruction to the intact urethra has been 1991; 145:459–467.
shown to decrease physician’s reluctance and increase 4. Stein JP, Skinner DG: Orthotopic bladder replacement. In
patient’s acceptance to undergo earlier cystectomy when Walsh PC, Retik AB, Vaughan ED, Wein AJ (eds):
the disease may be at a more curable stage.29 Campbell’s Urology, 8th edition, Chapter 108,
Currently, no equally effective alternative form of ther- pp 3835–3864. Philadelphia, WB Saunders, 2002.
apy for high-grade, invasive bladder cancer has evolved. 5. Lerner SP, Skinner DG, Lieskovsky G, et al: The
Bladder cancer appears to be resistant to radiation therapy, rationale for en bloc pelvic lymph node dissection for
even at high doses. Other bladder-sparing techniques bladder cancer patients with nodal metastases: long-term
results. J Urol 1993; 149:758–765.
employing chemotherapy alone, or in combination with
6. Thrasher JB, Crawford ED: Current management of
radiation therapy have substantially higher local recur- invasive and metastatic transitional cell carcinoma of the
rence rates and do not appear to result in long-term bladder. J Urol 1993; 149:957–972.
survival or recurrence rates comparable to radical cystec- 7. Montie JE: Against bladder sparing surgery. J Urol 1999;
tomy.6,7 Whether patients have a better quality of life fol- 162:452–457.
lowing cystectomy or following bladder-sparing 8. Stein JP, Skinner DG: Radical cystectomy in the female.
protocols that require significant treatment to the bladder In Montie JE (ed): Atlas of Urologic Clinics of North
with the potential for tumor recurrence, has not been America, Vol 5, No 2, pp 37–64. Philadelphia, WB
clarified. However, the authors firmly believe that the Saunders, 1997.
argument for bladder-sparing protocols has diminished 9. Stein JP, Skinner DG, Montie JE: Radical cystectomy and
with the advent and successful application of orthotopic pelvic lymphadenectomy in the treatment of infiltrative
bladder cancer. In Droller MJ (ed): Bladder Cancer:
diversion following radical cystectomy.
Current Diagnosis and Treatment, Chapter 10,
Unlike any other therapy, radical cystectomy patho- pp 267–307. Totowa, NJ, Humana Press, 2001.
logically stages the primary bladder tumor and regional 10. Stein JP, Quek MD, Skinner DG: Contemporary surgical
lymph nodes. This histologic evaluation will provide techniques for continent urinary diversion: continence
important prognostic information and may help identify and potency preservation. In Libertino JA, Zinman LN
high-risk patients who could benefit from adjuvant ther- (eds): Atlas of the Urologic Clinics of North America, Vol
apy. Patients with extravesical tumor extension, or with 9, pp 147–173. Philadelphia, WB Saunders, 2001.
lymph node positive disease, are at risk for recurrence 11. Utz DC, Schmitz SE, Fugelso PD, et al: A
and should be considered for adjuvant treatment strate- clinicopathologic evaluation of partial cystectomy
gies. Additionally, the recent application of molecular for carcinoma of the urinary bladder. Cancer 1973;
markers, based on pathologic staging and analysis, may 32:1075.
12. Novick AC, Stewart BH: Partial cystectomy in the
also serve to identify patients at risk for tumor recurrence
treatment of primary and secondary carcinoma of the
who may benefit from adjuvant forms of therapy.30 bladder. J Urol 1976; 116:570.
In conclusion, a properly performed radical cystec- 13. Herr HW, Bochner BH, Dalbagni G, et al: Impact of the
tomy with en bloc lymphadenectomy provides the best number of lymph nodes retrieved on outcome in patients
survival rates, with the lowest reported local recurrence with muscle invasive bladder cancer. J Urol 2002;
rates for high-grade invasive bladder cancer. The surgical 167:1295–1298.
technique is critical to optimize the best clinical and 14. Leissner J, Hohenfellner R, Thuroff JW, Wolf H.K:
technical outcomes with this procedure. Technical Lymphadenectomy in patients with transitional cell
388 Part IV Bladder

carcinoma of the urinary bladder; significance for staging intestinal preparation on the incidence of infectious
and prognosis. Brit J Urol Int 2000; 85:817–823. complications following colon surgery. Ann Surg 1973;
15. Poulsen AL, Horn T, Steven K: Radical cystectomy; 178:453–462.
extending limits of pelvic lymph node dissection improves 23. Pinaud MLJ, Blanloeil YAG, Souron RJ: Preoperative
survival for patients with bladder cancer confined to the prophylactic digitalization of patients with coronary artery
bladder wall. J Urol 1998; 160:2015. disease: a randomized echocardiographic and
16. Stein JP, Cai J, Groshen S, Skinner DG: Risk factors for hemodynamic study. Anesth Analg 1983; 62:685–689.
patients with pelvic lymph node metastases following 24. Burman SO: The prophylactic use of digitalis before
radical cystectomy with en bloc cystectomy: the concept thorocotomy. Ann Thorac Surg 1972; 14:359–368.
of lymph node density. J Urol 2003; 170:35–41. 25. Crawford ED, Skinner DG: Salvage cystectomy after
17. Faysal MH, Frieha FS: Evaluation of partial cystectomy radiation failure. J Urol 1980; 123:32–34.
for carcinoma of the bladder. Urology 1979; 14:352. 26. Colleselli K, Stenzl A, Eder R, et al: The female urethral
18. Magri J: Partial cystectomy: review of 104 cases. Br J Urol sphincter: a morphological and topographical study.
1962; 32:74. J Urol 1998; 160:49–50.
19. van der Werf-Messing B: Carcinoma of the bladder 27. Grossfeld GD, Stein JP, Bennett CJ, et al: Lower urinary
treated by suprapubic radium implants: the value of tract reconstruction in the female using the Kock ileal
additional external irradiation. Eur J Urol 1969; 5:277. reservoir with bilateral ureteroileal urethrostomy: update
20. Voges GE, Tauschke E, Stockle M, Alken P, Hohenfellner of continence results and fluorourodynamic findings.
R: Computerized tomography: an unreliable method for Urology 1996; 48:383–388.
accurate staging of bladder tumors in patients who are 28. Buscarini M, Stein JP, Lawrence MA, Skinner DG: Tube
candidates for radical cystectomy. J Urol 1989; gastrostomy following radical cystectomy and urinary
142:972–974. diversion: surgical technique and experience in 709
21. Pagano F, Bassi P, Galetti TP, et al:: Results of patients. Urology 2000; 56:150–152.
contemporary radical cystectomy for invasive bladder 29. Hautmann RE, Paiss T: Does the option of the ileal
cancer: a clinicopathological study with an emphasis on neobladder stimulate patient and physician decision
the inadequacy of the tumor, nodes and metastases toward earlier cystectomy? J Urol 1998; 159:1845–1850.
classification. J Urol 1991; 145:45–50. 30. Stein JP, Grossfeld GD, Ginsberg DA, et al: Prognostic
22. Nichols RL, Broido P, Condon RE, Gorbach SL, Nyhus markers in bladder cancer: a contemporary review of the
LM: Effect of preoperative neomycin-erythromycin literature. J Urol 1999; 160:645–659.
C H A P T E R

22 Selective Bladder Preservation by


Combined Modality Treatment
Donald S. Kaufman, MD, Alex F. Althausen, MD,
Niall M. Heney, MD, FRCS, Anthony L. Zietman, MD, M. Dror
Michaelson, MD, PhD, and William U. Shipley, MD

In the United States, the standard treatment of muscle- These older approaches included: (1) transurethral resec-
invasive transitional cell cancer of the bladder is radical tion alone done selectively for patients with small
cystoprostatectomy. This is an approach that results in tumors, which represented less than 20% of all muscle-
90% local control at 5 years but only 40% to 60% 5-year invading bladder tumors; (2) external beam radiation
overall survival. The usual cause of death is the result of therapy as monotherapy; (3) systemic multidrug
systemic spread of the primary tumor with the strong chemotherapy as monotherapy. Local control rates with
presumption that in the majority of cases micrometasta- radiation alone for muscle-invading tumors have been
tic disease is present at the time of cystectomy and the disappointingly low, and radiation as monotherapy has
patients so afflicted are therefore destined to die of dis- largely been abandoned.3–6
tant metastases. An analysis of cystectomy performed at It is now clear that monotherapies of various types are
Memorial Sloan Kettering Cancer Institute demon- in general unsuccessful in curing patients of invasive
strated a disease-specific survival of 67% with a median bladder cancer. Barnes et al.7 reported a 27% 5-year sur-
follow-up of 65 months and a median overall survival of vival in 85 patients with well-and moderately differenti-
only 45%.1 The clinical and pathologic stage of the dis- ated T2 transitional cell carcinomas (TCCs) treated with
ease is important in predicting long-term survival. The transurethral resection alone. Sweeney et al.8 found that
5-year recurrence-free survival for muscle-invasive blad- only 19% of patients with muscle-invasive tumors were
der cancer at the University of Southern California was selected for treatment by partial cystectomy, and these
89% in P2 node-negative tumors, 50% in P4 node-negative had a local recurrence rate of 38% to 78%. Hall et al.9
tumors, and 35% in patients with node-positive tumors.2 reported a 19% 3-year freedom from recurrence with
The effects of cystoprostatectomy on quality of life chemotherapy alone in 27 patients with localized disease,
have been carefully studied. The newest surgical tech- utilizing cisplatin, methotrexate, vinblastine, and epiru-
niques of nerve-sparing that may preserve male potency bicin. The local control rates for the monotherapies
and help increase the likelihood of continence with noted above are inadequate (Table 22-1) when compared
orthotopic urinary diversions have made cystectomy to radical cystectomy with its local control rate of 90%.
more tolerable, but even the most enthusiastic propo- Attempts at bladder sparing must be selective, as not all
nents of orthotopic bladder construction would agree patients are candidates for that approach. Favorable selec-
that a well-functioning natural bladder is superior in tion criteria include tumors that can be substantially
function to any of the technologic advances in bladder removed by transurethral resection and making certain
construction and reconstruction devised in the past that a complete response following initial chemoradiation
decade. induction is achieved, as measured by follow-up cytology
Present day combined modality treatment and selec- and cystoscopic biopsies. Only if there is a complete
tive bladder preservation by early response evaluation response of induction therapy, consolidation chemother-
differ from previous approaches utilizing monotherapy. apy is recommended and that followed by adjuvant

389
390 Part IV Bladder

Table 22-1 Bladder Sparing with Monotherapy The results of trimodality therapy for muscle-invasive
bladder cancer have led to further studies utilizing tri-
No. Recurrence
No. of an Invasive modality treatment with improved radiation techniques
Treatment Patients Tumor (%) and the use of newer chemotherapeutic agents in innova-
tive combinations in attempts to improve on the com-
Transurethral resection 331 2010 plete response rate and the long-term control rate in the
alone treatment of muscle-invasive bladder cancer.
Despite promising results there is reluctance among
Radiation alone 949 4011
urologic surgeons to accept trimodality therapy as an
Chemotherapy alone 27 199 alternative to cystectomy even in selected patients. In
part, this is due to improvements in urinary diversion
in patients undergoing cystectomy, but, more impor-
chemotherapy in those patients who have negative blad- tantly, concern persists among urologists that only cys-
der biopsies following consolidation radiochemotherapy. tectomy has the potential to rid the patient of the
If residual disease is found, cystectomy is recommended. disease permanently. Urologists have expressed the fol-
The combination of transurethral resection, radiation, lowing widely held views as arguments against bladder
and chemotherapy has yielded better results than any of sparing:
the monotherapies with an improved clinical complete
response rate (Tables 22-2 and 22-3). Substantial 1. Superficial relapse is common and very difficult to
improvements in local control have been reported with treat after radiochemotherapy.
combined modality therapy.12–14,19 This generally con- 2. Radiation burns the bladder rendering it useless,
sists of transurethral resection of the tumor for debulking with bleeding and scarring associated with frequency,
with the goal a visibly complete tumor removal followed urgency, nocturia, and perineal pain.
by radiation treatment with concurrent radiosensitizing 3. Innovative surgical techniques, including
chemotherapy. In the studies reported, the most com- neobladders, continent diversion, and other technical
monly used radiosensitizing drugs have been cisplatin, advances, have been successful and are subject to
5-fluorouracil (5-FU), and paclitaxel, used either singly continuing improvement, making bladder-sparing
or in various combinations. efforts unnecessary.

Table 22-2 Recent Results of Multimodality Treatment for Muscle-invasive Bladder Cancer
5-year 5-year Survival
Number of Overall with Intact Study
Multimodality Therapy Used Patients Survival (%) Bladder (%) Location References

External beam radiation + 42 52 42 RTOG 85-12 Tester et al.12


cisplatin

TURBT, external beam radiation 79 52 41 University of Dunst et al.13


+ cisplatin Erlangen

TURBT, MCV, external beam


radiation + cisplatin 91 51 44 (4 years) RTOG 88-02 Tester et al.14

TURBT, 5-FU, external beam 120 63 NA University of Housset et al.15,16


radiation + cisplatin Paris

TURBT, external beam radiation 162 (93-cisplatin; 55 44 University of Sauer et al.17


+ cisplatin or carboplatin 69-carboplatin) Erlangen

TURBT, ± MCV, external 123 49 38 RTOG 89-03 Shipley et al.18


beam radiation + cisplatin

TURBT, MCV, external beam 190 54 45 MGH Shipley et al.19


radiation + cisplatin

TURBT, external beam 123 49 NA RTOG 97-06 Hagan et al.20


radiation + cisplatin
Chapter 22 Selective Bladder Preservation by Combined Modality Treatment 391

Table 22-3 RTOG Bladder Protocols (1985–2001): Trimodality Therapy with Cystectomy for Poorly
Responding/Relapsing Patients
Protocol Induction Treatment Patients 5-year Survival Complete Response

85-12 TURBT, CP + XRT 42 52% 66%*

88-02 TURBT, MCV, CP + XRT 91 51% 75%*

89-03 TURBT, ± MCV then CP + XRT 123 49% 59%

95-06 TURBT, 5-FU plus CP + XRT 34 n.a. 67%

97-06 TURBT, CP + BID XRT adj. MCV 52 n.a. 74%

99-06 TURBT, TAX plus CP + XRT; adj. CP + GEM 84 n.a. n.a.

TURBT, transurethral resection of bladder tumor; XRT, external beam irradiation; CP, cisplatin; 5-FU, 5-fluorouracil; MCV, methotrexate,
cisplatin, vinblastine; TAX, paclitaxel; GEM, gemcitabine; n.a., not available.
*Urine or bladder wash cytology not included in the evaluation of the bladder tumor response.

4. Once the patient has been treated with der cancer (Tables 22-4 and 22-5). The list includes, in
chemotherapy and radiation therapy to the bladder, addition to cisplatin, methotrexate, vinblastine, ifos-
radical cystectomy becomes more difficult to perform famide, doxorubicin, gemcitabine, paclitaxel, and others.
and is associated with considerable morbidity. Not surprisingly, combinations of these drugs have been
5. Chemotherapy with its attendants nausea, anorexia, shown to be more effective than single agents in the per-
weight loss, and malaise is permanently destructive of centage of complete remissions (CR) achieved. With the
the quality of life. use of combination chemotherapy in advanced measurable
6. Delay in cystectomy risks patients’ lives. disease, CR became a common achievement compared to

Each of these concerns is dealt with in this chapter.


Table 22-4 Single Agent Activity in Bladder
In the past 15 years, the Radiation Therapy Oncology Cancer—Older Agents
Group (RTOG) has completed six prospective protocols
of combined modality therapy for patients with muscle- Drug Response Rate (%)
invasive cancer who are considered to be cystectomy can- Cisplatin 12–28
didates (see Table 22-3). Bladder preservation with
intravesical surgery, chemotherapy, and radiation therapy Methotrexate 29–45
are combined as initial treatment with radical cystectomy
recommended for all incomplete responders. Five of the Doxorubicin 17
RTOG protocols were phase I to phase II trials of con- 5-fluorouracil 15–17
current chemotherapy and radiation therapy and one
protocol was a phase III trial testing the efficacy of adju- Vinblastine 15
vant methotrexate, cisplatin, and vinblastine (MCV)
chemotherapy. A total of 426 patients were entered on Mitomycin C 13–20
these trials with 5-year overall survival of 50%, with
3-quarters of those patients being cured of their bladder
cancer, while maintaining a functioning bladder.21 Table 22-5 Single Agent Activity in Bladder
Cancer—Newer Agents
Current protocols are directed towards potentially more
effective chemotherapeutic regimens that will result in a Drug Response Rate (%)
high protocol compliance rate and possibly a higher
Paclitaxel 42–56
overall survival rate. A bladder-sparing strategy may be
offered to highly selected patients with the understand- Ifosfamide 20–31
ing that radical cystectomy is still an available option in
those patients who fail combined radiation therapy and Gemcitabine 23–28
chemotherapy.
Carboplatin 13–15
Investigators have become interested in clinical trials
with the demonstration that some older, as well as newer Docetaxel 13
drugs, have considerable activity against metastatic blad-
392 Part IV Bladder

a 15% to 20% rate of partial remission only utilizing a the question of neoadjuvant multidrug systemic
variety of single drugs, and with CR only rarely observed chemotherapy to reduce the appearance of distant metas-
with single-agent treatments. tases and increase cure rates. However, the 5-and 10-year
Successful bladder-sparing treatment is, and should be, metastasis-free survival rates were not influenced in any
very selective with frequent examinations of patients’ patient subgroup by the addition of two cycles of neoad-
bladders for signs of persistence or recurrence of disease. juvant MCV chemotherapy. The lack of efficacy of
Several protocols have been written explicitly directing neoadjuvant MCV was confirmed in a previously reported
discontinuation of the bladder-sparing effort in favor of phase III trial.18 We have not used neoadjuvant
radical cystectomy at the earliest sign of failure of local chemotherapy in subsequent protocols, directing our
control. All of the protocols have required for acceptance attention instead to adjuvant chemotherapy.
that patients be medically fit and willing to undergo cys- The MGH experience with 190 patients with invasive
tectomy immediately on recognition that the disease is bladder cancers with clinical stages T2–T4a entered on
found to be active in the bladder. Available data clearly successive prospective protocols has recently been
indicate that one-third of patients entering potential blad- updated.19 A common feature of all of the protocols was
der-sparing protocols require radical cystectomy either early bladder tumor response evaluation and the selec-
during or after the completion of radiochemotherapy. tion of patients for bladder conservation on the basis of
Over the years since the initial report of the effective- their initial response to transurethral resection of bladder
ness of cisplatin as a single agent in 1980 and the report tumor (TURBT) combined with chemotherapy and radi-
of cisplatinum given concurrently with radiation, it has ation therapy. Bladder conservation was reserved for
become appreciated that bladder cancer is a heteroge- those who had a complete clinical response at the mid-
neous disease, one with a rapid doubling time and with point in therapy (after a radiation dosage of 40 Gy).
micrometastases often present at the time of cystectomy. Approximately two-thirds of the total then received con-
It is important to review the evolution of the solidation with additional chemotherapy and radiation
Massachusetts General Hospital (MGH) and national therapy to a total tumor dose of 64 to 65 Gy. Incomplete
cooperative group approaches to bladder conservation. responders were advised to undergo radical cystectomy,
From 1981 to 1986, in a National Bladder Cancer Group as were patients whose invasive tumors persisted or
protocol, cisplatin was first used as a radiation sensitizer. recurred after treatment.
From 1986 to 1993, a phase II and phase III protocol uti- In these phase II and phase III protocols, the schedul-
lized MCV as neoadjuvant drugs. From 1994 to 1998, ing of the chemoradiation varied. In phase III study, mul-
twice daily (b.i.d.) radiation was introduced with concur- tidrug chemotherapy as neoadjuvant treatment was
rent cisplatin and 5-FU as radiation sensitizers and with evaluated compared without it and with all patients given
adjuvant MCV. From 1999 to 2002, b.i.d. radiation was TURBT plus concurrent cisplatin, and radiation therapy.
used, with cisplatin and paclitaxel as radiosensitizers and The median follow-up for all surviving patients was 6.7
with adjuvant cisplatin/gemcitabine. In 2003, a new pro- years with 81 patients having been followed for 5 years or
tocol was then opened, a randomized phase II program more and 28 patients for 10 or more years.19 The 5- and
utilizing b.i.d. radiation in all patients with randomiza- 1-year actuarial overall survival rates were 54% and 36%,
tion in the radiosensitizing drugs to either cisplatin plus respectively (stage T2 62% and 41%, stages T3–T4a 47%
5-FU or cisplatin/paclitaxel. The adjuvant regimen is to and 31%). The 5-and 10-year disease-specific survivals are
consist of cisplatin, gemcitabine, and paclitaxel, a three- 63% and 59%, respectively (stage T2 74% and 66%,
drug regimen proven effective in the treatment of stages T3–T4a, 53% and 52%). The 5-and 10-year dis-
advanced measurable bladder cancer.22 ease-specific survivals with an intact bladder are 46% and
The current eligibility criteria for bladder sparing 45%, respectively (stage T2, 57% and 50%, stages
include: (1) histologic proof of invasion of the muscularis T3–T4a 35% and 34%). The pelvic failure rate was 8.4%.
propria; (2) normal upper tracts; (3) the absence of No patient required cystectomy due to bladder morbidity.
hydronephrosis; (4) adequate renal function; (5) a normal The overall survival rate is provided in Figure 22-1,
hemogram; (6) medical fitness for cystectomy; and (7) the and the disease-specific survival rate is stratified by clini-
absence of malignant lymphadenopathy on imaging stud- cal stage in Figure 22-2. The current schema for multi-
ies utilizing CT or MRI, with biopsy as necessary. modality treatment of muscle-invasive bladder cancer is
Protocols carried out between 1986 and 2002 at provided in Figure 22-3, and the risk of relapse with a
the MGH Cancer Center included as complete a superficial tumor following multimodality treatment is
transurethral resection of the tumor as safely as possible shown in Figure 22-4.
and with the exception of one study, in which neoadjuvant The actuarial 5- and 10-year overall survivals and dis-
chemotherapy was employed, patients were treated fol- ease-specific survivals for all 190 patients and for some
lowing a transurethral resection with radiation and con- clinically important subgroups are shown in Table 22-6.
comitant chemotherapy.23 From 1986 to 1993, we studied The clinical stage (see Figures 22-1 and 22-2) signifi-
Chapter 22 Selective Bladder Preservation by Combined Modality Treatment 393

Overall survival versus 68%, p = 0.002. The 5- and 10-year disease-specific


survivals for all 66 patients undergoing cystectomy are
100 48% and 41%, respectively, indicating the important
contribution of this procedure in those patients treated
75 with this approach. The tumor stage did not influence
62% the 5- and 10-year disease-specific survivals in the 66
Stage T2 (n  90) patients who are undergoing either an immediate cystec-
50 tomy or a salvage cystectomy (stage T2, 57% and 39%,
47% stages T3–T4a, 42% and 42%).
Seventy-three (60% of 121) patients who were com-
25 p  0.02 Stage T3−4a (n  100) plete responders after induction therapy developed no
90 41 11 further bladder tumors. Twenty-four percent subse-
0 100 40 17 quently developed only a superficial occurrence and 16%
developed an invasive tumor.
0 1 2 3 4 5 6 7 8 9 10 Twenty-nine patients with superficial recurrence were
Time from protocol enrollment (years) managed conservatively by transurethral resection and
intravesical therapy. With a median follow-up of 4.1
Figure 22-1 Overall Survival. (From Shipley WU, Kaufman years since conservative treatment of the superficial
DS, Zehr E, Heney NM, Lane SC, Thakal HK, Althausen AF,
recurrence 18 (62%) have tumor-free bladders. Thus, 91
Zietman AL: Urology 2002; 60:62–68, with permission.)
of the 121 initially complete responding patients (or
75%) have tumor-free bladders. Seven of the 29 required
a subsequent cystectomy for further superficial or inva-
Disease-specific survival
sive recurrence. For these patients, the overall survival is
100 comparable to the 74 who had no failure, but one-third
of these patients required a salvage cystectomy.24
74 % Stage T2 (n  90) While there has been an understandable concern that
75 bladder-sparing treatment with high-dose radiation
53 % might cause irreparable damage to bladder function,
nearly all patients reported normal or near-normal func-
50
Stage T3−4a (n  100) tion and this is borne out by objective measurements of
bladder function, recently reported by Zietman et al.25
25 The quality of life in patients whose bladders have been
p  0.01
preserved has been studied in several centers with the
90 40 11
results summarized in Table 22-7. As reported by
0 100 40 17
Zietman et al. in the study of the long-term results of tri-
0 1 2 3 4 5 6 7 8 9 10 modality therapy for invasive bladder cancer, 78% of
patients had compliant bladders with normal capacity
Time from protocol enrollment (years)
and flow parameters. Eighty-five percent had no urinary
Figure 22-2 Disease-specific survival. (From Shipley WU, urgency or only occasional urgency. Twenty-five percent
Kaufman DS, Zehr E, et al: Urology 2002; 60:62–68, with had occasional to moderate bowel control symptoms and
permission.) 50% of men had preserved erectile function.
The MGH QOL questionnaire results of the late
cantly influences overall survival (p = 0.02) and disease- effects of chemoradiation on bladder function are very
specific survival (p = 0.01), as well as CR rate (stage T2, similar to two European cross-sectional studies recently
71%, stages T3–T4a, 50%, p = 0.04) and the rate of sub- reported. In both the studies, over 74% of the patients
sequent distant metastases (stage T2, 22%, stages reported good urinary function. In one of the studies all
T3–T4a, 37%, p = 0.03). We also found higher 5- and patients received concurrent chemotherapy and radiation
10-year disease-specific survivals with an intact bladder treatment as in the MGH series,26 while in the other,27
for stage T2 patients, 57% and 50%, respectively, than the patients received radiation alone. Concurrent
for stages T3–T4a patients, 35% and 24%, respectively, chemotherapy might, therefore, be inferred to add little
with a p value of 0.008. Neither the tumor grade nor the in terms of bladder morbidity.
use of neoadjuvant chemotherapy significantly improved Comparing our results to those of contemporary radi-
the CR rate (64%), overall survival, disease-specific sur- cal cystectomy series is confounded by the discordance
vival, or distant metastasis-free survival. The presence of between the clinical TURBT staging and pathologic (cys-
hydronephrosis significantly reduces the CR rate, 37% tectomy) staging. A recent prospective evaluation from
394 Part IV Bladder

Biopsy-proven muscle-invasive bladder cancer

Transurethral resection of all visible tumor, if possible

Induction therapy with external beam radiation and radiosensitizing chemotherapy (weeks 1 to 3)

Repeat cystoscopy with transurethral biopsy (week 7)

Complete response (T0) OR only a Any residual tumor at the original site OR a tumor
superficial tumor (Ta, Tcis) at a new site (T1 or greater) at a new site

Proceed with consolidation chemoradiation therapy (weeks 8 to 9) Radical cystectomy (week 9)

Repeat cystoscopy with transurethral biopsy (week 17) Adjuvant chemotherapy

Complete response Superficial persistence Muscle-invasive disease

Intravesical therapy or Radical cystectomy


radical cystectomy

Adjuvant chemotherapy

Adjuvant chemotherapy

Long-term cystoscopic surveillance

Figure 22-3 Current schema for multimodality treatment of muscle-invasive bladder cancer.
(From Michaelson MD, Zietman AL, Kaufman DS, Shipley WU: Invasive bladder cancer: the
role of bladder-preserving therapy and neoadjuvant chemotherapy. Urologia Integrada Y De
Investigacion (in press).
Chapter 22 Selective Bladder Preservation by Combined Modality Treatment 395

T2-4 TCC bladder


Trimodality therapy
n  190

Complete responders
n  121

No bladder Invasive
relapse relapse
n  73 n  16

Superficial
relapse
n  32

Cystectomy No treatment
n3 n1
Local therapy
n  28

No further relapse Invasive


n  18 n3
Further superficial cystectomy  3
n7
cystectomy  4

Figure 22-4 Outcome of 121 patients with clinical stages T2–T4 TCC of the bladder who
had a CR to trimodality therapy (chemotherapy, induction radiation, and transurethral
resection). (From Zietman AL, Grocela J, Kaufman DS, et al: Urology 2001; 58:380–385,
with permission.)

Stockholm28 has documented that clinical staging is more salvage cystectomy when necessary in the selective blad-
likely to understate the extent of the disease with regard der preservation series. The disease-specific survival rate
to penetration into muscularis propria or beyond than is of 40% at 10 years in the MGH series for patients having
pathologic staging. Thus, if any favorable outcome bias cystectomy underscores the need for close urologic fol-
exists, it is in favor of the pathologically reported radical low-up with cystoscopic surveillance and prompt bladder
cystectomy series. The University of Southern California removal when necessary.
recently reported on 633 patients undergoing radical cys- One of the concerns with bladder-sparing therapy is
tectomy with pathologic stages T2–T4a with an actuarial the risk of subsequent superficial relapses within the
overall survival rate at 5 years of 48% and 10 years of intact bladder, which could progress to life threatening
32%.2 The Memorial Sloan Kettering Cancer Center malignancy once again. Long-term follow-up from the
Contemporary Radical Cystectomy series showed that in MGH series has examined this issue in detail in 121
184 patients with tumors of pathologic stages P2–P4, the patients with a complete response (see Figure 22-4). Sixty
5-year overall survival rate was 36%. The actuarial 5-year percent of patients did not have evidence of relapse after
survival rate of all 269 patients undergoing radical cystec- a median follow-up of 7.1 years. Of the 32 superficial
tomy with pathologic stages ranging from P0 to P4 in this recurrences, 10 have required cystectomy and 18 have
series was 45%.1 been treated conservatively tumor-free bladders. Thus,
The results of these contemporary cystectomy series 91, or 75%, of the 121 complete responding patients
for muscle-invading bladder cancer are similar to the with a median follow-up of over 7 years have tumor-free
MGH series, as well as those from the University of bladders. The overall survival of those patients with a
Erlangen29 and the RTOG18 (Table 22-8). This similar- superficial recurrence is the same as those CR patients
ity in survival is likely in part due to the prompt use of without a bladder recurrence.24
396 Part IV Bladder

Table 22-6 Survival Outcomes by Patient and Tumor Characteristics


OVERALL SURVIVAL (%) DISEASE-SPECIFIC SURVIVAL (%)

Patient Group No. 5 year 10 year 5 year 10 year

All patients 190 54 ± 7.5* 36 ± 8.3* 63 ± 7.5* 59 ± 8.0*

Age at entry

<75 years 155 55 40 p = 0.04 65 60 n.s.

>75 years 35 51 22 56 56

Gender

Female 47 59 40 p = 0.67 60 52 p = 0.50

Male 143 52 34 64 62

Clinical Stage

T2 90 62 41 p = 0.02 74 66 p = 0.01

T3–T4a 100 47 31 53 52

Hydronephrosis

No 163 55 37 p = 0.15 64 61 p = 0.09

Yes 27 48 29 53 49

From Dunst J, Sauer R, Schrott KM, et al: Int J Radiol Oncol Biol Phys 1994; 30:261-266.
*95% confidence interval.

Table 22-7 Results of a Urodynamic/QOL Analysis gested that paclitaxel is an active agent in TCCs and a
Long-term results of trimodality therapy for invasive bladder phase II study of the combination of cisplatin and pacli-
cancer taxel demonstrated a 50% response rate in 52 patients
with metastatic disease.30 Three phase II trials demon-
1. 78% have compliant bladders with normal capacity and strated that gemcitabine combined with cisplatin is a
flow parameters well-tolerated active regimen.31–33 The combination of
cisplatin and gemcitabine has been compared to
2. 85% have no urgency or occasional urgency
methotrexate, vinblastine, doxorubicin, and cisplatin
3. 25% have occasional to moderate bowel control (MVAC) in a phase III study and the two combinations
symptoms were shown to have similar efficacy in metastatic disease.
The gemcitabine/cisplatin combination, however, was
4. 50% of men have normal erectile function better tolerated and led to fewer hospital days for the
treatment of toxic side effects.32 It is, therefore, consid-
ered by many to represent the new standard of care for
Thus, lifelong surveillance with cystoscopy is crucial metastatic TCC and is under investigation in the adju-
in patients treated with bladder-sparing therapy. vant setting as well.
Prompt salvage therapy for either superficial or recur- The latest national protocol for bladder-sparing treat-
rent invasive disease likely has prevented a survival ment (RTOG 02-33) was approved in January 2003. This
disadvantage. is a randomized phase II study comparing two combina-
In an attempt to improve safety, as well as to increase tions of radiosensitizing chemotherapy, each given con-
efficacy, newer studies of multimodality therapy, utilizing currently with a short course of b.i.d. radiation
newer chemotherapeutic agents, have recently shown a treatment. Patients received either the combination of
high degree of activity in metastatic transitional cell fluorouracil and cisplatin or paclitaxel and cisplatin. This
tumors. The major drugs in this category are gemc- is followed, in patients whose tumors are successfully
itabine and paclitaxel. Many recent studies have sug- controlled with chemotherapy and radiation, by a three-
Chapter 22 Selective Bladder Preservation by Combined Modality Treatment 397

Table 22-8 Invasive Bladder Cancer Survival Outcomes in


Contemporary Series
OVERALL SURVIVAL (%)

Series Stages Number 5-Year 10-Year

Cystectomy

USC2 (2001) P2–P4a 633 48 32

MSKCC1 (2001) P2–P4a 181 36 27

Selective bladder preservation

Erlangen29 (2002) cT2–T4 326 45 29

MGH19 (2001) cT2–T4a 190 54 36

RTOG18 (1998) cT2–T4a 123 49 —

drug adjuvant treatment program utilizing cisplatin, 2. Stein JP, Lieskovsky G, Cote R, et al: Radical
gemcitabine, and paclitaxel. This regimen has shown the cystectomy in the treatment of invasive bladder cancer:
highest response rate yet observed in published reports in long-term results in 1054 patients. J Clin Oncol 2001;
advanced measurable bladder cancer.22 19: 666–675.
3. Mameghan H, Fisher R, Mameghan J, et al: Analysis of
The 10-year overall survival and disease-specific sur-
failure following definitive radiotherapy for invasive
vival rates in our bladder-sparing protocols are compara-
transitional cell carcinoma of the bladder. Int J Radiat
ble to the results reported with contemporary radical Oncol Biol Phys 1995; 31:247–254.
cystectomy. For patients of similar clinical and patho- 4. Jenkins BJ, Blandy JP, Caulfield JF, et al: Reappraisal of
logic stage, one-third of patients treated on protocol with the role of radical radiotherapy and salvage cystectomy in
the goal of bladder sparing ultimately required a cystec- the treatment of invasive bladder cancer. Br J Urol 1988;
tomy. A trimodality approach with bladder preservation 62:343–346.
based on the initial tumor response is therefore safe and 5. Gospodarowicz MK, Hawkins MV, Rawling GA, et al:
appropriate treatment with the majority of long-term Radical radiotherapy for muscle invasive transitional cell
survivors retaining functional bladders. It is clear, how- carcinoma of the bladder: failure analysis. J Urol 1989;
ever, that lifelong bladder surveillance is essential 142:148–154.
6. DeNeve W, Lybeert ML, Goor C, et al: Radiotherapy
because only prompt salvage therapy can prevent a focus
for T2 and T3 carcinoma of the bladder: the influence
of new or recurrent bladder cancer from disseminating.
of overall treatment time. Radiol Oncol 1995;
We would conclude that selective bladder sparing 36:183–188.
should be one of the approaches considered in the treat- 7. Barnes RW, Dick AL, Hadley HL, et al: Survival
ment of invasive bladder cancer. While it is not sug- following transurethral resection of bladder carcinoma.
gested that it will replace radical cystectomy, sufficient Cancer Res 1977; 37:2895–2898.
data now exist from many international prospective 8. Sweeney P, Kursh ED, Resnick MI: Partial cystectomy.
studies to demonstrate that it represents a valid alterna- Urol Clin North Am 1992; 2(Suppl 2):75–87.
tive. This approach contributes significantly to the 9. Hall RR, Robert JT, Marsh MM: Radical transurethral
quality of life of patients so treated and represents a surgery in chemotherapy aiming at bladder preservation.
unique opportunity for urologic surgeons, radiation In Splinter AW, Scher HI (eds): Neoadjuvant
Chemotherapy in Invasive Bladder Cancer, pp 169–174.
oncologists, and medical oncologists to work hand in
New York, Wiley Liss, 1990.
hand in a joint effort to provide patients with the best
10. Herr HW: Conservative management of muscle-
treatment for this disease. infiltrating bladder cancer: prospective experience. J Urol
1987: 138:1162–1163.
11. Scher HI, Shipley WU, Herr HW: Cancer of the bladder.
REFERENCES In DeVita VT Jr, Hellman S, Rosenberg SA (eds): Cancer:
Principles and Practice of Oncology, 5th edition, pp
1. Dalbagni G, Genega E, Hashibe M, et al: Cystectomy for 1300–1318. Philadelphia ; JB Lippincott, 1997.
bladder cancer: a contemporary series. J Urol 2001; 12. Tester W, Porter A, Asbell S, et al: Combined modality
165:1111–1116. program with possible organ preservation for invasive
398 Part IV Bladder

bladder carcinoma: results of RTOG protocol 85-12. Int J combined pelvic irradiation plus cisplatin chemotherapy.
Radiat Oncol Biol Phys 1993; 25:783–790. J Urol 1990; 144:1128–1136.
13. Dunst J, Sauer R, Schrott KM, et al: Organ-sparing 24. Zietman AL, Grocela J, Kaufman DS, et al: Selective
treatment of advanced bladder cancer. A 10-year bladder conservation using transurethral resection,
experience. Int J Radiat Oncol Biol Phys 1994; chemotherapy and radiation: management and
30:261–266. consequences of Ta, T1 and Tis recurrence within the
14. Tester W, Porter A, Heaney J, et al: Neoadjuvant retained bladder. Urol 2001; 58:380–385.
combined modality therapy with possible organ 25. Zietman AL, Sacco UE, Skowronski E, et al: Organ
preservation for invasive bladder cancer. J Clin Oncol conservation as an alternative to radical cystectomy for
1996; 14:119–126. invasive bladder cancer: urodynamic and quality of life
15. Housset M, Maulard C, Chretien YC, et al: Combined evaluation of patients treated by trimodality therapy. Int J
radiation and chemotherapy for invasive transitional-cell Radiat Oncol Biol Phys 2002; 54(2 Suppl 1) 62.
carcinoma of the bladder: a prospective study. J Clin 26. Caffo O, Fellin G, Graffer U, et al: Assessment of quality
Oncol 1993; 11:2150. of life after cystectomy or conservative therapy for
16. Housset M, Dufour E, Maulard-Durtux C: Concomitant patients with infiltrating bladder carcinoma. Cancer 1996;
5-Fluorouracil, cisplatin and bifractionated split course 78:1089–1097.
radiation therapy for invasive bladder cancer. Proc Am 27. Henningsohn L, Wijkstrom H, Dickman PW, et al:
Soc Clin Oncol 1997; 16:319A. Distressful symptoms after radical radiotherapy for
17. Sauer R, Birkenhake S, Kuhn R, et al: Efficacy of urinary bladder cancer. Radiother Oncol 2002;
radiochemotherapy with plating derivatives compared to 60:215–225.
radiotherapy alone in organ-sparing treatment of bladder 28. Wijkstrom H, Norning U, Lagerkvist M, et al: Evaluation
cancer. Int J Radiat Oncol Biol Phys 1998; 40:121–127. of clinical staging before cystectomy transitional cell
18. Shipley WU, Winter KA, Kaufman DS, et al: A phase III bladder carcinoma, a long-term follow-up of 276
trial of neoadjuvant chemotherapy in patients with consecutive patients. Br J Urol 1998; 81:686–691.
invasive bladder cancer treated with selective bladder 29. Rodel C, Grabenbauer GG, Kuhn R, et al: Combined-
preservation by combined radiation therapy and modality treatment and selective organ preservation in
chemotherapy: initial results of RTOG 89-03. Clin Oncol invasive bladder cancer: long-term results. J Clin Oncol
1998; 16:357–383. 2002; 20(14):3061–3071.
19. Shipley WU, Kaufman DS, Zehr E, et al: Selective 30. Dreicer R, Manola J, Roth B, et al: Phase II study
bladder preservation by combined modality protocol of cisplatin and paclitaxel in advanced carcinoma
treatment: long-term outcomes of 190 patients with of the urothelium: an Eastern cooperative oncology
invasive bladder cancer. Urol 2002; 60:62–68. group (ECOG) study. Clin Oncol 2000;
20. Hagan MP, Winter KA, Kaufman DS, et al: RTOG 18:1056–1061.
97-06: initial report of a phase I/II trial of bladder 31. Kaufman DS, Raghavan D, Carducci M, et al: Phase II
conservation employing TURB, accelerated irradiation trial of gemcitabine plus cisplatin in patients with
sensitized with cisplatin followed by adjuvant MCV metastatic urothelial cancer. J Clin Oncol 2000;
chemotherapy. Int J Radiat Oncol Biol Phys 2001; 18:1921–1927.
51(Suppl 1):20A. 32. Von der Maase H, Hansen SW, Roberts JT, et al:
21. Shipley WU, Kaufman DS, Tester WJ, et al: Overview of Gemcitabine and cisplatin versus methotrexate,
bladder cancer trials in the radiation therapy oncology vinblastine, doxorubicin, and cisplatin in advanced or
group. In Raghavan D (ed): Cancer Supplement (in press). metastatic bladder cancer: results of a large, randomized,
22. Bellmunt J, Guillem V, Paz-Ares L, et al: Phases I and II multicenter, phase III study. J Clin Oncol 2000;
study of paclitaxel, cisplatin, and gemcitabine in advanced 18(17):3068–3077.
transitional-cell carcinoma of the urothelium. J Clin 33. Moore MJ, Winquist EW, Murray N, et al: Gemcitabine
Oncol 2000; 18(18):3247–3255. plus cisplatin, an active regimen in advanced urothelial
23. Prout GR Jr, Shipley WU, Kaufman DS, et al: cancer: a phase II trial of the National Cancer Institute of
Preliminary results in invasive bladder cancer with Canada Clinical Trials Group. J Clin Oncol 1999;
transurethral resection, neoadjuvant chemotherapy and 17(287):876–881.
C H A P T E R

23 Noncontinent and Continent


Cutaneous Urinary Diversion
Tracy M. Downs, MD, Maxwell V. Meng, MD,
and Peter R. Carroll, MD

Urinary diversion is necessary in patients who undergo describes permanent forms of cutaneous urinary diver-
cystectomy for bladder cancer, as well as in some patients sion, both noncontinent and continent. Temporary forms
with severe functional or anatomic abnormalities of the of urinary diversion, such as percutaneous nephrostomy,
lower urinary tract. Alternative urinary drainage may be are discussed elsewhere.
either temporary or permanent. Permanent forms of uri-
nary diversion can be accomplished by establishing direct
PREOPERATIVE PREPARATION
continuity between the urinary tract and the skin or,
more commonly, by interposing a segment of bowel Preoperative counseling is crucial. The discussion should
between the urinary tract and skin. Nearly all segments include the specific goals and complications of the
of bowel have been described for use in urinary diversion, planned procedure, as well as what the patient should
and despite extensive study, no single method or portion expect after surgery in terms of changes in lifestyle.
is ideal for all patients and clinical settings. The specific Noncontinent diversion may be the most appropriate
method of urinary diversion should be selected based on alternative in the debilitated patient and those who lack
the indication for diversion, individual patient prefer- the manual dexterity or motivation to care for a conti-
ence, anatomy, renal function, and overall health. nent reservoir. In addition, a thorough history should be
Urinary diversions can be broadly categorized into obtained, including previous surgical procedures, associ-
those that are continent (i.e., store urine with intermit- ated medical conditions, systemic diseases, and previous
tent drainage) and those that are noncontinent (i.e., con- irradiation. The patient should be specifically questioned
tinuous urine output). Continent forms of urinary regarding a history of regional enteritis, ulcerative colitis,
diversion are achieved with either a continence mecha- diverticulosis and diverticulitis, and other gastrointestinal
nism to an abdominal stoma or with an orthotopic blad- problems; this helps identify which segment of bowel can
der substitute relying on intrinsic continence after be used or avoided. Although the ileal conduit is most
anastomosis to the native urethra. Noncontinent types of popular, use of the colon has several advantages.
urinary diversion generally act simply as a conduit Antireflux mechanisms are possible using the ileocecal
through which the urine exits the body, thus requiring an valve or tenia, and the large lumen reduces the incidence
external appliance to collect the urine. Although conti- of stomal stenosis.
nence after reconstruction permits freedom from an An assessment of renal function is important and pre-
external collection bag, these operations may be techni- operative imaging of the upper urinary tract can reveal
cally more difficult and associated with higher complica- abnormalities, such as nephrolithiasis, hydronephrosis,
tion rates. Continent diversion, however, can benefit the and renal scarring. Additional evaluation of the bowel by
patient with regard to psychologic aspects, with pre- contrast imaging or endoscopy should be considered in
served self-image and sexuality. Overall, patients report a patients with a history of gastrointestinal diseases or signs
high level of satisfaction with both continent and non- and symptoms suggesting such diseases. The patient’s
continent forms of urinary diversion.1 This chapter overall state of health and fitness for surgery needs to be

399
400 Part IV Bladder

evaluated, especially in the older individual with bladder leak is provided by the volume of drain output and quan-
cancer and typical comorbidities. titation of fluid creatinine. In patients with a continent
It is essential to address issues of stomal care before cutaneous or orthotopic diversion, a Foley catheter is
surgery. Patients who are physically or mentally chal- used to drain the pouch. Manual irrigation of the catheter
lenged may need assistance from a family member or vis- (60 to 100 ml saline) is performed at 4- to 6-hour inter-
iting nurse. In general, the collecting device is changed vals to prevent mucus obstruction. While hospitalized,
every 4 to 5 days. The stoma site should be carefully patients become familiar with the catheterization and
selected prior to surgery, and consultation and subse- irrigation process. Plain radiography with instillation of
quent follow-up with an enterostomal therapist can be iodinated contrast into the pouch is performed prior to
valuable. The patient is evaluated in both the sitting and removing the Foley catheter to ensure watertight
standing position to ensure that the stoma is located away integrity. If a suprapubic tube is placed at the time of sur-
from bony prominences, fat folds, prior abdominal scars, gery, it can be removed after the contrast imaging study
and clothing waistbands (i.e., belts), all of which can or left as a safety valve, while the patient demonstrates
make it difficult for the external appliance to fit securely. adequate emptying by catheterization. At home, the
The usual site is located along a line from the anterior patient continues to irrigate the pouch 2 to 3 times per
superior iliac spine and the umbilicus, at the lateral edge day, helping eliminate mucus debris. Since continent
of the rectus abdominus muscle. It is important to bring reservoirs develop chronic bacteriuria, antibiotics should
the stoma through the rectus muscle to help prevent the only be given if the patient is symptomatic.
development of a parastomal hernia.
Adequate preoperative bowel preparation is important
NONCONTINENT URINARY DIVERSIONS:
for reducing infectious complications. Patients are typi-
SURGICAL TECHNIQUE
cally placed on a clear liquid diet 2 days prior to their
Urinary Diversion to the Abdominal Wall
scheduled procedure. A mechanical bowel prep (Go-
Cutaneous Pyelostomy
lytely) is started one day prior to surgery and oral antibi-
otics (neomycin + erythromycin) are given after Cutaneous pyelostomy was originally reported by
completion of the mechanical bowel prep. Systemic Immergut et al.2 as a method of diverting urine in chil-
antibiotics are administered during the perioperative dren with tortuous, dilated ureters. The relative lack of
period. subcutaneous fat, underdevelopment of the flank muscu-
lature, and renal mobility in children facilitate the oper-
ation.3 Although useful as a temporizing procedure in
POSTOPERATIVE CARE
children with gross infection, urinary obstruction, and
Several general principles of postoperative care apply especially in the setting of renal failure, current indica-
to the various types of urinary diversion. As with most tions are limited. The technique requires the presence of
intra-abdominal operations, paralytic ileus commonly an extrarenal pelvis. Pyelostomy is an alternative to per-
occurs after using intestinal segments for urinary diver- cutaneous nephrostomy in small infants, where place-
sion. Thus, a nasogastric tube is routinely placed for gas- ment of a nephrostomy tube can be difficult and
tric decompression and removed when bowel function long-term maintenance may pose a problem for parents.4
returns. More recently, the nasogastric tube is removed Technique. The renal pelvis is identified and mobi-
immediately after the operation with infrequent need for lized through a subcostal incision. There is no need to
replacement and improved patient’s comfort. In order to mobilize the ureter or disrupt its collateral circulation.
minimize antibiotic-associated complications, prophylac- The kidney is rotated anteriorly, and a 3-cm incision is
tic antibiotics should be continued for only 24 hours after made in the renal pelvis well away from the ureteropelvic
surgery. Fluid balance should be closely monitored and junction (Figure 23-1A). The renal pelvis is brought out
postoperative fluid replacement should be calculated to the skin incision and anastomosed to the skin using
from maintenance requirements, third-space loss, drain interrupted absorbable sutures (Figure 23-1B). The
output, and systemic conditions leading to hypermetabo- remainder of the incision is closed in the usual fashion.
lism. Serum electrolytes are checked for imbalances.
Patients undergoing pelvic surgery are at risk for venous
Cutaneous Ureterostomy
thromboembolic events, which can be reduced with the
use of sequential compression devices and early ambula- The first cutaneous ureterostomy was performed by
tion. Incentive spirometry can improve pulmonary func- LeDentu in 1889 and represents a simple method of uri-
tion and lessen respiratory complications. If ureteral nary diversion.5 End cutaneous ureterostomy has been
stents are used, they are typically removed on the fifth used as a means of temporary diversion in infants and pal-
postoperative day and after 3 weeks for an ileal conduit liative diversion in adults6,7; delayed reconstruction after
and continent diversion, respectively. Evidence of urinary cutaneous ureterostomy can be successfully performed.8
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 401

Figure 23-1 A, The renal pelvis is incised for approximately 3 cm. B, The renal pelvis is
anastomosed to the skin using interrupted sutures.

The advantages of cutaneous ureterostomy include If only a single ureter is obstructed, a simple stoma can
decreased operative and recuperative times, as well as the be created by sewing the end of the ureter flush with the
use of an extraperitoneal approach.9 In addition, compli- skin at the stoma site. Another option, when the ureter is
cations associated with the use of bowel, such as intestinal narrow, is to create a V-flap stoma (Figure 23-2A–C). If
anastomosis and absorption of urinary constituents, are both ureters are dilated, a single stoma can be created by
avoided.10 Disadvantages of cutaneous ureterostomy suturing the ureters together, everting them and anasto-
include the risk of stomal stenosis, chronic bacteriuria, mosing the ends to the skin. Alternatively, a Z-plasty can
and stone formation. be performed when the ureters are not adequately dilated
A dilated ureter is usually necessary for a successful (Figure 23-3A–C).12 When a single ureter is dilated,
outcome, facilitated by the thick-walled, well-vascular- transureterostomy with retroperitoneal passage of the
ized ureter.11 If both ureters are dilated and have ade- smaller ureter to the contralateral side is combined with
quate length, each can be brought out to the skin. If only the cutaneous ureterostomy. Both ureters need to be free
one ureter is dilated, this should be used for the cuta- from angulation and tension. The larger ureter is incised
neous ureterostomy with proximal transureteroureteros- 2 cm on its medial aspect, and the smaller ureter is
tomy of the contralateral, nondilated ureter.8,12 trimmed to the appropriate length and spatulated. The
Technique. When both ureters are dilated, a double- ureteral–ureteral anastomosis is performed using a run-
barreled cutaneous ureterostomy is fashioned with the ning absorbable suture; ureteral stents may be placed
stoma on the side of the shorter ureter. In the case of prior to this portion. The stoma is fashioned as previ-
unilateral ureteral dilation, the stoma is created on that ously described for a single ureter.
side. Only a single stoma site, with one appliance, is
required whether one or both ureters are brought to the
Conduits to the Abdominal Wall
skin.
Ileal Conduit
The ureter is mobilized extraperitoneally with careful
preservation of the collateral blood supply. If possible, Seiffert13 first described an ureteroileocutaneous diver-
the ureter should be freed to the level of the bladder to sion in 1935. The procedure was subsequently popular-
maximize length. Prior to transecting the ureter, the ized by Bricker14 and remains the most commonly used
ureter is measured to ensure that it is adequate. method of noncontinent urinary diversion in the United
402 Part IV Bladder

Figure 23-2 A, The ureter is brought through the skin and spatulated. B, C, The Ureter is
everted and the anastomosis is performed using several interrupted absorbable sutures.

Figure 23-3 A–C, After making a Z-shaped incision, the ureters are spatulated on their
lateral aspects and sutured to the skin.
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 403

States. Patient selection, proper stoma location, and devices. The ileal segment is brought below the level of
careful technique in creating a well-vascularized stoma the ileoileal reanastomosis. The conduit is positioned cau-
are all important to the success of the procedure. dally, usually with the end in the right lower quadrant in
An ileal conduit may be contraindicated in patients an isoperistaltic direction; the base of the conduit is closed
with a history of regional enteritis or extensive pelvic with either absorbable sutures or a stapler (Figure 23-4B).
irradiation.15 In these situations, a colon conduit may be The left ureter is brought under the sigmoid mesocolon
preferable. The typical stoma is located in the right lower and both ureters are spatulated and reimplanted into the
quadrant, as previously described. In order to minimize base of the conduit. The various types of ureteral anasto-
the absorptive surface of the bowel in contact with urine, moses will be discussed later. Ureteral stents are placed
the ileal segment should be as short as feasible. and brought out via the abdominal stoma to facilitate uri-
Reabsorption of urinary constituents does not cause a nary drainage and stent the anastomoses. The mesentery
significant problem in patients with normal renal func- is reapproximated using nonabsorbable sutures to prevent
tion but metabolic abnormalities may develop in those internal bowel herniation.
with renal insufficiency.16 To create the stoma, a small circle of skin is excised at
Technique. The conduit is usually constructed using a the premarked site, and the underlying cylinder of fat
segment of ileum approximately 15 cm proximal to the is removed. A cruciate incision is made in the anterior
ileocecal valve. The exact length of the conduit varies fascia, large enough to accommodate two fingers (Figure
with patient habitus but averages between 15 and 20 cm 23-5A). The stents and the end of the conduit are brought
in most patients (Figure 23-4A). Preservation of the vas- through the rectus abdominus muscle and fascia; the end
cular supply to the ileal segment is vital. Transillumination of the conduit is then secured to the fascia (Figure 23-5B).
of the mesentery helps identify the blood vessels supply- If present, the distal staple line is excised and the stoma is
ing the bowel, and at least two major vascular arcades everted to create a “rosebud” (Figure 23-5C). The stoma
should be included with the ileal segment. Once the should ideally protrude, without tension, 2 to 3 cm above
appropriate area of ileum is identified and isolated, the the surface of the skin. In patients with short mesentery or
mesentery is divided proximally and distally with individ- thick abdominal wall, a Turnbull loop can be created to
ual ligation of vessels. The bowel is divided between maximize stomal vascularity.17 When properly con-
clamps or using a GIA stapler. Bowel continuity is structed, both end- and loop-cutaneous stomas have com-
reestablished by suturing or with the aid of stapling parable long-term results in patients with ileal conduits.18

Figure 23-4 A, An appropriate segment of ileum is selected, containing two major vascular
arcades. B, The conduit is positioned in the right lower quadrant and the ureters are
reimplanted into the base of the conduit.
404 Part IV Bladder

Figure 23-5 A, The rectus fascia is incised in a cruciate fashion. B, The end of the conduit
is brought through the rectus muscle and anchored to the fascia. C, The stoma is everted
and sutured to the skin.

preserve an adequate blood supply to the segment. The


Jejunal Conduit
jejunum is divided between either clamps or the GIA sta-
Jejunal conduit urinary diversion should be reserved for pler and then continuity is restored. In contrast to the
patients in whom other, more suitable options are ileal conduit, the isolated jejunum should lie above the
unavailable. These include those with prior pelvic radia- reanastomosed jejunum. The proximal end of the con-
tion, inflammatory bowel disease, or loss of the middle duit is directed towards the retroperitoneum and the
and distal ureters. Electrolyte disturbances are common conduit is oriented in an isoperistaltic direction. The
in those with urinary conduits using the jejunum.19 ureters are anastomosed to the jejunum, with placement
Technique. As with other bowel segments incorpo- of stents to reduce early postoperative electrolyte abnor-
rated into the urinary tract, the length of jejunum should malities.15 The mesenteric window is closed using non-
be as short as possible to reduce metabolic abnormalities; absorbable sutures. The stoma is created in the same
in some cases, the stoma can be placed above the umbili- fashion as described for an ileal conduit; the stoma is usu-
cus. The site, distant from the belt line, should be care- ally located in the right upper quadrant when using
fully considered and marked before surgery. jejunum.
Patients who have received significant pelvic radiation
may require transection of the ureters at the level of the
Colonic Conduits
true pelvis (pelvic inlet). The ureters are brought out
from the retroperitoneum below the ligament of Treitz.20 The colon conduit was popularized by Turner-
An appropriate segment of jejunum is identified and iso- Warwick21 in 1960. Urinary conduits constructed from
lated, as described for ileal conduits. It is important to large intestine have several potential advantages when
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 405

compared to those made using small intestine. First, non- flux mechanism depends on the competency of the ileo-
refluxing ureteral anastomoses are possible using either a cecal valve, which can be reinforced by wrapping the
short tunnel through the teniae coli or the ileocecal redundant cecum around the ileum and securing this
valve. Second, stomal stenosis is less common due to the configuration with seromuscular sutures.22 The proximal
larger lumen of the colon. Third, a suitable segment of portion of the conduit is fixed to the retroperitoneum,
colon outside of the field of previous abdominal or pelvic and the distal cecal portion is brought out to the skin. An
irradiation is usually available. When use of the colon is everted stoma is created in the standard rosebud fashion.
anticipated, a preoperative contrast imaging study of the
large bowel or colonoscopy should be considered.
Transverse Colon Conduit
The transverse colon conduit is well suited for patients
Ileocecal Conduit
who have received extensive pelvic irradiation, making
The ileocecal conduit was first described by Zinman and the small bowel suboptimal, or situations where the dis-
Libertino22 in 1975. This method utilizes the ileocecal tal ureters are damaged or absent. In situations of inade-
valve as an antireflux mechanism for the ureters. Other quate ureteral length, the transverse colon segment can
advantages of this portion of bowel include the constant be directly anastomosed to both renal pelves.
and abundant blood supply and location in the right Technique. A 15-cm segment of transverse colon is
lower quadrant, facilitating stoma formation.3 The ileo- used for the conduit. The blood supply is based on the
cecal segment is rarely affected by generalized bowel dis- middle colic artery, and transillumination of the trans-
orders, such as diverticulitis. verse mesocolon assists in identifying the appropriate
Technique. The blood supply of the ileocecal region segment (Figure 23-6A). The greater omentum is dis-
is based on the ileocolic artery. The segment, including sected from the superior aspect of the transverse colon
10 cm of terminal ileum, is isolated and the appendix is and the mesentery is incised, with ligation of individual
removed. After restoring continuity between the ileum blood vessels. The colon is divided both proximally
and ascending colon, the ureters are anastomosed and distally, and bowel continuity is restored. The iso-
directly into the ileal portion of the segment. The antire- lated transverse colonic segment should lie below the

Figure 23-6 A, An appropriate segment of transverse colon is selected. B, After


colocolostomy, the ureters are brought into the abdominal cavity and the ureteral
anastomoses are performed.
406 Part IV Bladder

reanastomosed transverse colon. Mobilization of the


RECTAL BLADDER URINARY DIVERSION
right colon can ensure that the colocolostomy is created
without tension. The proximal end of the colon conduit The rectal bladder urinary diversions allow excretion of
is closed and fixed posteriorly. The ureters are mobi- urine my means of evacuation. Innovative techniques
lized and then the ends are spatulated and brought have been advocated for separating the fecal and urinary
through the posterior peritoneum into the abdominal streams, but all still employ the principle of ureterosig-
cavity. The ureters are reimplanted into the base of the moidostomy. Variations of the original ureterosigmoi-
conduit in either a direct or nonrefluxing manner and dostomy include the ureter ileosigmoidostomy, folded
ureteral stents are placed (Figure 23-6B). A flap of peri- rectosigmoid bladder, and augmented valve rectum pro-
toneum is fashioned to cover the ureteral anastomoses. cedures, in which different segments of small intestine
A standard rosebud technique is used to create the are anastomosed to the rectum. These operations will be
stoma, which can be positioned on either side in the grouped and discussed together as “rectal bladder urinary
upper or lower quadrant. diversions.”
In each of these operations, with the exception of cer-
tain types of augmented valve rectum procedures, the
Sigmoid Colon Conduit
ureters are transplanted to the rectal stump and the prox-
The sigmoid colon conduit should be avoided in patients imal colon is managed by terminal sigmoid colostomy or,
with a history of pelvic irradiation. In these cases, the more commonly, by bringing the sigmoid to the per-
superior hemorrhoidal artery is often injured, leaving an ineum, using the anal sphincter to achieve both bowel
inadequate blood supply to the sigmoid segment. A sig- and urinary control. Although these operations continue
moid conduit is also questionable in those undergoing to enjoy some popularity abroad, they have never been
cystectomy because the vascular supply to the rectum widely accepted in the United States.
may be compromised if the internal pudendal artery is
ligated. However, the sigmoid can be used after pelvic
Ureterosigmoidostomy
exenteration, since it obviates the need for intestinal
reanastomosis. A sigmoid colon conduit is commonly Ureterosigmoidostomy can be regarded as the original
used for the management of benign conditions of the continent urinary diversion with reports of this procedure
lower urinary tract, such as neurogenic bladder, requiring dating as far back as the 1850s.23,24 Its appeal to the uro-
urinary diversion. logic surgeon has waned given issues of transplanting the
Technique. The sigmoid colon is mobilized by lateral ureters directly to an intact fecal stream and development
incision along the white line of Toldt; adequate mobiliza- of malignancy, and considerable complications, including
tion makes the bowel reanastomosis easier. The optimal hyperchloremic acidosis, hypokalemia with nephropathy,
segment is identified, typically 10 to 12 cm in length. and pyelonephritis. Nevertheless, ureterosigmoidostomy
After short incisions in the mesentery are made, the prox- offers a simple form of continent urinary diversion when
imal and distal ends of the colonic segment are divided. compared with the newer techniques requiring complex
The superior hemorrhoidal artery can be divided for refashioning of bowel segments.25,26 Currently, most
increased mobility. Bowel continuity is reestablished, with urologists reserve the procedure only for those individu-
the conduit placed laterally to the anastomosis. The right als of advanced age where adverse long-term sequelae are
ureter is brought under the sigmoid colon, and the ureters less important. Several other factors argue against the use
are sewn using either a direct or nonrefluxing technique. of ureterosigmoidostomy. In patients with neurogenic
The preferred stoma site, created as previously described, bladder, there may be associated bowel or anal sphincter
is on the left side of the abdomen. dysfunction. Individuals with dilated ureters are at
increased risk for developing either obstruction or reflux,
while extensive prior radiation may affect both the bowel
CONTINENT URINARY DIVERSIONS: SURGICAL
and distal ureters. Patients with preexisting renal insuffi-
TECHNIQUE
ciency are poor candidates for ureterosigmoidostomy or
Continent cutaneous (i.e., nonorthotopic) techniques can any continent diversion27,28; similarly, those with underly-
be divided into two major categories: (1) urinary diver- ing hepatic dysfunction are at risk for developing ammo-
sion into the rectosigmoid and (2) continent urinary nia intoxication.29 Preoperative evaluation of the patient
pouches requiring clean intermittent catheterization. should include barium enema and/or colonoscopy that
Because numerous variants of continent urinary diver- would detect bowel disease, such as diverticulitis, polyps,
sion are used worldwide, a complete review of all opera- or cancer.
tive techniques is beyond the scope of this chapter. This Incontinence of the mixture of stool and urine is a
chapter focuses on the most commonly utilized and calamitous complication. Thus, an adequate test of pre-
described continent urinary diversions. operative anal sphincter integrity is mandatory and anal
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 407

sphincteric tone must be judged competent before one


selects this procedure. Various tests have been proposed
to measure sphincter function. The most useful are those
that require the retention of an enema solution of both
solid and liquid material for a specified time in the
upright and ambulating positions without soilage.
Bissada et al.26 use a saline retention enema that the
patient holds for 1 hour, while Benson et al.15 advocate
the use of a 400- to 500-ml thin mixture of oatmeal and
water that the patient attempts to retain for 1 hour in the
upright position.28

Technique
Figure 23-7 Leadbetter-Clarke ureterointestinal anastomosis.
If indicated, cystectomy is performed first. At the start of A linear incision is made in the taenia, the taenia is raised and
urinary diversion part of the procedure, a large-caliber the mucosa is identified. A small button of mucosa is
(24Fr to 28Fr) red rubber catheter is placed in the rec- removed and the ureter is spatulated and then sutured to the
tum, advanced through the anus and sutured in place to mucosa with 5-0 PDS. The sero-muscular layer is sutured
the perianal skin. This tube should be advanced suffi- over the ureter with care taken not to compromise or occlude
ciently far that it can be easily reached and remains in the ureter.
place after surgery to drain urine and potentially to
decrease the risk of anastomotic leakage. inner layer is closed with a running suture of absorbable
A permanent end colostomy is created by dividing the material taken through all layers of intestine, whereas the
descending sigmoid colon at the junction of its middle outer layer is closed with seromuscular nonabsorbable
and lower thirds with complete closure of the distal rec- material. Although intubation of the ureters and stent
tosigmoid segment with absorbable sutures, rather than placement are feasible using the Leadbetter technique of
staples, to reduce the potential for postoperative calculus ureteral implantation, the transcolonic approach (anterior
formation in the rectosigmoid reservoir. The proximal colotomy) (see Figure 23-8B) affords greater ease in
sigmoid colon is diverted to a premarked end colostomy directing the ureteral stents to the outside.
stoma site. The ureterocolonic anastomoses can be retroperi-
The most distal portion of the rectosigmoid is the pre- tonealized. In addition, it is often helpful to suture the
ferred site for the ureteral anastomosis. In the Leadbetter proximal end of the isolated rectosigmoid to the sacral
technique,30 the left ureter is brought through the sig- promontory to prevent excessive mobility. One or 2
moid mesentery. After both ureters have been mobilized, intraabdominal drains are typically placed, as well as a
stay sutures are placed proximally and distally in an rectal tube.
antimesenteric tenia 10 to 15 cm apart and a longitudinal
incision of 5 to 7 cm is made. The distal end of the ureter
CONTINENT CATHETERIZABLE URINARY
is spatulated and a mucosal to mucosal anastomosis is
DIVERSIONS
performed with fine absorbable suture material (Figure
23-7). The muscularis of the tenia is then closed over the Numerous operative techniques have been developed for
ureter with interrupted absorbable sutures to form the continent catheterizable urinary diversions. Patient
submucosal tunnel. The contralateral anastomosis is selection is important in deciding which patients are
made in the same fashion; generally, the right ureter is most suitable for a continent catheterizable urinary
anastomosed most distally. diversion. To perform clean intermittent catheterization,
Goodwin and colleagues30a published their experience it is paramount that one has good hand-eye coordination.
with the transcolonic (open colon technique) of ureteral Thus, patients with spinal cord injury (e.g., quadriplegia)
anastomosis (Figure 23-8A–D). This technique differs or some with multiple sclerosis or other neurologic dis-
from Leadbetter’s method in that the rectosigmoid is orders may not be candidates. Patient with any cognitive
opened anteriorly and the ureters are brought through a condition, such as dementia, that could lead to a poor
separate submucosal tunnel in the posterior wall into the understanding of the catheterization process would not
bowel lumen. The ureters are sutured, separately or con- be an appropriate candidate.
joined (i.e., Wallace technique), to the mucosa from within The choice of where to place the catheterizable stoma
the bowel lumen. After ureteral anastomosis and ureteral is based on the preference of the surgeon. The two most
stent placement have been completed, the colon is closed common sites are at the umbilicus and in the lower quad-
with a simple 2-layer technique (see Figure 23-8C ). The rant of the abdomen, through the rectus bulge and below
408 Part IV Bladder

Figure 23-8 A, The open colostomy approach is preferred because it allows the placement
of indwelling ureteral stents bilaterally. B, The stents are directed to the outside through the
lumen of the rectal tube. C, The colon is closed with a simple two-layer technique. The
retroperitoneal windows are closed with a running suture.
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 409

Figure 23-8 cont’d D, The ureteral colonic anastomoses are retroperitonealized if possible.
(From Walsh P, Retik A, Vaughn EJ, et al (eds): Campbell’s Urology, 8th ed, Philadelphia,
WB Saunders, 2002.)

the “bikini line.” The latter site affords both men and which avoids the need for intussusception; (4) the
women the opportunity to conceal the stoma. In certain hydraulic valve (e.g., Benchekroun nipple).31
patients the umbilicus is the preferred location. For Two techniques of appendiceal continence mecha-
example, in the individual confined to a wheelchair, the nisms have been reported. Mitrofanoff43 reported excis-
umbilical stoma location may have a lower incidence of ing the appendix with a button of cecum and reversing it
stomal stenosis, especially when fashioning an appen- on itself before tunneled reimplantation.44 Alternatively,
diceal stoma. The umbilical location is also ideal in Riedmiller et al.45 have left the appendix attached to the
paraplegics because it allows easier catheterization cecum and buried it into the adjacent taenia by rolling it
without the need for chair transfer and disrobing. back onto itself. A wide tunnel is created in the taenia
Cosmetically, the umbilical location of a stoma is typi- extending 5 to 6 cm from the base of the appendix
cally indistinguishable from a normal umbilical dimple. (Figure 23-9A,B) and windows are created in the
Patients are instructed to cover the stomal site with a mesoappendix between blood vessels. The appendix is
gauze or square adhesive bandage to avoid mucus soil- folded cephalad into the tunnel, and seromuscular
ing of clothing. sutures are placed through the mesoappendix windows to
complete the tunneling. The tip of the appendix is ampu-
tated and brought to the selected stomal site.
CONTINENCE MECHANISM
For right colon pouches, an appendiceal tunneling
Perhaps the single most demanding technical aspect dur- technique is the simplest to perform. The in situ or trans-
ing creation of a continent urinary diversion is construc- posed appendix is tunneled into the cecal taenia, similar to
tion of the continence mechanism. It is the success or that of the previously described ureterocolonic anastomo-
failure of this mechanism that ultimately determines the sis. Appendiceal continence mechanisms have been criti-
success or failure of the urinary diversion. Four surgical cized for three reasons. First, the appendix may be
techniques have been incorporated to create a reliable, unavailable in some patients because of prior appendec-
catheterizable continence mechanism: (1) appendiceal; tomy. Second, the appendiceal stump may be too short to
(2) pseudoappendiceal tubes fashioned from ileum or reach the anterior abdominal wall or umbilicus, while still
right colon; (3) intussuscepted nipple valve or flap valve, maintaining sufficient length for tunneling. Third, the
410 Part IV Bladder

large amount of mucus produced by an intestinal reser- the mesentery that otherwise induces eversion and
voir is more easily emptied or irrigated with a larger effacement of the intussusception. A second major mod-
catheter (20 Fr to 22 Fr) rather than the typical smaller ification has been the attachment of the nipple valve to
diameter catheter (14 Fr or 16 Fr) that is accommodated the reservoir wall itself. This has been achieved by 2 or 3
through an appendiceal stump. Several of these criticisms different stapling techniques, as well as by a suturing
have been addressed by modifying surgical technique. In technique described by Hendren38 and King.39,40 Despite
individuals who had a prior appendectomy, a pseudoap- meticulous surgical technique and the most experienced
pendiceal tube can be fashioned from ileum32 or from the surgeons, nipple valve failure can be anticipated in 10%
wall of the right colon.33 In cases where the appendiceal to 15% of cases. In addition to being subject to slippage,
stump is too short to reach the anterior abdominal wall or nipple valves may develop ischemic atrophy; if this
umbilicus, Burns and Mitchell34 lengthen the appendiceal occurs, a new nipple valve must be fashioned from a new
stump by including a tubular portion of the proximal bowel segment.
cecum. This has the added advantage of allowing a Several institutions have introduced numerous modi-
slightly larger stoma made of cecum that is less prone to fications of the original Kock technique for constructing
stomal stenosis. Overall, the appendiceal or pseudoappen- a nipple valve given the disappointing long-term stability.
diceal continence mechanism remains a very attractive The group at the University of Southern California has
and reliable continence mechanism. developed the T pouch using a flap valve.41 The proce-
The second major type of continence mechanism used dure is much simpler, avoiding the need for intussuscep-
in right colon pouches is the tapered and/or imbricated tion, and is used to create both a continence mechanism
terminal ileum and ileocecal valve. The technique is and an antireflux mechanism.
rather simple, with imbrication or plication of the ileoce- Benchekroun42 describes a hydraulic valve nipple. In
cal valve region along with tapering of the more proximal this procedure, a small bowel segment is isolated and a
ileum in the fashion of a neourethra35–37 and afford a reli- reversed intussusception is carried out, apposing the
able continence mechanism. mucosal surfaces of the small bowel. Tacking sutures are
The creation of intussuscepted nipple valves is the taken to a portion of the circumference of the intussus-
most technically demanding of continence mechanisms ception to stabilize the nipple valve, while allowing urine
and also is associated with the highest rates of complica- to flow freely between the leaves of apposed ileal mucosa.
tion and reoperation. The procedure has a significant The Benchekroun hydraulic ileal valve may be the one
learning curve and, therefore, should probably not be diversion in which reservoir detubularization actually
chosen by the surgeon who infrequently constructs con- impairs the continence rate because the valve mechanism
tinent pouches. depends on pouch pressure for continence. Without ele-
A major advance in nipple valve construction includes vated pouch pressure, there is no forceful apposition of
removal of mesenteric attachments from the middle 6 to the serosal leaves of the valve. Although there are reports
8 cm of bowel,38 thereby reducing the tethering effect of in the literature of reasonable success, concerns regard-

Figure 23-9 A and B, The appendix is left attached to the cecum and buried into the
adjacent cecal taenia by rolling it back onto itself. A wide tunnel is created that extends 5 to
6 cm from the base of the appendix. Windows are created in the mesoappendix between
blood vessels. The appendix is folded cephalad into the tunnel, and seromuscular sutures
are placed through the mesoappendix.
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 411

ing the long-term durability of this mechanism have remaining 40 to 50 cm of the bowel segment are
resulted in lack of widespread use. brought together as a U-shaped plate, with the apex
directed to the proposed stoma site. The U-shaped
plate is opened using electrocautery and an absorbable
INTRAOPERATIVE POUCH TESTING
suture is used to close the posterior wall of the opened
Following pouch construction and completion of the intestinal plate (Figure 23-10A–E). The proximal and
continence mechanism, it is essential that both the pouch distal valves are then created by intussuscepting
and continence mechanism be examined in detail in the approximately 10 cm of ileum at each end; the proxi-
operating room. Typically, the pouch is filled with saline mal 10 cm serves as the valve and the distal 5 to 10 cm
or water, the catheter is removed, and the pouch is com- the patch (Figure 23-10A). The middle 6 to 8 cm of
pressed lightly as the surgeon examines the pouch for any the 10-cm proximal intussuscepted segment is
points of leakage, as well as observing the ability of the denuded of mesentery by electrocoagulation on each
pouch to contain urine. Next, the pouch is recatheterized side flush with the serosa of the ileum. Allis or
to ensure smooth passage of catheter passage. This is a Babcock clamps are passed through the open limbs,
critical maneuver because the inability to catheterize is a and intussuscepted nipple valves are created by pulling
serious complication that often results in the need for back the intraluminal bowel wall into the opening of
reoperation. the reservoir to be created (Figure 10B). In order to
In general, all redundancy should be removed from maintain the intussusception, 3 to 4 parallel rows of
the continence mechanism. It is often useful to secure the staples (TA-55, 4.8-mm staples) are placed along the
reservoir to the anterior abdominal wall in a manner that entire length of the nipple valves (Figure 23-10C). To
allows the reservoir to surround the aditus, preventing minimize the risk of stone formation, the distal 6 sta-
the development of a false passage or a kink and thereby ples from each cartridge are removed before staple
facilitating catheterization. application to ensure that the tip of the valve is free of
the staples.
The nipple valve is then fixed by 1 or 2 stapling
CONTINENT RESERVOIRS
techniques to the back wall of the patch.39 A small but-
Continent Ileocecal Reservoirs
tonhole may be made in the back wall of the ileal plate
Kock Pouch
so that the anvil of the stapler can be passed through
In 1982, Kock et al.46 reported the first 12 cases on the buttonhole and advanced into the nipple valve
their continent ileocecal reservoir, stimulating world- before application of the fourth row of staples (Figure
wide interest in continent urinary diversion proce- 23-10D). If this is done, the buttonhole is oversewn
dures. However, the initial enthusiasm with this with absorbable material. Alternatively, the anvil of
technique is tempered by a complication rate requiring the stapler can be directed between the two leaves of the
additional surgery in half of patients, primarily for mal- intussuscipiens and the fourth row of staples used to fix
function of the efferent valve. Since then, the Kock the inner leaf of the nipple valve to the pouch wall
pouch has undergone several modifications. Skinner et (Figure 23-10E).
al.47,48 have carefully studied and improved the tech- Skinner advocates the use of an absorbable polygly-
nique over the years. Although some individuals still cotic acid (PGA) mesh collar to anchor the base of the
perform the original procedure, technical difficulties nipple valve to prevent late slippage or extussusception.
and significant complication rates have led to the pro- A 2.5-cm wide strip of absorbable mesh is fashioned into
cedure being abandoned by most urologic surgeons. a collar and passed through a separate window in the
This operation and the similarly constructed T pouch mesentery and sewn with seromuscular stitches to
are the only catheterizable continent diversions that the bowel wall to secure the intussuscepted nipple from
preserve the ileocecal valve. the outside (Figure 23-10F,G). The mesh of the efferent
Technique. An 80-cm small bowel segment is iso- nipple can also be fixed to the abdominal wall to facili-
lated approximately 15 to 50 cm proximal to the ileo- tate catheterization. The ureters are spatulated and end-
cecal valve. A stapling device is used to isolate the to-side, mucosa-to-mucosa anastomoses are performed
segment of small bowel and the continuity of the small to the free portion of the afferent nipple; each ureteral
bowel is reestablished with closure of the mesenteric anastomosis is stented. The reservoir is then folded and
window to prevent incarceration of the bowel. Division closed with a single or double layer continuous
of the mesentery should extend to the base of the absorbable suture. The abdominal stoma is created by
mesentery to ensure sufficient mobility of the efferent suturing the efferent nipple valve to the rectus fascia at
limb of the pouch. Next, the proximal and distal 15 to the predetermined site and a flush stoma is created. A
20 cm of the isolated segment are preserved for cre- Foley catheter is passed through the efferent limb and
ation of the afferent and efferent nipple valves, and the left inside the reservoir. Posteriorly the pouch is
412 Part IV Bladder

Figure 23-10 A, A 15-cm segment of terminal ileum is isolated and opened along its
antimesenteric wall. The proximal 10 cm will serve as the continent intussusception and the
distal 5 to 10 cm as the patch. The size of the patch varies according to the size of the
excised segment. B, An Allis clamp or Babcock clamp is advanced into the ileal terminus,
the full thickness of the intussuscipiens is grasped and it is prolapsed into the pouch. C,
Three rows of 4.8-mm staples are applied to the intussuscepted nipple valve using the TA-
55 stapler. D, A small buttonhole is made in the back wall of the ileal plate to allow the anvil
of the staples to be passed through and advanced into the nipple valve. E, The anvil of the
stapler can be directed between the two leaves of the intussuscipiens and the fourth row of
staples applied in this manner. This figure shows two valve mechanisms. In this instance,
there would be only one.
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 413

Figure 23-10 cont’d F, A 2.5-cm-wide strip of absorbable mesh is placed through


additional windows of Deaver at the base of each nipple valve the mesh strips are fashioned
into collars. G, The collars are sewn to the base of the pouch, as well as to the ileal
terminus with seromuscular sutures.

attached to the sacral promontory with interrupted segments can be of various lengths, depending on ureteral
sutures. length and the thickness of the anterior abdominal wall.
The middle 44 cm of ileum is folded into a W, with each
limb measuring 11 cm (Figure 23-11B). The afferent
T Pouch
antireflux mechanism is created by opening the windows
The group at the University of Southern California has of Deaver between the mesenteric vascular arcades along
devised a novel continence mechanism created entirely the distal 3 to 4 cm. The efferent continence mechanism
from ileum, based on a technique described by Abol- is created by opening the proximal 7 to 8 cm of vascular
Enein and Ghoneim49,50 and Bochner et al.51 Abol- arcades (antiperistaltic) (Figure 23-11C). Quarter-inch
Enein and Ghoneim created an extramural serosal Penrose drains are then placed in each window of Deaver
tunnel into which the ureters were implanted, with the to facilitate the passage of the 3-0 silk horizontal mattress
extramural trough creating a pseudotunnel that pre- sutures that are used to approximate the serosa of the
vented reflux with, theoretically, a lower risk of obstruc- corresponding 11-cm limbs of the W (Figure 23-11D).
tion than either of the techniques of direct transmural The 3- to 4-cm anchored portion of the proximal limb is
ureteral implantation described by Goodwin, Leadbetter, then tapered over a 30Fr catheter and the 7- to 8-cm
or LeDuc et al.52 Stein et al.41 first reported on the use anchored portion of the efferent limb is tapered over a
of a tapered ileal segment implanted into a serosal 16Fr catheter. In both portions, tapering is performed
trough as the antireflux mechanism for neobladder.53 with a GIA stapler. In the efferent limb, care must be
Stein et al.54 presented the early experience with a taken to create a gradual taper so that the catheter does
double T pouch as a replacement for the Kock pouch, not hit a false cul-de-sac (Figure 23-11E). The portions
detailing the technique in 9 patients (2 men and 7 of the 11-cm W-limbs not forming the troughs are
women) followed a mean time of 12 months. sutured together with a running suture of 3-0 PGA. The
Technique. A 70-cm segment of terminal ileum is bowel is now incised along its antimesenteric border in
isolated 15 to 20 cm from the ileocecal valve. The proxi- the portion where the serosal trough exists and in close
mal isoperistaltic 10- to 12-cm segment is isolated and proximity to the medial PGA suture lines when beyond
serves as the antireflux mechanism, while the distal 12- to the two limbs (Figure 23-11F). The incised mucosa is
15-cm segment is isolated and rotated in an antiperi- then closed in 2 layers with a running suture of 3-0 PGA.
staltic fashion and becomes the cutaneous continence The incised intestinal flaps (antimesenteric incision)
mechanism (Figure 23-11A,B). A short (2 to 3 cm) are then sutured to each ostium with interrupted
mesenteric incision is made to isolate the proximal limb sutures of 3-0 PGA and the 2 ileal flaps sutured over
(afferent antireflux limb), and a 4-cm incision is made for each segment with a running suture of 3-0 PGA
the distal limb (efferent continence limb), thereby pre- (Figure 23-11G). The reservoir is closed side-to-side in
serving the major vascular arches. The proximal and distal 2 layers with 3-0 PGA, completing its construction
414 Part IV Bladder

Figure 23-11 A, 70-cm segment of terminal ileum is isolated 15 to 20 cm from the


ileocecal valve. B, Proximal 10-cm segment is isolated and rotated toward the reservoir in
an antiperistaltic direction. C, The windows of Deaver are opened to allow the walls of the W
reservoir to be apposed behind the valve mechanisms. Penrose drains are passed to guide
suture passage.
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 415

Figure 23-11 Cont’d D, Some 3-0 silk horizontal mattress sutures are passed through
each window. The distal continence mechanism is longer than the proximal antireflux
mechanism. E, The proximal and distal mechanisms are tapered with a metal GIA stapler. F,
The bowel is incised along its antimesenteric border where it will overlie the two Ts. Distal to
the Ts, the bowel is incised closed to the approximated limbs of the reservoir. G, The ostia
of the valves are secured to the bowel wall with interrupted absorbable sutures. The two
flaps of ileum are closed over the Ts with running absorbable sutures.
416 Part IV Bladder

Figure 23-11 Cont’d H, The back wall of the reservoir is closed with running absorbable
sutures. I, The lateral walls are folded medially and the construction completed with running
absorbable sutures

(Figure 23-11H,I). The ureters are anastomosed end- cal valve as a means of further stabilizing the intussus-
to-side over stents to the proximal limb, which has ception.59 The technique described herein includes
been closed with a running absorbable Parker-Kerr these modifications rather than the earlier versions of
suture. The efferent limb is brought to the abdominal the operation.
wall stoma site, the redundant ileum resected, and the Technique. A 10- to 15-cm segment of cecum and
stoma is matured. The reservoir lies immediately adja- ascending colon is isolated, along with two equal-sized
cent to the anterior abdominal wall. loops of terminal ileum and an additional portion of
This procedure appears to have many advantages over ileum measuring 20 cm for the creation of the reservoir
the Kock pouch, but long term complications and dura- and the continence mechanism, respectively (Figure
bility of this technique cannot be assessed at this time 23-12A). The entire colonic segment and distal seg-
because of small patient numbers and short follow-up. ment of ileum are split open along the antimesenteric
border, with care taken to preserve the ileocecal valve.
These three bowel segments are folded in the form of
Mainz Pouch
an incomplete W, and the posterior aspects sutured
In 1986, Thüroff et al.55 presented initial results with a together to form a broad posterior plate (Figure 23-12B).
cecoileal reservoir (Mainz I pouch). A sutured ileoileal The ileal mesentery of the intact (nondetubularized)
intussusception valve served as the continence mecha- proximal ileum is freed of its mesentery for a distance
nism in the first series of patients. Subsequently, the of 6 to 8 cm and intussusception of the segment is
Mainz pouch has undergone considerable modification achieved. The ileum is intussuscepted and stabilized
over the years primarily due to problems with the with 2 rows of staples (TA 55 staples) (Figure 23-12C).
intussuscepted nipple valve.56–58 The operation has Next, the intussuscepted ileum is pulled through the
now been modified by replacing sutures with staples to natural tunnel of the intact ileocecal valve and fixed to
prevent deintussusception and using the intact ileoce- the ileocecal valve with two more rows of staples. The
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 417

third row of staples is applied to stabilize the nipple the urodynamic and continence rates for the continent
valve to the ileocecal valve (Figure 23-12D) and the cutaneous reservoirs from various published series.
fourth row of staples is applied inferiorly, securing the
inner leaf of the intussusception to the ileal wall
(Figure 23-12E). Indiana Pouch
Both ureters are implanted by means of a 5-cm long One of the first continent cutaneous pouches to gain
submucosal tunnel at the uppermost part of the cecal wide acceptance in the urologic community was the
portion of the reservoir. Next, the reservoir is folded on Indiana pouch developed in 1985 by Rowland et al.88
itself in a side-to-side fashion and closed with a single The major contribution was the creation of a reliable
layer of continuous sutures to complete the pouch con- continence mechanism. Originally, the entire ileal seg-
struction. A suprapubic tube is placed through a separate ment was buttressed with imbricating sutures to establish
stab incision in the pouch and abdominal wall to ensure the continence mechanism. Pressure profile studies
adequate drainage. The entire pouch is rotated cephalad demonstrated that the continence zone was confined to
and the intussuscepted ileum is brought to the umbilicus. the region of the ileocecal valve; therefore, imbricating
A small button of skin is removed from the depth of the sutures were only necessary in the region of the ileocecal
umbilicus and the ileal terminus is directed through this valve.37
buttonhole (Figure 23-12F). The reservoir is secured to
the posterior fascia with interrupted absorbable sutures, Technique. In the present form of an Indiana pouch,
and the intussuscepted ileal limb is sewn similarly to the 20 to 25 cm of cecum and ascending colon are isolated
anterior fascia. Extra ileal length is resected, and the along with 10 to 15 cm of terminal ileum (Figure 23-
ileum is sutured to the umbilicus with interrupted 13A). After bowel continuity is reestablished, appendec-
absorbable sutures. Although the cutaneous stoma can be tomy is performed and the appendiceal fat pad obscuring
placed in several different locations, the preferred stoma the inferior margin of the ileocecal junction is removed
site by the Mainz group is the umbilicus. (Figure 23-13B). The entire right colon is detubularized
The introduction of the more reliable appendiceal along its antimesenteric border about three-fourths of
continence mechanism has greatly increased the accept- its length starting from the distal end, and the cecum is
ance of the Mainz I procedure. Starting in 1988, the in partially spatulated. The ureters are anastomosed
situ appendix was used for creation of the continence through a submucosal tunnel along the posterior colonic
mechanism.60 The advantages of this modification are tenia.
reducing of the operative time by 30 to 40 minutes, a The continence mechanism consists of plicated termi-
shorter ileal segment, eliminating nipple sliding or pro- nal ileum, which reinforces the ileocecal valve. The ileal
lapse, and reduced risk of stone formation with the plication, consisting of 2 rows of Lembert sutures 8 to 10
absence of staples. The Mainz group has also developed mm apart, begins at the ileocecal valve and extends 3 to 4
2 new techniques for construction of a Mitrofanoff cm proximally (Figure 23-13C); the continence mecha-
(appendiceal) type of tube for use in patients whose nism is tested and when no leakage is present, a second
appendix is either unsuitable or absent.33,61,62 layer of continuous silk sutures over a catheter (12Fr or
14Fr) is used to reinforce the suture line. Alternatively, the
University of Miami group uses purse-string sutures in the
Continent Right Colon Reservoirs with
same region of the ileum.37 In the Florida pouch, apposing
Intussuscepted Terminal Ileum
Lembert sutures are applied on each side of the terminal
Similar to the Mainz pouch, these right colon pouches ileum (Figure 23-13D). Any excess ileum is tapered over a
use the nipple valve concept for continence by using the catheter and removed using a GIA stapler. It is important
ileocecal valve. Unlike the Mainz pouch, the right colon to imbricate while the cecal reservoir is still open, allowing
is used solely for the construction of the pouch portion of one to observe the gradual closure of the ileocecal valve.
the diversion. Several right colon pouches using the valve The pouch is formed by folding down the opened cecum
technology have been described—Indiana pouch, UCLA in a Heineke-Mikulicz configuration and closing with con-
pouch, Duke pouch, LeBag pouch, Florida pouch, and tinuous 2-0 PGA suture. Ureteral stents are placed and a
the Penn pouch.63–65 These operations differ from one suprapubic tube is brought through the lower abdominal
another by only minor features, predominantly related to wall. A 1-cm buttonhole is made in the skin, anterior and
the technique employed for stabilizing the nipple valve posterior fascia. The catheterizable ileal limb is advanced
and the amount of large intestine harvested (Table 23-1). between the rectus muscles. Excess ileum is removed and
In this section, we discuss in detail only the Indiana and a flush skin stoma is created.
Penn pouches. In all instances, appendectomy is per- At Indiana, as well as other institutions, it was recog-
formed because the in situ appendix would serve as a nized that by only marsupializing a portion of the ascend-
nidus for infection and abscess formation. Table 23-2 lists ing colon, the cecal region maintained sufficient
418 Part IV Bladder

Figure 23-12 A, A 10- to 15-cm portion of cecum and ascending colon is isolated along
with two separate equal-sized limbs of distal ileum and an additional portion of ileum
measuring 20 cm. B, A portion of the intact proximal ileal terminus is freed of its mesentery
for a distance of 6 to 8 cm.

peristaltic integrity to generate pressures high enough to rate,89 such as the use of absorbable staples to fashion the
overcome the continence mechanism. Variations on the reservoir. First introduced by Bejany and Politano,37 a
Indiana pouch, including the Florida pouch,36 harvest the GIA stapler with metal staples can facilitate fashioning of
entire right colon and one-third to one-half of the trans- the efferent limb.
verse colon to create the reservoir. The University of
Miami pouch37 retained the original Indiana pouch
design but marsupialized the entire ascending colon and Penn Pouch
cecum and refashioned the pouch in a Heineke-Mikulicz This ileocecal pouch, described by Duckett and Snyder44
configuration. in 1986, was the first continent diversion employing the
Additional modifications to the Indiana reservoir Mitrofanoff principle, wherein the appendix served as the
allow more rapid construction with a lower complication continence mechanism.
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 419

Figure 23-12 cont’d C, The intact ileum is intussuscepted and two rows of staples are taken
on the intussuscipiens itself. D, The intussuscipiens is led through the intact ileocecal valve
and a third row of staples is taken to stabilize the nipple valve to the ileocecal valve. E, A fourth
row of staples is taken inferiorly, securing the inner leaf of the intussusception to the ileal wall.
420 Part IV Bladder

Figure 23-12 cont’d F, A button of skin is removed from the depth of the umbilical funnel
and the ileal terminus is directed through this buttonhole. Excess ileal length is resected and
ileum is sutured at the depth of the umbilical funnel.

Table 23-1 Comparison of Techniques of the Various Forms of Continent Right Colon Pouches
with Intussuscepted Terminal Ileum
Pouch Year Reservoir Continence Mechanism Ureteral Anastomosis

UCLA Pouch* 199463 Entire right colon Terminal ileum is tapered over a 14Fr LeDuc (tunneling
+ 15 cm distal ileum Foley using a GIA stapler technique)
Ileal limb (nipple) is fixed against the
wall of the continent reservoir with
continuous sutures
Duke Pouch* 198764 Entire right colon Ileum is intussuscepted through the Tunneling technique
+ 15 cm distal ileum ileocecal valve
Ileal limb (nipple) is stabilized with
sutures or staples
A patch of mucosa is excised from the
posterior cecal plate
One entire thickness of the intussuscipiens
is incised to reveal the inner leaf of the
intussuscipiens (serosa)
Florida Pouch† 198736 Entire right colon Similar to Indiana Pouch (exception: End-to-side (colonic
+ transverse colon opposing Lembert sutures on each side segment) nonrefluxing
(right 1/3 or 1/2) + 10 of the terminal ileum to buttress the
to 12 cm distal ileum ileocecal valve
LeBag* 198665 20 cm right colon Standard nipple valve from Goodwin technique (similar
+ 20 cm distal ileum intussuscepted ileum to ureterosigmoidostomy)
Staples are used to stabilize the
nipple valve
No mesh collar for nipple valve
stabilization

*The long-term outcome of this pouch is unknown.


†Ureteral obstruction occurred in 4.9% of patients with nontunneled ureterocolonic anastomosis; hyperchloremia in 70% of patients (only 4

patients required treatment for this electrolyte imbalance).


Table 23-2 Comparison of Continent Cutaneous Reservoirs: Reservoir Volume, Urodynamic Maximum Fill Pressures,
and Continence Rates

RESERVOIR CONTINENCE
No.
Pouch References Patients Volume (ml) Pressure (cm H2O) Overall Daytime Nighttime

Kock pouch Kock et al.66 12 500 NR NR NR NR

T pouch Stein et al.67 9 400–700 NR 100% NR NR

Mainz pouch Thuroff et al.68 100 510–620 23–33 at half capacity NR NR NR


31–41 at full capacity
Lampel et al.69 440 NR NR 90% stapled
ileocecal
intussusception
Gerharz et al.70 193 NR NR 100%

UCLA pouch* deKernion et al.71 19 560 (mean) NR 71% NR 69%


Raz63 NR 600–700 NR 88.2% NR NR

Indiana pouch Rowland et al.72 29 291–508 ≤20 intraluminal NR 93 76


Rowland et al.73 29 291–508 ≤20 intraluminal NR 93 76
Scheidler et al.74 91 NR NR 93% NR NR
Carroll et al.75 14 400–1500 24 (mean) reservoir 85.7% NR NR
675 (mean) 40 (mean) plicated ileal segment
Ahlering et al.76 70 400–800 15–25 resting pressure 98.6% NR NR
Carroll et al.77 21 NR 41 (mean) plicated ileum 85.7 NR NR
Rowland et al.78† 81 NR NR NR 98% 98%
63 (mean) stapled ileum
Bihrle et al.79‡ 50 500 (mean) NR NR 94% 94%
733 (maximum)

Continued
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 421
422
Part IV Bladder

Table 23-2 Comparison of Continent Cutaneous Reservoirs—cont’d

RESERVOIR CONTINENCE
No.
Pouch References Patients Volume (ml) Pressure (cm H2O) Overall Daytime Nighttime

Florida pouch Lockhardt et al.80 11§ 400–1200 28–55 at capacity 88% NR NR


650 (mean)
Lockhardt et al.81 107 550–1200 10–58 at capacity 97.5% NR NR
747 (mean) 35 (mean)

Duke pouch* Webster et al.82 7 500–700 50 (near capacity) 42.8%¶ NR NR

U. Miami pouch Bejany et al.83 10 750–800 20 at capacity (pouch) 100% NR NR


20–30 tapered ileum (empty)
50–60 tapered ileum (at capacity)
Benson et al.84 75 ≥750 20 at capacity 98.6% NR NR

Penn pouch Sumfest et al.85 NR NR NR 96% NR NR

Gastric pouch# Adams et al.86 3 245 35 end filling pressure NR NR NR


Carr et al.87 12 309 12.9 100 NR NR

*The long-term outcome of these pouches is unknown.


†The last 81 patients operated on by Rowland and associates underwent construction of a stapled efferent limb; In the last 20 patients, the reservoir was created with absorbable

staples.
‡Mean follow-up in this study was 2.5 years.
§8/11 patients had a continent colonic reservoir performed.
¶One patient suffered incontinence due to evagination of the intussusception; three patients had intermittent incontinence secondary to cecal segment hyperactivity. This was

controlled with oxybutynin hydrochloride 5 to 10 mg TID.


#Of the 12 patients, seven had urinary reservoirs totally constructed from stomach, and five had composite reservoirs.

NR, not reported.


Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 423

Figure 23-13 A, Segment of terminal ileum approximately 10 cm in length along with the
entire right colon is isolated. B, Appendectomy is performed and the appendiceal fat pad
obscuring the inferior margin of the ileocecal junction is removed by cautery. C, Interrupted
Lembert sutures are taken over a short distance (3 to 4 cm) in two rows for the double
imbrication of the ileocecal valve as described at Indiana University. D, Application of
opposing Lembert sutures on each side of the terminal ileum.
424 Part IV Bladder

Technique. A segment of cecum up to the junction of mesoappendix between blood vessels. The appendix is
the ileocolic and middle colic arteries is isolated, in addi- folded cephalad into the tunnel and seromuscular sutures
tion to a similar length of terminal ileum. The isolated are placed through the mesoappendix windows to com-
bowel is marsupialized on the antimesenteric borders and plete the tunneling. The tip of the appendix is amputated
sutured to one another. The superior margin of the pouch and brought to the selected stomal site.
is sutured transversely using absorbable material.
Mitrofanoff technique. A button of cecum surrounding
Gastric Pouches
the origin of the appendix is circumscribed and the
resulting cecal aperture closed with running absorbable The stomach has many unique properties91 and can be
suture. The mesentery of the appendix is dissected care- used in selected circumstances. Advantages to using
fully from the base of the cecum, preserving the blood stomach as a urinary reservoir include diminished elec-
supply. The appendix is then reversed on itself, so that trolyte reabsorption, inherent barrier to absorption of
the cecal button can reach the anterior abdominal wall ammonium, absence of hyperchloremic acidosis, and
and the tip of the appendix can be directed to the taenia reduced bacterial colonization in the acidic environment.
of the colon (Figure 23-14). The appendiceal tip is tran- Situations favoring consideration of the gastric uri-
sected obliquely and spatulated prior to a tunneled nary diversion include extensive irradiation of the lower
appendiceal-taenial implantation. If additional appen- bowel, preexisting metabolic acidosis or renal insuffi-
diceal length is required, the variation proposed by Burns ciency, and patients in whom shortening the small bowel
and Mitchell34 may be employed (cecal tube). could result in malabsorption. Experience with gastric
An alternative technique can be applied when fashion- pouches and composite reservoirs has been reported in
ing the continent catheterizable stoma. Riedmiller et al.45 both the pediatric90 and adult populations.91,92
leave the appendix attached to the cecum and bury it into Technique. A 7- to 10-cm wedge-shaped segment is
the adjacent taenia by rolling it back onto itself. A wide isolated from the greater curvature of the stomach, typi-
tunnel is created in the taenia extending 5 to 6 cm from cally using a 70- to 90-mm GIA stapler. The incision is
the base of the appendix. Windows are created in the not extended to the lesser curvature because of the risk of
vagal injury and resulting problems with gastric empty-
ing. Before the gastric wedge is taken, the decision has to
be made as to which gastroepiploic artery will serve as a
pedicle for the gastric flap. The segment is preferentially
mobilized on the left gastroepiploic vascular stalk by
dividing the short gastric vessels proximal to the segment
up to the gastric fundus (Figure 23-15A,B). If the left
artery cannot be used, then the right gastroepiploic ves-
sel is used and the small gastric vessels are ligated to the
level of the pylorus. The stomach is then closed based on
surgeon’s preference.
A gastroduodenostomy or gastrojejunostomy tube is not
mandatory unless the antrum of the stomach has been used.
The gastric wedge is brought through the transverse meso-
colon and the root of the small bowel to lie in a retroperi-
toneal position. The isolated wedge is refashioned into
nearly a sphere by folding it back on itself and suturing the
edges together with running absorbable material. Before
closing the pouch, the ureters are reimplanted using the
submucosal tunneling technique (Goodwin).
A continent catheterizable reservoir can be con-
structed using appendix or the distal ureter tunneled sub-
mucosally into either the dome or posterior portion,
respectively. If the distal ureter is used to create the con-
Figure 23-14 A segment of cecum up to the junction of the
ileocolic and middle colic blood supplies along with a similar
tinence mechanism, a proximal transureteroureteros-
length of terminal ileum is isolated and marsupialized on the tomy can be performed. The free portion of the ureter
antimesenteric borders. A button of cecum surrounding the can then be brought to the skin or to the introitus
origin of the appendix is circumcised. The mesentery of the (female) or urethral stump (male) to serve as a catheteri-
appendix is dissected carefully from the base of the cecum, zation portal. If neither option is available, a gastric tube
preserving its blood supply. and nipple can be created from a gastric flap.
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 425

Figure 23-15 A and B, A wedge-shaped segment of stomach whose greatest width is 7 to


10 cm is fashioned from the greater curvature. The left gastroepiploic artery is preferentially
used as the blood supply for the isolated gastric wedge by dividing the short gastric vessels
up to the gastric fundus. Alternatively, if there is a problem with the left artery, the right
gastroepiploic vessel may be employed.

Alternatively, the wedge of stomach can be incorporated fluxing anastomoses are easier to perform on the colon
into a reservoir composed of detubularized ileum with or because of the presence of teniae coli. Numerous tech-
without tapering (Figure 23-16A–C). When stomach is niques of ureterointestinal anastomoses have been
used as a bladder augment or as a portion of a neoblad- described and we present several common methods.52,97,98
der, the dysuria and hematuria syndrome has been
reported, limiting use in sensate patients.93 Ureteral-Small Bowel Anastomoses
The construction of reservoirs entirely from stomach Direct Anastomosis
has not seen widespread acceptance. Rather, use of stom-
ach segments has been limited to bladder augmentation Bricker,14 in his description of ureteroileocutaneous
or as part of a composite reservoir.94,95 While the use of diversion, used a freely refluxing, direct end-to-side
stomach has particular appeal in the pediatric popula- ureteral anastomosis. It remains the simplest method of
tion,95 its various unique intrinsic properties as a reser- connecting the ureter to the ileum. After spatulation of
voir suggest that its use will continue in only selected the ureters, a small ellipse of bowel serosa and mucosa is
clinical situations. excised from the site of implantation. The spatulated
ureter is anastomosed directly to the bowel, with care
taken to incorporate both the muscular and mucosal ele-
URETEROINTESTINAL ANASTOMOSES ments of the intestine (Figure 23-17A,B). Only small
Ureterointestinal anastomoses can be performed in either portions of the mucosa should be included to ensure that
a refluxing (direct) or nonrefluxing fashion. Theoretically, the mucosa everts and results in precise mucosa-to-
a nonrefluxing anastomosis protects the upper urinary mucosa apposition. In contrast, if too much mucosa is
tract from infected urine and may decrease renal deterio- included in the anastomosis, this can narrow the lumen of
ration. This aspect, however, remains controversial. the ureterointestinal anastomosis. The anastomosis can
Reflux alone does not necessarily lead to upper urinary be performed in 1 or 2 layers using interrupted
tract deterioration and is related to factors, such as infec- absorbable sutures. Prior to completing the anastomosis,
tion, stones, and obstruction. Patients with nonrefluxing the ureter may be stented, especially if the patient is at
anastomoses have not been shown to have lower rates of risk for impaired wound healing.
bacterial colonization.96 There is no long-term evidence
that patients without reflux have a lower rate of renal dys- Wallace Technique
function. In addition, nonrefluxing techniques of ureteral
anastomosis are associated with higher stricture rates. In this method, the ureters are joined together prior to
Both types of ureteral anastomoses can be performed performing the ureterointestinal anastomosis99; the cre-
into either the small or large bowel. In general, nonre- ation of a single, larger urinary segment may decrease the
426 Part IV Bladder

Figure 23-16 A, An 11-cm long segment of stomach is isolated on the right gastroepiploic
blood supply with the use of a GIA-90 stapler. Usually, two staplers are required. B, A 22-
cm segment of ileum is then isolated, opened along its antimesenteric border and
refashioned in a U shape.
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 427

Figure 23-16 cont’d C, The edges of the stomach are then sutured to edges of the ileum
with a running absorbable suture of 2-0 PGA.

Figure 23-17 A and B, After removing a small ellipse of bowel, the ureter is spatulated and
anastomosed directly to the bowel wall using interrupted absorbable sutures.
428 Part IV Bladder

risk of subsequent ureteroenteric stenosis. Each ureter is lumen, a small ellipse of mucosa is excised. The ureter
spatulated for a distance approximately equal to the is anastomosed to the bowel mucosa (Figure 23-19B)
diameter of the conduit. The ureters are joined together and then the muscular layer of the tenia coli is reap-
using a running absorbable suture and then anastomosed proximated to create a tunnel over the ureter (Figure
to the open proximal end of the bowel in a refluxing 23-19C).
manner (Figure 23-18A,B). The anastomosis can be per-
formed in 1 or 2 layers with absorbable sutures. The
COMPLICATIONS
ureters may be stented.
General Complications
Numerous complications may occur after all types of uri-
Tunneled Ureterointestinal Anastomosis
nary diversion. Problems, both early and delayed, can
In this method, the ureter is brought through a submu- results from surgical technique, the incorporation of
cosal tunnel, creating a nonrefluxing anastomosis to the intestine into the urinary tract, and the patient’s underly-
bowel.100 A short tunnel is made between the mucosal and ing disease processes. Early postoperative complications,
seromuscular layers at each anastomotic site; saline can be such as hemorrhage, wound infection, dehiscence, uri-
injected submucosally using a small needle to aid with the nary leakage, and bowel obstruction, are uncommon.
dissection.101 Small plugs of mucosa are removed from Delayed complications, including problems with the
the ends of each tunnel where the ureter enters the bowel stoma, metabolic abnormalities, renal deterioration,
lumen. After advancing each ureter through its tunnel, infection, and calculi, are encountered more frequently.
the ureter is anastomosed to the mucosa of the bowel.
Stoma Complications
Ureterocolonic Anastomoses
If patients are followed long enough, stomal complica-
The ureters can be implanted into the large bowel in tions are common and lead to significant dissatisfaction.
either a direct refluxing fashion, as previously described Training in proper stomal care, preferably by an enteros-
for small bowel, or a nonrefluxing manner. In the latter, tomal therapist, is important in reducing these issues.
a 3-cm incision is made in a tenia coli (Figure 23-19A). The location of the stoma is also vital in optimizing the
The incision is carried through the muscular layer, stoma, and guidelines for selecting the site have been
sparing the mucosa. At the site of entry into the colonic previously outlined.

Figure 23-18 A, The ureters are spatulated and joined together using a running suture.
B, The ureters are anastomosed to the open proximal end of the conduit.
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 429

Figure 23-19 A, A 3-cm incision is made in a tenia coli and a small ellipse of bowel mucosa
is removed. B and C, The ureter is anastomosed to the mucosa and the muscular layer of
the tenia is reapproximated.

Stomal stenosis is a common late complication of the stoma. Large parastomal hernias should be repaired
cutaneous ureterostomy, occurring in up to 50% of surgically.
patients.102 The incidence of stenosis is lower when Problems with the skin around the stoma are common
dilated, well-vascularized, thick-walled ureters are used. and can be categorized as infectious (i.e., fungal), irrita-
If the ureter is only minimally dilated, a V-flap technique tive (e.g., urine), erosive (e.g., mechanical trauma), or
may be beneficial. The treatment of stomal stenosis after pseudoverrucous.107 Treatment depends on the type and
cutaneous ureterostomy is surgical, generally necessitat- etiology of lesion present. In general, parastomal skin
ing urinary intestinal diversion. problems are greatly reduced with meticulous and gentle
Stomal stenosis is also seen with other types of intes- stoma care, the use of nonirritative adhesives, and a prop-
tinal urinary conduits. In ureteroileal urinary diversion, erly fitting appliance. The aperture in the appliance
the incidence of stenosis is as high as 42% in children and should be just slightly larger than the stoma itself to min-
6.7% in adults.103,104 Similar rates have been reported for imize erosion and prolonged contact between the skin
jejunal conduits.20 Because the colon has a larger lumen, and urine.
one would expect a lower rate of stomal stenosis.
However, long-term studies still demonstrate a signifi-
Ureterointestinal Anastomotic Complications
cant rate of stenosis, up to 61% in children.105,106 Stomal
stenosis can lead to conduit elongation and upper urinary The use of soft Silastic stents reduces urinary extravasa-
tract obstruction. The diagnosis can be made by passing tion, which may lead to periureteral scarring and
a catheter through the stoma and measuring the volume ureterointestinal anastomotic strictures. In general,
of residual urine. The treatment consists of surgical revi- ureteral stents should be placed in those at risk for poor
sion of the stoma. healing. Nonrefluxing anastomotic techniques have
Parastomal hernias are another delayed complications higher rates of stricture formation, although strictures can
associated with intestinal urinary conduits. Parastomal occur at or distant from the intestinal anastomosis. Many
hernia occurs more frequently with colon conduits (10% ureteral strictures can be successfully treated endoscopi-
to 15%) compared with the use of small bowel. The risk cally with either dilation or incision; however, some will
of parastomal hernias is reduced by bringing the bowel ultimately require open surgical repair. The incidence of
through the rectus abdominus muscle and removing ureteral obstruction and stomal stenosis for noncontinent
excess fat from the mesentery of the distal conduit near urinary diversions is summarized in Table 23-3.
430 Part IV Bladder

Table 23-3 Complications of Noncontinent Intestinal Urinary Conduits


No. % Ureteral % Stomal
References Patients Population % Pyelonephritis Obstruction % Stones Stenosis

Ileal conduit

Butcher et al.104 307 Adults 13.8 2.3 3.3 6.7

Johnson et al.108 181 Adults 5.5 15.5 3.3 3.9

Johnson and Lamy109 214 Adults 15.2 18.4 2.5 5.1

Sullivan et al.110 336 Adults 19.2 14.7 4.0 5.1

Middleton and Hendren103 90 Children 20 10 9 42

Shapiro et al.116 90 Children 16.7 22.3 8.9 38

Pitts and Muecke111 242 Both 10.7 4.1 5.8 14

Jejunal conduit

Golimbu and Morales20 30 Both — 3.3 — 6.6

Colon conduit

Schmidt et al.112 22 Adults — 4.5 — 0

Morales and Golimbu105 46 Both 17 13 4.3 13

Althausen et al.113 70 Both 7.1 8.6 4.3 2.8

Elder et al.106 41 Children — 22 16 61.5

Ileocecal conduit

Matsuura et al.114 147 Both 13.6 9.5 5.4 2

Renal Complications
attributed to pyelonephritis, reflux, or obstruction (e.g.,
Pyelonephritis, both early and late, can occur in all types stones, ureteral stricture). The risk of renal dysfunction
of urinary diversion. The incidence of pyelonephritis with increases with time, with obstruction representing a
cutaneous ureterostomy has been reported to be 3%.8 A major cause.110 The reported incidence is variable, but
significantly greater incidence has been found in patients ranges from 18% to 56% and from 8% to 48% for ileal
with ileal and colon conduits, as high as 20% in some and colon conduits, respectively.103,105,106,113, 115, 116
series.110 Although many ureterocolonic anastomoses are
performed using nonrefluxing techniques, even patients
Urinary Calculi
without reflux of urine develop bacterial colonization of
the upper urinary tract.96 Pyelonephritis should be treated Any patient with an intestinal urinary diversion is at risk
with specific antibiotic therapy based on cultures and sus- for urinary stone formation, with an incidence of 5% to
ceptibility of the infecting organism. The sample of urine 10% in adults and 4% to 16% in children. Many factors
should be obtained by direct catheterization of the stoma contribute to the formation of these stones.117 Staples
rather than simply from the collection device. used during conduit construction can serve as a nidus for
Renal deterioration has been associated with all forms stones. Bacteriuria exists in most patients with a urinary
of intestinal urinary diversion. This deterioration can be conduit, where the presence of urease-producing organisms
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 431

can lead to struvite stones. The concomitant metabolic aci- electrolytes move between the extracellular fluid and
dosis in many individuals leads to hypocitraturia and hyper- urine within the conduit. The magnitude and direction
calciuria, further exacerbating the risk of nephrolithiasis.118 of electrolyte shifts are dependent on the differential con-
In patients with an ileal conduit urinary diversion, hyper- centration of the electrolytes. Sodium and chloride are lost
oxaluria and calcium oxalate stones may also result from the into the urine, while potassium and urea are reabsorbed.
use of the terminal ileum. The terminal ileum absorbs bile The loss of sodium leads to decreased extracellular fluid
salts and plays a key role in fat metabolism. When the ter- volume and reduced renal blood flow, causing activation
minal ileum is excluded from the intestinal tract, faulty bile of the renin–angiotensin system and subsequent stimula-
salt and fat absorption leads to saponification of both cal- tion of aldosterone production by angiotensin II.
cium and magnesium within the gut lumen. Decreased Aldosterone increases reabsorption of hydrogen and
cation binding with free gut oxalate causes increased excretion of potassium into the distal renal tubule with
oxalate absorption and hyperoxaluria. resultant acidosis and intracellular potassium release,
The management of stones after urinary diversion can respectively. The reabsorption of urea and dehydration
be difficult. Stones located in the upper tract can be treated leads to azotemia. Treatment of the jejunal conduit syn-
using a combination of percutaneous and retrograde drome consists of oral sodium chloride replacement.
endoscopic techniques along with shock wave lithotripsy.119 Metabolic acidosis can occur when either the ileum or
Shock wave lithotripsy should be avoided when there colon is used as a urinary conduit. Both sodium and chlo-
is stricture distal to the stone.119 Close monitoring is ride are absorbed across the bowel surface, with chloride
crucial because the cause is often multifactorial and absorption greater than sodium. This results in a net loss
recurrence rates are high. Those patients with struvite of bicarbonate into the urine; renal insufficiency con-
stones may benefit from chronic suppressive antibiotics tributes to the severity of the acidosis. Excess potassium
and/or a urease inhibitor. is excreted into the urine as a result of chronic acidosis.
Since the ileum has greater capacity for absorbing uri-
nary potassium compared to the colon, patients with an
Metabolic Complications
ileal conduit tend to have normal total body potassium,
Metabolic abnormalities are common after the introduc- while those with a colon conduit have total body potas-
tion of bowel into the urinary system (Table 23-4). These sium depletion. The treatment for electrolyte abnormal-
include electrolytes disturbances, altered drug kinetics, ities associated with ileal and colon diversions is
osteomalacia, and nutritional disturbances. The severity alkalinization with oral bicarbonate or citrate.
of metabolic problems is determined by length and spe-
cific segment of bowel used, renal function, concentra-
Altered Drug Metabolism
tion of urinary solutes, and degree of urinary stasis.16
Direct drainage of the urine via cutaneous ureterostomy Phenytoin toxicity has been reported in patients with
and pyelostomy avoids these metabolic problems. intestinal urinary conduits.120 Any drug that is excreted by
the kidneys unchanged and can be reabsorbed by the intes-
tine has the potential for toxicity. Patients with continent
Electrolyte Disturbances
urinary diversion and receiving methotrexate chemother-
The use of jejunum as a urinary conduit may result in apy are at risk for toxicity; a catheter should be placed in
hyponatremic, hypochloremic, hyperkalemic metabolic the reservoir during methotrexate administration.
acidosis in up to 40% of patients.19 Clinical symptoms
include nausea, vomiting, anorexia, weakness, and
Nutritional Disturbances
lethargy. These electrolyte changes are the expected
result of normal intestinal physiology. The jejunum is Vitamin B12 is absorbed in the distal ileum. Deficiency of
unable to maintain a large solute gradient; therefore, this vitamin, occasionally the result of using this bowel

Table 23-4 Metabolic Complications Associated with Use of Bowel for Urinary Diversion
Segment Complications Treatment

Jejunum Hypochloremic metabolic acidosis Sodium chloride

Hyperkalemia Volume replacement

Ileum + Colon Hyperchloremic metabolic acidosis Alkalinizing agent (sodium or potassium citrate)

Hypokalemia (colon > ileum) Block chloride transport (chlorpromazine, nicotinic acid)
432 Part IV Bladder

segment, results in megaloblastic anemia and peripheral tinal pouches (mucus and sediment) when compared to
neuropathy. As mentioned earlier, loss of the terminal the native bladder. Intestinal crypts may also serve as bac-
ileum impairs bile salt reabsorption and leads to terial sanctuaries. Therefore, a minimum 10-day course
decreased uptake of fat-soluble vitamins (A, D, E, K). of antibiotic administration is indicated for treating
The patient may have chronic diarrhea from the combi- infection; of course, pyelonephritis may require longer
nation of fat malabsorption, excess bile salts, and loss of courses of therapy.
the ileocecal valve. Cholestyramine and agents that reduce Urinary retention is an infrequent but serious occur-
bowel motility can be used when the diarrhea persists. rence in continent pouches. It is most commonly seen
Chronic acidosis leads to osteomalacia in adults and rickets with continence mechanisms consisting of a nipple valve.
in children.121 Bone remineralization can be accom- If the chimney of the nipple valve is not near the abdom-
plished with oral alkalinization therapy and supplementa- inal surface, the catheter can be directed into folds of
tion with calcium and vitamin D. bowel rather than into the nipple valve proper such that
poor drainage is achieved. Pouch urinary retention rep-
resents a true emergency, and the patient must seek
Continence Mechanism Complications
immediate attention so that catheterization and drainage
One aspect of right colon pouches that has been criti- is promptly performed. The use of a coudé-tipped
cized is loss of the ileocecal valve. Although some catheter is often helpful in this regard, and, rarely, a flex-
patients experience frequent bowel movement in the ible cystoscope is needed.
short term, the majority reports regular bowel function After the immediate problem has been addressed by
through intestinal adaptation or use of pharmacologic emptying the pouch, a catheter is left indwelling for 2 to
therapy. Nevertheless, some patients develop striking 3 days to allow the edema and trauma to the catheteriza-
diarrhea and/or steatorrhea after loss of the ileocecal tion portal to resolve. Subsequently, the patient should
valve. This may be particularly true in the pediatric be observed for the ability to successfully catheterize on
patient in whom there is underlying neurogenic bowel a number of occasions. The appropriate angle of entry
(e.g., myelomeningocele). can be taught until the patient is comfortable with the use
A limitation of stapled nipple valves is the potential for of the new catheter; our preference is to routinely use
stone formation on exposed staples. This has been signif- coudé catheters with nonnipple valve pouches.
icantly decreased by the omitting staples at the tip of the Intraperitoneal rupture of catheterizable pouches has
intussuscepted nipple valve as suggested by Skinner et been reported.122–125 In general, this occurs more com-
al.39 However, more proximal staples occasionally erode monly in the neurologic patient in whom sensation of
into the pouch and serve as a nidus for stone formation. pouch fullness may be less distinct. Oftentimes, associ-
These stones are usually manageable with endoscopic ated mild abdominal trauma, such as a fall, is antecedent
intact extraction with forceps or after fragmentation to the rupture. Patients with rupture require immediate
using electrohydraulic, ultrasonic, or laser lithotripsy. pouch decompression and radiologic studies. For
Although exposed staples may serve as a nidus for stone patients with large defects, surgical exploration and
formation, continent urinary diversion itself results in repair are required. If the amount of urinary extravasa-
greater urinary excretion of calcium, magnesium, and tion is small and the patient does not have an acute
phosphate compared with ileal conduit diversion.118 abdomen, catheter drainage and antibiotic administra-
Thus, all patients undergoing continent diversion are at tion may suffice in treating intraperitoneal rupture.
an increased risk for the formation of reservoir stones. However, patients managed conservatively require care-
“Pouchitis” is a condition that is manifested by pain in ful monitoring. If there are any signs of progressive peri-
the region of the pouch and increased pouch contractil- tonitis, surgical exploration and repair are imperative.
ity. Although occurring infrequently, pouchitis may
result in temporary failure of the continence mechanism
Complications by Specific Type of Urinary
because of the hypercontractility of the bowel segment
Diversion
employed for construction of the pouch. The patient typ-
ically presents with a history of sudden explosive dis- Overall complication rates, reoperation rates, and mor-
charge of urine through the continence mechanism, tality rates are reported in Table 23-5 for each different
rather than dribbling incontinence, along with discom- continent cutaneous reservoir.
fort in the region of the pouch. Appropriate antibiotic
therapy usually results in the resolution of symptoms. We
Ureterosigmoidostomy
have found that a short course of antibiotics (i.e., 3 days)
is not successful in treating pouch infections, perhaps The most serious immediate complication from
due to the larger amount of foreign material within intes- ureterosigmoidostomy is anuria if the anastomosis is not
Table 23-5 Comparison of Continent Cutaneous Reservoirs: Reoperation Rate, Mortality and Complication Rates
No Reoperation COMPLICATIONS (%)
Pouch Study Patients Rate Mortality (%) Early Late Stomal Stenosis Pouch Stones

Kock Pouch Koch et al.66 12 58% 0 16.7 58 NR NR


Skinner et al.125 489 10–15% 1.9 16.2 29 NR 7.6%

T Pouch Stein et al.67 9 NR 0 0 11.1 NR 11.1%

Mainz pouch Thuroff et al.68 100 11% (nipple 0 7 18* NR NR


revision)
Lampel et al.69 440 NR NR 12 37 11.7% ileal nipple 6.8%
14.7% appendiceal stoma 3% ileal nipple
patients
Gerharz et al.70 193 24% appendix 0 NR NR 18% appendix stoma 0%
stoma
12% ileal nipple 1.9% Ileal nipple 2.8%

UCLA pouch deKernion et al.71 19 NR 0 NR NR NR NR

Indiana pouch Rowland et al.73 29 17.2% 0 20.6 NR NR NR


Scheidler et al.74 91 26% 1.1 NR NR NR NR
Ahlering et al.76 70 4% to 10% 1.4 13 17.1 NR NR
Carroll et al.77 21 4.7% 0 NR NR 4.7% 0%
Rowland et al.78† 81 17.3% 0 12.3 28.4 NR NR
Bihrle et al.79‡ 50 4% 0 20 14 0% NR

Florida pouch Lockhardt et al.80 11 NR NR NR NR NR NR


Lockhardt et al.81 107 3.7% 0 12.4 10.3 1.9%§ 1.9%

Duke pouch¶ Webster et al.82 7 28.5% 0 NR NR 14.2% NR

U. Miami pouch Benson et al.84 75 NR NR 25 21 NR NR

Penn pouch Sumfest et al.85 NR NR NR NR 10.6 19.1% NR

Complication rates are reservoir related complications.


*11 patients had valve prolapse or incontinence; none of these problems were noted in later series after the nipple valve stabilization had been modified.
†The last 81 patients operated on by Rowland and associates underwent construction of a stapled efferent limb; in the last 20 patients, the reservoir was created with absorbable staples.
‡Mean follow-up in this study was 2.5 years.
§No specific comments regarding stomal stenosis. This percentage represents the number of cases reported as having catheterization difficulties.
¶The long-term outcome of these pouches is unknown.

NR, not reported.


Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 433
434 Part IV Bladder

stented. Anuria after removal of the ureteral stents indi- emboli (0.7%). The late complication rate was 37% and
cates bilateral obstruction, likely secondary to tissue edema. was predominantly attributable to the pouch. Stomal fail-
In both cases, percutaneous nephrostomy tubes should be ure requiring open revision occurred in 45 patients (8%)
placed, especially in patients with fever or flank pain. and was directly related to the continence mechanism.
Urinary leakage from the ureterocolonic anastomosis Only 2 of 46 patients (1.4%) with an appendiceal conti-
or from the colostomy incision can occur early in the nence mechanism were incontinent, but stomal stenosis
postoperative period. The incidence of this complication occurred in 21%. The stapled ileocecal intussusception
can be minimized by watertight ureterocolonic anasto- described previously is the current standard, with a
moses, use of ureteral stents, and continuous drainage of reduction of long-term incontinence rates to 10%. Other
the rectal contents. Proper drainage of the rectal con- late complications include ureteral reimplants in 4.9%,
tents can be achieved by using two rectal tubes that have stomal stenosis in 11.7% of patients with an ileal nipple,
multiple perforations. Stents and tubes should be irri- and 14.7% of patients with an appendiceal stoma.
gated regularly (4 to 6 hours) to ensure patency. If the Patients who developed calculi within the pouch
patient is stable, it is reasonable to wait because the leak (6.8%) were primarily treated with percutaneous proce-
may seal by itself. In patients in whom significant leaks dures for stone removal. Despite the loss of the terminal
persist, treatment options include immediate reoperation ileum, it has been rare to see a significant drop in serum
and reanastomosis, percutaneous nephrostomy tube vitamin B12 levels, and very few patients have developed
placement, or cutaneous diversion. Renal deterioration a macrocytic anemia or neurologic symptoms. Twenty-
can occur late secondary to obstruction, reflux, and ascend- five percent of patients are taking oral alkalinization
ing infections. When the rectal tube is removed, the patient drugs to avoid metabolic acidosis. Additionally, since
must be closely monitored for the development of hyper- 1988 the incontinence rate has been only 3.2%, and less
chloremic acidosis. Because this is common, empiric bicar- than 2% of the patients with an appendiceal continence
bonate replacement may be initiated immediately. Potassium mechanism have been incontinent.
citrate should also be considered at the outset because of the Gerharz and associates58 from Marburg, Germany,
risk for hypokalemia. In patients who cannot maintain elec- reported a single institution experience with the Mainz I
trolyte homeostasis with oral medication, nighttime rectal ileocecal pouch. From 1990 to 1996, 202 consecutive
urinary drainage is mandated. The incidence of colorectal patients underwent continent diversion, 96 with a sub-
cancer after ureterosigmoidostomy is 3.5% to 13.3%, repre- mucosally embedded in situ appendix and 106 with an
senting an 80- to 550-fold greater risk when compared the intussuscepted ileal nipple. All patients had an umbilical
general population.126–128 Due to the concern for developing stoma. In 172 of 200 patients (85%), no stomal compli-
malignancy, it is suggested that patients have routine moni- cations occurred. In 17 of 96 patients (18%) with an
toring for blood in the stool, cytologic examination of the appendiceal stoma, 23 revisions were performed for
urine-feces mixture, and annual colonoscopy. Barium ene- stomal stenosis. In contrast, only 13 of 106 patients
mas are contraindicated because of the potential for reflux (12%) with an intussuscepted ileal nipple developed a
of this material, which could lead to a septic event. stomal problem. However, these patients required more
invasive, major procedures for correction, and the stomas
with an appendiceal stenosis could usually be repaired
Kock Pouch
with a minor procedure. Three patients with an ileal nip-
Skinner et al. have the most experience with this tech- ple (3%) developed pouch calculi, whereas none of the
nique and reported early and late complication rates of patients with an appendiceal continence mechanism
16.2% and 22%, respectively. The reoperation rate developed stones. As a result, the authors concluded that
approaches 10% to 15% in their last series, mostly for when available, the appendix should be the intestinal
correction of sliding of the continence nipple or erosion continence mechanism of choice.
of the stabilizing efferent nipple collar into the lumen.
Upper urinary tract deterioration, electrolyte imbalance,
Indiana Pouch
and stone formation are rare.
The reoperation rate, mainly caused by complications
with the efferent limb, was 26%. Because of the high rate
Mainz Pouch
of nighttime incontinence in Rowland’s series,35 Ahlering
In the mid-1990s, the 10- and 12-year experience with and colleagues129 detubularized the entire cecum to avoid
the Mainz pouch and the variations created by its devel- high pressure contractions from the cecum. Complete
opers was presented.56,57 The early complication rate was detubularization seems to be superior to ileal or sigmoid
12%; complications included mechanical ileus requiring colon patches.
open revision (1.6%), pouch leakage requiring revision Elegant urodynamic studies were conducted in
(0.9%), wound dehiscence (0.7%), and fatal pulmonary Indiana pouch variants by Carroll et al.130 They found
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 435

only 86% of patients totally continent in a small series. ments confounds these data. Bjerre et al.134 also com-
However, their pouch capacities exceeded 650 ml. Peak pared the ileal conduit with Kock pouch and found no
contractions of 47 cm H2O were recorded at capacity. differences in body image, partner relations, and global
life satisfaction. Despite a higher quality of life meas-
Gastric Pouches urement in those with a neobladder, nearly all patients
in both groups (>90%) felt in good health or compara-
Over a 10-year period from January 1985 to June 1995, ble to people of similar age.
Carr and Mitchell96 reported on the use of stomach in 12 McGuire et al.135 used the validated Rand 36-Item
patients. Seven had urinary reservoirs totally constructed Health Survey (SF-36) to measure quality of life in
from stomach and 5 had composite reservoirs. They patients with bladder cancer undergoing cystectomy
report continence in all patients but that the continence and urinary diversion. No difference in the physical
mechanisms have oftentimes required revision. The aver- component summary (PCS) score was apparent after
age bladder capacity was 309 ml, and average compliance ileal conduit, neobladder, and Indiana pouch construc-
was 12.9 ml/cm H2O. When stomach is used as a bladder tion when compared to age- and sex-based population
augment or as a portion of a neobladder, a dysuria and norms. However, patients with ileal conduits had signif-
hematuria syndrome has been reported.94 icantly decreased mental quality of life (p = 0.01), while
those with other forms of diversion were similar to
QUALITY OF LIFE matched population norms. Several more recent studies
have applied validated surveys to address this question.
With improvements in surgical technique and experi- Fujisawa et al.136 also used the SF-36 survey to assess
ence, orthotopic urinary reconstruction is a viable effects of different forms of urinary reconstruction.
alternative in many patients. The lack of a stoma or the There were no significant differences between patients
need for an appliance or catheterization should benefit with an ileal conduit and neobladder at follow-up of 45
patient acceptance and self-image. Whether these and 31 months, respectively. Although similar in both
changes translate into an improved quality of life groups, role-physical functioning and role-emotional
remains unclear. function were lower than U.S. population norms. Dutta
Early reports merely examined the impact of conduit et al.137 assessed both general health and cancer-specific
creation in patients receiving an ileal conduit. Overall, aspects in 112 patients using the SF-36 and Functional
these studies do not demonstrate a negative effect from a Assessment of Cancer Therapy-General (FACT-G)
noncontinent urinary diversion. Fossa et al.131 found that questionnaires, respectively. SF-36 scores were signifi-
few patients experienced a reduction in general quality of cantly better for patients with a neobladder compared
life (13%) or restriction in professional and daily func- with an ileal conduit (p = 0.008); however, after control-
tioning (16%); however, the instrument used was not a ling for age and disease variables, the differences were
validated survey. Similarly, Cespedes et al.132 reported less (p = 0.09). Global satisfaction in both groups was
that 83% of patients had no change or an increase in high (>85%). They conclude that patients with an ortho-
activity level after ileal conduit creation. Others have topic neobladder enjoy a marginal increase in quality of
found an improved quality of life (60% to 80%) after life when compared with patients receiving an ileal con-
surgery and ileal conduit diversion; this may reflect duit. Using the QLQ-C30 and an institutional question-
improvements in urinary function in a population with- naires, Hobisch et al.138 provided stronger evidence that
out cancer and does not directly compare outcomes with the neobladder is associated with improved quality of life
other forms of reconstruction. relative to the ileal conduit. Overall satisfaction was 97%
Hart et al.133 compared quality of life after radical in patients with an orthotopic neobladder and 35% in
cystectomy for cancer and reconstruction using an ileal patients with an ileal conduit, respectively. Table 23-6
conduit, cutaneous Kock pouch, or urethral Kock summarizes the literature concerning quality of life after
pouch. No significant differences were noted among ileal conduit diversion using validated instruments.
these groups with respect to overall quality of life, The recognition that quality of life is an important
emotional distress, and problems with social, physical, treatment outcome in urologic malignancies is a recent
or functional activities. Most patients (95%) rated qual- phenomenon. This is particularly true for radical cystec-
ity of life as good to excellent, although urinary and tomy, where surgery entails not only radical extirpation
sexual functions were significantly affected in all for cancer cure but also requires complex reconstruction
groups. Over half of patients with ileal conduits of the urinary tract. Although the data do not demon-
reported frequent difficulty in caring for the collection strate a clear advantage to orthotopic or continent reser-
device and 30% had occasional problems with skin irri- voirs, it appears that these procedures provide excellent
tation. The influence of disease-related factors on the quality of life, with minimal increase in morbidity, and
measurements of a nonvalidated questionnaire instru- should be strongly considered when technically feasible.
436 Part IV Bladder

Table 23-6 Studies of Quality of Life (QOL) After Urinary Diversion Utilizing Validated Instruments
(All Compared with Ileal Conduit)
References Number Type Instrument Cystectomy Results

Bjerre et al.134 67 Kock neobladder Qualitative interview Yes Higher QOL with neobladder
Similar overall satisfaction

Hart et al.133 224 Koch* Validated† Yes No differences in QOL


Urinary problems, sexual dysfunction

Fujisawa et al.136 56 Neobladder SF-36 Yes No difference in QOL


Both lower in role-physical,
emotional
Overall high satisfaction

McGuire et al.135 92 Neobladder, SF-36 Yes Conduit reduced mental health


Indiana component
Physical component similar to norms

Hobisch et al.138 102 Neobladder QLQ-C30‡ Yes High QOL with neobladder
Lower satisfaction with conduit

Dutta et al.137 112 Neobladder SF-36, FACT-G Yes No differences on multivariate


analysis
High satisfaction with both

Satoh142 45 Ileocecal rectal QLQ-C30 Yes Conduit worse in three domains


bladder

Hara143 85 Neobladder SF-36 Yes Both with lower QOL than norms
Lower role-emotional with conduit

*Both cutaneous and urethral.


†Adapted from various validated questionnaires.
‡European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-Core Questionnaire 30.

Conversely, the ileal conduit is preferable in some and mobilize the ureter so that anastomosis to the skin
patients and this alone does not result in a significant was possible. The initial model for performing the ileal
reduction in overall quality of life. Patients can continue conduit was described by Sanchez de Badajoz et al.139 in
with an active, normal lifestyle after nearly all of the 1992. Case reports describe successful operations in 2
reconstructive options. Complete preoperative discus- patients requiring supravesical diversion without pelvic
sion and explanation of the various forms of urinary exenteration. It is important to note that the bowel exclu-
reconstruction are crucial factors in patient preparation sion, reanastomosis, and ureteroileal anastomoses were
for the changes in their quality of life. performed extracorporeally via a port site.
With the development of laparoscopic devices and
experience with intracorporeal suturing, more centers
NOVEL TECHNIQUES
are attempting laparoscopic ileal conduits. In combina-
The realm of laparoscopy in urology has evolved from tion with laparoscopic radical cystectomy, most sur-
diagnosis to simple procedures (e.g., renal biopsy) to geons are performing at least a portion of the operation
complete organ removal and now to complex reconstruc- through an extended port site or the specimen extrac-
tion. Many reports have documented the use of mini- tion incision. Fergany and colleagues140 have developed
mally invasive techniques to create noncontinent and a porcine model where the entire ileal conduit is per-
continent urinary drainage. The earliest descriptions of formed intracorporeally, demonstrating technical feasi-
laparoscopic urinary drainage in humans involved the bility, as well as excellent short- and long-term
creation of cutaneous ureterostomy for palliation in outcomes in these animals. These experiences formed
advanced pelvic malignancy. Laparoscopy, both the basis for the operation in 2 patients undergoing rad-
transperitoneal and retroperitoneal, was used to identify ical cystoprostatectomy with ileal conduit for bladder
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 437

Table 23-7 Experience with Laparoscopic Ileal Conduit


Completely Time EBL
References Number Cystectomy Intracorporeal (hours) (ml) Follow-up Complications

Kozminski144 1 No No 6.3 NR 3 months None

Vara-Thorbeck145 1 No No 4 NR 4 months None

Sanchez de Badajoz146 1 Yes No 8 NR 1 year None

Gill147 2 Yes Yes 10.8 1100 NR None

Potter et al.141 1 No Yes 4.5 NR 5 years Ileus

Gupta148 5 Yes Yes 7.5 360 2 years Bowel


obstruction

*1 unit blood transfusion.


NR, not reported.

cancer; again, the entire operation, including bowel 8. Rainwater L, Leary F, Rife C: Transureteroureterostomy
manipulation and suturing, was completed entirely with cutaneous ureterostomy: a 25-year experience.
within the abdomen. Potter et al.141 also reported the J Urol 1991; 146:13.
long-term follow-up (5 years) in a patient, where 9. Persky L, McDougal W, Kedia K:
Transureteroureterostomy: an adjunct to cystectomy.
supravesical diversion was created by a pure laparo-
Urology 1980; 16:20.
scopic ileal conduit. Table 23-7 summarizes the litera-
10. Chute R, Sallade R: Bilateral side-to-side cutaneous
ture regarding surgical outcomes with laparoscopic ileal ureterostomy in the midline for urinary diversion. J Urol
conduit urinary diversion. 1961; 85:280.
The role of minimally invasive surgery in urinary 11. Rinker J, Blanchard T: Improvement of the circulation
diversion remains unclear. Although promising, further of the ureter prior to cutaneous ureterostomy: a clinical
studies are needed to confirm benefits, such as reduced study. J Urol 1966; 96:44.
pain and shorter hospitalization. Moreover, the tech- 12. Straffon R, Kyle K, Corvalan J: Techniques of cutaneous
nique must mimic the open operation in that complica- ureterostomy and results in 51 patients. J Urol 1970;
tions are minimal and durable results are achieved, given 143:138.
the challenging nature of the operation. 13. Seiffert L: Die “Darm-siphonblase.” Arch Klin Chir
1935; 183:569.
14. Bricker E: Bladder substitution after pelvic evisceration.
Surg Clin North Am 1950; 30:1511.
REFERENCES 15. Benson M, Olsson C: Urinary diversion. In Walsh P,
Retik A, Stamey T, et al (eds): Campbell’s Urology, 6th
1. Boyd S, Feinberg S, Skinner D, et al: Quality of life edition. Philadelphia, WB Saunders Co, 1992.
survey of urinary diversion patients: comparison of ileal 16. McDougal W: Metabolic complications of urinary
conduits versus continent Kock ileal reservoirs. J Urol intestinal diversion. J Urol 1992; 147:1199.
1987; 138:1386. 17. Turnbull RJ, Hewitt C: Loop-end myotomy ileostomy in
2. Immergut M, Jacobson J, Culp D: Cutaneous the obese patient. Urol Clin North Am 1978; 5:423.
pyelostomy. J Urol 1969; 101:276. 18. Chechile G, Klein E, Bauer L: Functional equivalence of
3. Gleeson M, Griffith D: Urinary diversion. Br J Urol end and loop ileal conduit stomas. J Urol 1992; 147:582.
1990; 66:113. 19. Klein E, Montie J, Montague D: Jejunal conduit urinary
4. Rosen M, Roth D, Gonzales EJ: Current indications for diversion. J Urol 1986; 135:244.
cutaneous ureterostomy. Urology 1994; 43:92. 20. Golimbu M, Morales P: Jejunal conduits: technique and
5. Whitmore WJ: Ureteral diversion. In Bergman H (ed): complications. J Urol 1975; 113:787.
The Ureter, p 603. New York, Harper & Row, 1967. 21. Turner-Warwick R: Colonic urinary conduits. Proc R
6. Sarduy G, Crooks K, Smith J: Results in children managed Soc Med 1960; 53:1032.
by cutaneous ureterostomy. Urology 1982; 19:486. 22. Zinman L, Libertino J: Ileocecal conduit for temporary
7. Young JJ, Ajedia F: Further observations on flank and permanent urinary diversion. J Urol 1975; 113:317.
ureterostomy and cutaneous transureteroureterostomy. 23. Simon J: Ectopia vesicae (absence of the anterior walls of
J Urol 1966; 95:327. the bladder and pubic abdominal parietes): operation for
438 Part IV Bladder

directing the orifices of the ureters into the rectum: 42. Benchekroun A: The ileocecal continent bladder.
temporary success; subsequent death; autopsy. Lancet In King L, Stone A, Webster G (eds): Bladder
1852; 2:568. Reconstruction and Continent Urinary Diversion,
24. Smith T: An account of an unsuccessful attempt to treat pp 224–237. Chicago, Year Book Medical, 1987.
extroversion of the bladder by a new operation. St Barth 43. Mitrofanoff P: Cystostomie continente trans-
Hosp Rep 1879; 15:29. appendiculaire dans le traitement des vessies
25. Allen T: Editorial comment. J Urol 1995; 153:1431. neurologiques. Chir Pediatr 1980; 21:297.
26. Bissada N, Morcos R, Morgan W, et al: 44. Duckett J, Snyder HI: Use of the Mitrofanoff principle
Ureterosigmoidostomy: Is it a viable procedure in the in urinary reconstruction. Urol Clin North Am 1986;
age of continent urinary diversion and bladder 13:271.
substitution? J Urol 1995; 153:1429. 45. Riedmiller H, Steinbach F, Thuroff J, et al: Continent
27. Ambrose S: Ureterosigmoidostomy. In Glenn J (ed): appendix stoma—a modification of the Mainz pouch
Urologic Surgery, pp 511–520. Philadelphia, JB technique. Presented at the EAU Congress, Amsterdam,
Lippincott, 1983. 1990.
28. Spirnak J, Caldamone A: Ureterosigmoidostomy. Urol 46. Kock N, Nilson A, Nilsson L, et al: Urinary diversion
Clin North Am 1986; 13:285. via a continent ileal reservoir: clinical results in 12
29. Silberman R: Ammonia intoxication following patients. J Urol 1982; 128:469.
ureterosigmoidostomy in a patient with liver disease. 47. Skinner D: The Kock pouch for continent urinary
Lancet 1958; 2:937. reconstruction focusing on the afferent segment and the
30. Leadbetter W: Consideration of problems incident to reservoir. Scand J Urol Nephrol Suppl 1992; 142:77.
performance of ureteroenterostomy: report of a 48. Skinner D, Lieskovsky G, Boyd S: Continent urinary
technique. J Urol 1951; 65:818. diversion. J. Urol 1989; 141:1323.
30a. Goodwin WE. Ureterosigmoidoscopy (open 49. Abol-Enein H, Ghoneim M: A novel uretero-ileal
transcolonic ureterointestinal anastomosis. In Cooper P reimplantation technique: the serous lined extramural
(ed): The craft of surgery; p 1367, Boston; Little, Brown tunnel. A preliminary report. J Urol 1994; 151:1193.
& Co, 1964.) 50. Abol-Enein H, Ghoneim M: Optimization of
31. Benchekroun A: Hydraulic valve for continence and ureterointestinal anastomosis in urinary diversion: an
antireflux: a 17 year experience of 210 cases. Scand J experimental study in dogs. III. A new antireflux
Urol Nephrol Suppl 1992; 142:70. technique for ureteroileal anastomosis: a serous lined
32. Woodhouse C, MacNeily A: The Mitrofanoff principle. extramural tunnel. Urol Res 1993; 21:135.
Br J Urol 1994; 74:447. 51. Bochner B, Stein J, Ginsberg D, et al: A serous lined
33. Lampel A, Hohenfellner M, Schultz D, et al: antireflex valve: in vivo fluorourodynamic evaluation of
Submucosal seromuscular tube and submucosal bowel antireflux continence mechanism. J Urol 1998;
flap tube: two new stoma techniques for Mainz pouch 160:112.
continent cutaneous urinary diversion. J Urol 1995; 52. LeDuc A, Camey M, Teillac P: An original antireflux
153:305A. ureteroileal implantation technique: long-term follow-
34. Burns M, Mitchell M: Tips on constructing the Mitrofanoff up. J Urol 1987; 137:1156.
appendiceal stoma. Contemporary Urol 1990;2:10. 53. Stein J, Buscarini M, DeFilippo R, et al: Application of the
35. Rowland R, Mitchell M, Bihrle R: The cecoileal T pouch as an ileo-anal reservoir. J Urol 1999; 162:2052.
continent urinary reservoir. World J Urol 1985; 3:185. 54. Stein J, Lieskovsky G, Skinner D: T-mechanism applied
36. Lockhart J: Remodeled right colon: an alternative to urinary diversion: the orthotopic T-pouch ileal
urinary reservoir. J Urol 1987; 138:730. neobladder and cutaneous double T-pouch ileal
37. Bejany D, Politano V: Stapled and nonstapled tapered reservoir. Tech Urol 2001; 3:209.
distal ileum for construction of a continent colonic 55. Thuroff J, Alken P, Riedmiller H: The Mainz pouch
urinary reservoir. J Urol 1988; 140:491. (mixed augmentation ileum ′n zecum) for bladder
38. Hendren W: Urinary diversion and undiversion in augmentation and continent diversion. World J Urol
children. Surg Clin North Am 1976; 56:425. 1985; 3:179.
39. Skinner D, Boyd S, Lieskovsky G: Clinical experience 56. Stein R, Young M, Doi Y, et al: Continent urinary
with the Kock continent ileal reservoir for urinary diversion using the Mainz pouch I technique—ten years
diversion. J Urol 1984; 132:1101. later. J Urol 1995; 153:251A.
40. King L: Protection of the upper tracts in undiversion. 57. Lampel A, Fisch M, Stein R, et al: Continent diversion
In King L, Stone A, Webster G (eds): Bladder with the Mainz pouch. World J Urol 1996; 14:85.
Reconstruction and Continent Urinary Diversion, pp 58. Gerharz E, Kohl U, Weingartner K, et al: Complications
127–153. Chicago, Year Book Medical, 1987. related to different continence mechanisms in ileocecal
41. Stein J, Lieskovsky G, Ginsberg D, et al: The T pouch: reservoirs. J Urol 1997; 158:1709.
an orthotopic ileal neobladder incorporating a 59. Thuroff J, Alken P, Riedmiller H, et al: 100 cases of
serosal lined ileal antireflux technique. J Urol 1998; Mainz pouch: continuing experience and evolution.
159:1836. J Urol 1988; 140:283.
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 439

60. Riedmiller H, Steinbach F, Thuroff J, et al: Continent 81. Lockhart J, Pow-Sang J, Persky L, et al: Results,
appendix stoma: a modification of the Mainz pouch complications and surgical indications of the Florida
technique. AUA 85th Annual Meeting, 13–17 May, New pouch. Surg Gynecol Obstet 1991; 173:289.
Orleans, 1990. 82. Webster G, Bertram R: Continent catheterizable urinary
61. Lampel A, Hohenfellner M, Schultz-Lampel D, et al: In diversion using the ileocecal segment with stapled
situ tunneled bowel flap tubes: 2 new techniques of a intussusception of the ileocecal valve. J Urol 1986; 135:465.
continent outlet for Mainz pouch cutaneous diversion. 83. Bejany D, Politano V: Stapled and nonstapled tapered
J Urol 1995; 153:308. distal ileum for construction of a continent colonic
62. Lampel A, Thuroff J: Bowel-flap tubes for continent urinary reservoir. J Urol 1988; 140:491.
urinary diversion. World J Urol 1998; 16:235. 84. Benson M, Olsson C: Cutaneous continent urinary
63. Raz S: The UCLA Neobladder. 1994 (edited by diversion. In Walsh P, Retik A, Vaughn EJ, et al (eds):
J. deKernion). Campbell’s Urology, Vol 4, 8th edition, Section 13
64. Webster G, King L: Further commentary: cecal bladder. Urologic Surgery. Philadelphia, Elsevier Science, 2003.
In King L, Stone A, Webster G (eds): Bladder 85. Sumfest J, Burns M, Mitchell M: The Mitrofanoff
Reconstruction and Continent Urinary Diversion, principle in urinary reconstruction. J Urol 1993;
pp 206–208. Chicago, Year Book Medical, 1987. 150:1875.
65. Light J, Scardino P: Radical cystectomy with 86. Adams M, Mitchell M, Rink R: Gastrocystoplasty: an
preservation of sexual and urinary function: use alternative solution to the problem of urological
of the ileocolonic pouch (Le Bag). Urol Clin North Am reconstruction in the severely compromised patient.
1986; 13:261. J Urol 1988; 140:1152.
66. Kock N, Nilson A, Nilsson L, et al: Urinary diversion 87. Carr M, Mitchell M: Continent gastric pouch. World
via a continent ileal reservoir: clinical results in 12 J Urol 1996; 14:112.
patients. J Urol 1982; 128:469. 88. Rowland R, Mitchell M, Bihrle R: The cecoileal
67. Stein J, Lieskovsky G, Skinner D: The double-T-pouch: continent reservoir. World J Urol 1985; 3:185.
a novel continent cutaneous ileal reservoir. J Urol1999; 89. Rowland R: Present experience with the Indiana pouch.
161:253A (pg 67). World J Urol 1996; 14:92.
68. Thuroff J, Alken P, Riedmiller H, et al: 100 cases of 90. Adams M, Mitchell M, Rink R: Gastrocystoplasty: an
Mainz pouch: continuing experience and evolution. alternative solution to the problem of urological
J Urol 1988; 140:283. reconstruction in the severely compromised patient.
69. Lampel A, Fisch M, Stein R, et al: Continent diversion J Urol 1988; 140:1152.
with the Mainz pouch. World J Urol 1996; 14:85. 91. Lockhart J, Davies R, Cox C, et al: The gastroileal
70. Gerharz E, Kohl U, Weingartner K, et al: Complications pouch: an alternative continent urinary reservoir for
related to different continence mechanisms in ileocecal patients with short bowel, acidosis and/or extensive
reservoirs. J Urol 1997; 158:1709. pelvic radiation. J Urol 1993; 150:46.
71. deKernion J, Raz S, Wahle G, et al: The UCLA 92. Austin P, DeLeary G, Homsy Y, et al: Long-term
neobladder: clinical urodynamic and metabolic results. metabolic advantages of a gastrointestinal composite
J Urol 1994; 151:242A (no. 59). urinary reservoir. J Urol 1997; 158:1704.
72. Rowland R, Mitchell M, Bihrle R: The cecoileal 93. Nguyen D, Bain M, Salmonson K, et al: The syndrome
continent urinary reservoir. World J Urol 1985; 3:185. of dysuria and hematuria in pediatric urinary
73. Rowland R, Mitchell M, Bihrle R, et al: Indiana reconstruction with stomach. J Urol 1993; 150:707.
continent urinary reservoir. J Urol 1987; 137:1136. 94. Gosalbez R, Padron O, Singla A, et al: The gastric
74. Scheidler D, Klee L, Rowland R, et al: Update on the augment single pedicle tube catheterizable stoma: a
Indiana continent urinary reservoir. J Urol 1989; useful adjunct to reconstruction of the urinary tract.
141:302A. J Urol 1994; 152:2005.
75. Carroll P, Presti JJ, McAnnich J, et al: Functional 95. Carr M, Mitchell M: Continent gastric pouch. World
characteristics of the continent ileocecal urinary J Urol 1996; 14:112.
reservoir: mechanisms of urinary continence. J Urol 96. Gonzales R, Reinberg Y: Localization of bacteriuria in
1989; 142:1032. patients with enterocystoplasty and nonrefluxing
76. Ahlering T, Weinberg A, Razor B: Modified Indiana conduits. J Urol 1987; 138:1104.
pouch. J Urol 1991; 145:1156. 97. Yang W: Yang needle tunneling technique in creating
77. Carroll P, Presti JJ: Comparison of plicated and stapled antireflux and continent mechanisms. J Urol 1993; 150:830.
continent ileocecal stoma. Urology 1992; 40:107. 98. Hirdes W, Hoekstra I, Vliestra H: Hammock
78. Rowland R: Present experience with the Indiana pouch. anastomoses: a nonrefluxing ureteroileal anastomosis.
World J Urol 1996; 14:92. J Urol 1988; 139:517.
79. Bihrle R: The Indiana pouch continent urinary reservoir. 99. Wallace D: Ureteric diversion using a conduit: a
Urol Clin North Am 1997; 24:773. simplified technique. Br J Urol 1966; 38:522.
80. Lockhart J: Remodeled right colon: an alternative 100. Starr A, Rose D, Cooper J: Antireflux ureteroileal
urinary reservoir. J Urol 1987; 138:730. anastomosis in humans. J Urol 1975; 113:170.
440 Part IV Bladder

101. Menon M, Yu G, Jeffs R: Technique for antirefluxing 121. McDougal W, Koch M, Shands CI, et al: Bony
ureterocolonic anastomosis. J Urol 1982; 127:236. demineralization following urinary intestinal diversion.
102. Feminella J, Lattimer J: A retrospective analysis of J Urol 1988; 140:853.
70 cases of cutaneous ureterostomy. J Urol 1971; 122. Kristiansen P, Mansson W, Tyger J: Perforation of
106:538. continent cecal reservoir for urine twice in one patient.
103. Middleton AJ, Hendren W: Ileal conduit in children at Scand J Urol Nephrol 1991; 25:279.
the Massachusetts general hospital from 1955 to 1970. 123. Thompson S, Kursh E: Delayed spontaneous rupture of
J Urol 1976; 115:591. an ileocolonic neobladder. J Urol 1992; 148:1890.
104. Butcher HJ, Sugg W, McAfee C, et al: Ileal conduit 124. Wantanabe K, Kato H, Misawa K, et al: Spontaneous
method of ureteral urinary diversion. Ann Surg 1962; perforation of an ileal neobladder. Br J Urol 1994;
156:682. 73:460.
105. Morales P, Golimbu M: Colonic urinary diversion: 10 125. Skinner D, Lieskovsky G, Boyd S: Continent urinary
years of experience. J Urol 1975; 113:302. diversion. J Urol 1989; 141:1323.
106. Elder D, Moisey C, Rees R: A long-term follow-up of 126. Silverman S, Woodhouse C, Strachan J, et al: Long-term
the colonic conduit operation in children. Br J Urol management of patients who have had urinary diversions
1979; 51:462. into colon. Br J Urol 1986; 58:634.
107. Borglund E, Nordstrom G, Nyman C: Classification of 127. Stewart M: Urinary diversion and bowel cancer. Ann R
peristomal skin changes in patients with urostomy. J Am Coll Surg Engl 1986; 68:98.
Acad Dermatol 1988; 19:623. 128. Kalble T, Mohring K, Tricker A, et al: Adenocarcinoma
108. Johnson DE, Jackson L, Guinn GA: Ileal conduit of the colon following ureterosigmoidostomy. Akt Urol
diversion for carcinoma of the bladder. South Med J 1989; 20:173.
1970; 63:1115. 129. Ahlering T, Weinberg A, Razor B: Modified Indiana
109. Johnson DE, Lamy SM: Complications of a single stage pouch. J Urol 1991; 145:1156.
radical cystectomy and ileal conduit diversion: review of 130. Carroll P, Presti JJ, McAnnich J, et al: Functional
214 cases. J Urol 1977; 117:171. characteristics of the continent ileocecal urinary
110. Sullivan J, Grabstald H, Whitmore WJ: Complications reservoir: mechanisms of urinary continence. J Urol
of ureteroileal conduit with radical cystectomy: review of 1989; 142:1032.
336 cases. J Urol 1980; 124:797. 131. Fossa S, Reitan J, Ous S, et al: Life with an ileal conduit
111. Pitts WJ, Muecke E: A 20-year experience with ileal in cystectomized bladder cancer patients: expectations
conduits: the fate of the kidneys. J Urol 1979; 122:154. and experience. Scand J Urol Nephrol 1987; 21:97.
112. Schmidt JD, Buchsbaum HJ, Jacobo EC: Transverse 132. Cespedes R, McGuire E, Donat S, et al: Bladder
colon conduit for supravesical urinary tract diversion. preservation and continent urinary diversion in T3b
Urology. 1976; 8:542. transitional cell carcinoma of the bladder. Semin Urol
113. Althausen A, Hagen-Cook K, Hendren WI: Oncol 1996; 14:103.
Nonrefluxing colon conduit: experience with 70 cases. 133. Hart S, Skinner E, Meyerowitz B, et al: Quality of life
J Urol 1978; 120:35. after radical cystectomy for bladder cancer in patients
114. Matsuura T, Tsujihashi H, Park Y, et al: Assessment of with an ileal conduit, cutaneous or urethral Koch pouch.
the long-term results of ileocecal conduit urinary J Urol 1999; 162:77.
diversion. Urol Int 1991; 46:154–158. 134. Bjierre B, Johansen C, Steven K, et al: Health-related
115. Schwarz G, Jeffs R: Ileal conduit urinary diversion in quality of life after cystectomy: bladder substitution
children: computer analysis of follow-up from 2 to compared with ileal conduit diversion: a questionnaire
26 years. J Urol 1975; 114:285. survey. Br J Urol 1995; 75:200.
116. Shapiro S, Lebowitz R, Colodny A: Fate of 90 children 135. McGuire M, Grimaldi G, Grotas J, et al: The type of
with ileal conduit urinary diversion a decade later: urinary diversion after radical cystectomy significantly
analysis of complications, pyelography, renal function impacts on the patient’s quality of life. Ann Surg Oncol
and bacteriology. J Urol 1975; 114:289. 2000; 7:4.
117. Dretler S: The pathogenesis of urinary tract calculi 136. Fujisawa M, Isotani S, Gotoh A, et al: Health-related
occurring after ileal conduit diversion. I. Clinical study. quality of life with orthotopic neobladder versus ileal
II. Conduit study. III. Prevention. J Urol 1973; 109:204. conduit according to the SF-36 survey. Urology 2000;
118. Terai A, Arai Y, Kawakita M, et al: Effect of urinary 55:862.
intestinal diversion on urinary risk factors for 137. Dutta S, Chang S, Coffey C, et al: Health related quality
urolithiasis. J Urol 1995; 153:37. of life assessment after radical cystectomy: comparison of
119. Wolf JJ, Stoller M: Management of upper tract calculi in ileal conduit with continent orthotopic neobladder.
patients with tubularized urinary diversions. J Urol 1991; J Urol 2002; 168:164.
145:266. 138. Hobisch A, Tosun K, Kinzl J, et al: Life after cystectomy
120. Savarirayan F, Dixey G: Syncope following and orthotopic bladder versus ileal conduit urinary
ureterosigmoidostomy. J Urol 1969; 101:844. diversion. Semin Urol Oncol 2001; 19:18.
Chapter 23 Noncontinent and Continent Cutaneous Urinary Diversion 441

139. Sanchez de Badajoz E, del Rosal Samaniego J, Gomez 144. Kozminski M, Partamian KO. Case report of
Gamez A, et al: Laparoscopic ileal conduit. Arch Esp laparoscopic ileal loop conduit. J Endourol 6:147-150,
Urol 1992; 45:761. 1992
140. Fergany A, Gill I, Kaouk J, et al: Laparoscopic 145. Sanchez de Badajoz E, Gallego Perales JL, Reche
intracorporeally constructed ileal conduit after porcine Rosado A, et al. J Endourol 9:59-62, 1995
cystoprostatectomy. J Urol 2001; 166:285. 146. Vara-Thorbeck C, Sanchez de Badajoz E. Laparoscopic
141. Potter S, Charambura T, Adams JN, et al: ileal loop conduit. Surg Endosc 8:114-5, 1993
Laparoscopic ileal conduit: five year follow-up. 147. Gill IS, Fergany A, Klein EA, et al: Laparoscopic radical
Urology 2000; 56:22. cystoprostatectomy with ileal conduit performed
142. Satoh S, Sato K, Habuchi T, et al: Health-related quality completely intracorporeally: the initial 2 cases. Urology
of life of ileoceccal rectal bladder compared with ileal 2000 56(1): 26-29.
conduit diversion: a questionnaire survey. 2002; 9: 385. 148. Gupta NP, Gill IS, Fergany A, et al. Laparoscopic radical
143. Hara I, Miyake H, Hara S, et al: Health-related quality cystectomy with intracorporeal ileal conduit diversion:
of life after radical cystectomy for bladder cancer: a five cases with a 2-year follow-up. BJV Int 90:391-396,
comparison of ileal conduit and orthotopic bladder 2002.
replacement. 2002; 89:10.
C H A P T E R

24 Orthotopic Bladder Substitution


in the Male and Female
Celi Varol, MD, Fiona C. Burkhard, MD,
Werner W. Hochreiter, MD, and Urs E. Studer, MD

Orthotopic bladder substitutions are here to stay and have sexes.3,4 To achieve such results, certain essential factors
become the main option following cystectomy. need to be considered during specific time periods. These
Improvement in cosmetic results and documented are segregated in this chapter into the preoperative, oper-
increases in quality-of-life outcomes will encourage more ative, and postoperative periods.
urologists to perform such surgical procedures.1,2
Excellent long-term results following orthotopic bladder
substitution can be attained, provided that attention is PREOPERATIVE PATIENT SELECTION
given to appropriate preoperative patient selection, spe- AND ASSESSMENT
cific intraoperative surgical details, and, most importantly, The preoperative assessment is identical to that for any
meticulous postoperative patient follow-up. The general radical cystectomy case, where bone, lung, and lymph
principles of patient selection and follow-up apply to both node metastases must be excluded. Besides establishing
sexes with only the obvious anatomic and operative varia- patient operability, major liver, renal, bowel, or mental
tions to be taken into account in each case. Although blad- insufficiency must be recognized, as these may be con-
der substitutions in men have been widely performed over traindications for a bladder substitution.5
the past two decades and are an accepted mode of urinary
diversion, trepidation still exists when considered in
women. Currently, the majority of cystectomies per- Mental Capacity and Compliance
formed in women are for malignant bladder disease, where A successful outcome of a bladder substitution requires
the risk of metachronous or synchronous urethral transi- the cooperation of the patient and the willingness to
tional cell tumor recurrence and voiding dysfunction are comply with long-term follow-up. Adequate mental
the main factors preventing this from being a routine pro- capacity to comprehend the basic function of the bladder
cedure. With evolving data over the last decade, a less cau- substitute with sufficient physical dexterity is needed.
tious approach to female bladder substitution is The patient needs to be educated to ensure satisfactory
developing. The rarity of urethral recurrence reported and longevity of the bladder substitute. If this is not possible
a better understanding of the natural history of transitional or a question of compliance exists, an alternative form of
cell carcinoma (TCC) has led to more appropriate patient treatment should be discussed. A dedicated nurse who
selection in women undergoing bladder substitution sur- specializes in the education and rehabilitation of patients
gery. Advances in surgical techniques together with with bladder substitutions should be routinely involved
increased understanding of the pelvic anatomy, general in the preoperative selection process.
physiology, and female continence mechanism have
resulted in improved functional outcomes emulating those
in men. The ileal low pressure bladder substitute with an Renal Function
afferent tubular segment is easily constructed and has doc- The commonest biochemical abnormalities detected fol-
umented good long-term functional results in both lowing bladder substitution are metabolic acidosis and

443
444 Part IV Bladder

electrolyte abnormalities. The frequency and severity biopsies from the paracollicular region in the prostatic
observed depends on the length and type of intestinal urethra or the bladder neck in women (site of urethral
segment used, duration of urine contact with the bowel, anastomosis) are associated with a high likelihood of a
and the compensatory functional reserve of the kidneys. urethral recurrence. These patients require a urethrec-
Preoperative renal function with a serum creatinine level tomy and should not be considered for an orthotopic
of ≤150 μmol/l is required to limit such complications. bladder substitute. Infiltration by urothelial tumors
Malignant ureteric obstruction or any other mechanical (superficial or stromal) of the proximal prostatic urethra,
causes of renal insufficiency, where improvement of renal carcinoma in situ, and multifocal transitional cancer in
function can be expected with surgery, may be taken into the bladder confer a higher chance of urethral recur-
account.6,7 rence.11,14 These findings are not absolute contraindica-
tions to the performance of a bladder substitution but
require close follow-up with urethral wash cytology.
Hepatic Function
Management of urethral recurrence with CIS can be per-
To maintain a metabolic equilibrium following surgery formed with BCG therapy, attaining a success rate of
adequate liver function is needed in bladder substitute approximately 80%. These results, however, cannot be
candidates. The bowel mucosa of the bladder substitute replicated in the presence of papillary or invasive urethral
allows ammonium from the urine to shift into the circu- tumor recurrence where urethrectomy is advocated.14
lation. In a urinary tract infection with a urease splitting
organism, the ammonium load becomes even more
Continence
important. In the presence of liver insufficiency hyper-
ammonemia can result in dehydration, neurologic disor- Identification of incontinence prior to surgery, especially
ders and eventual coma.8 in females is essential. Urge incontinence is likely to be
treated by cystectomy; however, stress incontinence,
which is due to inadequate sphincter function, needs to
Bowel Function
be further evaluated by urodynamic assessment with an
The gastrointestinal tract allows up to 60 cm of ileal urethral pressure profile. Patients with a grade I genuine
resection without major repercussions, provided that the stress incontinence, a normal functional urethral length,
ileocecal valve remains intact. When utilizing longer ileal a hypermobile bladder neck and absence of a large cysto-
segments of 60 to 100 cm or the ileocecal valve, long- cele should benefit from a bladder substitute. However, if
term bowel motility disturbances may result.9 Therefore, there is documented evidence for a shortened functional
if ileum is resected, an attempt should always be made to urethral length or intrinsic sphincter deficiency, inconti-
preserve the terminal ileum and the ileocecal valve, as nence following an orthotopic bladder substitute can be
this not only minimizes the risk of bowel dysfunction but expected.
also maintains vitamin B12 and bile salt metabolism.
Evidence of prior bowel disease, resection or radiation
General Preoperative Patient Preparation
needs to be elucidated as it results in functional bowel
shortening causing malabsorption or diarrhea when fur- On the evening prior to the operation, a liquid diet is
ther bowel is used for a bladder substitute. Preservation implemented with two high colonic enemas. In contrast,
of the right colon allows an extended length of left colon colonic bladder substitutes require a full mechanical
to be used for reconstruction with lesser malabsorption bowel preparation (Glycoprep). Application of compres-
side effects. This is because fluid reabsorption occurs sion stockings and subcutaneous deep venous thrombosis
predominantly in the right colon, whilst the left colon prophylaxis should be commenced the evening before
serves more as a conduit. However, even with right colon surgery. Injections should be given in the upper body to
preservation, metabolic complications will still occur.8,10 prevent pelvic lymphocele formation.

Anastomotic Margin Biopsy and Urethral OPERATIVE TECHNIQUE


Recurrence:
To achieve a good functional result, the bladder substitu-
Urethral tumor recurrence is an uncommon phenome- tion has to have a good capacity, a low-pressure system
non following bladder substitution, with a reported inci- made of detubularized bowel segments with minimal
dence of 2% to 4%.11–14 The main factor appears to be outlet resistance and preserved sphincter function. There
the closeness of the primary tumor in the bladder to the are certain critical surgical steps that need to be followed
urethral sphincter. In men, the prostatic urethral length during the cystectomy to achieve these properties. These
allows for more separation from the bladder tumor than steps vary slightly between the genders in regards to the
in women, when the tumor is in the trigone. Positive obvious pelvic anatomy and the position of the respective
Chapter 24 Orthotopic Bladder Substitution in the Male and Female 445

neurovascular bundles that supply the pelvic floor and the paraprostatic neurovascular bundles. At the seminal
the urethral sphincter. The general principles, however, vesicles, minimal trauma should occur by dissecting close
remain the same in the performance of the cystectomy to them on the lateral wall in an antegrade fashion, away
with the construction of the bladder substitute not being from the pelvic plexus, without touching it. The pelvic
particular to either sex. The following points should be plexus will be located dorsolateral at this point and will
noted carefully during the cystectomy and formation of continue onto the dorsomedial border of the prostatic
the bladder substitute for optimal surgical outcome. capsule. If the dorsomedial pedicle of the bladder is sev-
ered too far dorsally, the nerves are at risk of being tran-
sected here.17 A nerve-sparing prostatectomy should be
Cystectomy
performed by freeing the dorsolateral neurovascular bun-
Nerve Preservation
dle off the prostatic capsule. On the contralateral tumor
Preservation of the autonomic nerves to the sphincter bearing side, the dorsomedial pedicle of the bladder
and urogenital diaphragm is controversial, but we feel should be resected along the pararectal/presacral plane
that it should be attempted on the nontumor bearing side and the prostatic neurovascular bundles removed with
(in bladder cancer) as it will increase the chance of the prostate if necessary.
potency in the male and aid urinary continence in both A female nerve-sparing cystectomy requires similar
sexes.15 These nerves originate from the inferior attention during ligation of the dorsomedial pedicle of
hypogastric plexus and continue as the pelvic plexus lying the bladder. Care is taken during the dissection of the lat-
dorsal to the bladder in the pararectal/paravaginal area, eral vagina as the neurovascular bundles lie in the lateral
ending as the paraprostatic neurovascular bundle or paravaginal wall. The dissection needs to be in the
paravaginal plexus prior to supplying the urogenital anterolateral paravaginal plane, close to the bladder base
diaphragm, sphincter, and erectile organs. Probably on the nontumor bearing side. This can be facilitated by
equally important is the preservation of the intrapelvic the use of sponge-holding forceps in the vagina. On the
branch of the pudendal nerve.16 In benign pathology, tumor bearing side, the preparation goes along the dor-
bilateral nerve-sparing cystectomy should be performed solateral aspect of the vaginal wall where the anterior
with simultaneous prostatic capsule preservation in the vaginal wall is removed en bloc with the cystectomy spec-
male. imen (Figure 24-1). The proximal part of the paravaginal
In the male, the critical areas to preserve the pelvic plexus will therefore be severed here (Figure 24-2). The
plexus and neurovascular bundles are in the dorsomedial intrapelvic branches of the pudendal nerve that also
pedicles of the bladder, lateral to the seminal vesicles, and innervate the external sphincter mechanism pass under

Figure 24-1 The preparation goes along the dorsolateral aspect of the vaginal wall where
the anterior vaginal wall is removed en bloc with the cystetectomy specimen.
446 Part IV Bladder

the endopelvic fascia along the medial aspect of levator


ani.18 Therefore, minimal trauma to the endopelvic fas-
cia should occur by only incising it in a ring-like fashion
at the junction of the urethra and bladder neck.
On the contralateral tumor bearing side, the paravagi-
nal tissue is widely excised as far as the pararectal region
to remove the lymphatics that drain the bladder base
together with the autonomic nerves (see Figure 24-10).

Atraumatic Dissection of The Urethra


Sharp, atraumatic dissection of the urethra with limited
use of only bipolar electrocautery at the prostatic apex in
men and bladder neck in females is essential. Maximum
urethral length must be retained. Preservation of the pub-
oprostatic and pubourethral ligaments and incision of the
endopelvic fascia along the bladder neck in female patients
will allow for more urethral stability and improved conti-
nence.19 In women, preserving the autonomic nerve sup-
ply to the proximal one-third of the urethra will also
maintain its tubular shape. If this is denervated, a flaccid,
hypotonic urethra results, which tends to kink. If in doubt
of the innervation, the proximal one-third of the urethra
should be resected. This may result in dribble inconti-
nence when walking due to the shortened functional ure-
thral length. The pudendal nerve still innervating the
distal two-thirds of the urethra and pelvic floor will main-
tain continence at times of sudden increased abdominal
pressure, such as during coughing.19,20 Figure 24-2 The proximal part of the paravaginal plexus is
severed in a ring-like fashion at the junction of the urethra
and bladder neck.
Ureters
The ureters need to be mobilized en bloc with all their
paraureteric tissue. This allows for preservation of the ileum segment is measured along the border of the ileal
blood supply to the proximal ureter, whilst division of the mesentery without stretching the bowel. The distal
ureters at the level of the iliac bifurcation, allows en bloc mesenteric incision transects the main vessels, whereas
removal of the distal ureter and paraureteric lymphatic the proximal incision must be shorter in order to pre-
vessels with the cystectomy specimen. The left ureter is serve the main vessels perfusing the future reservoir seg-
mobilized retrocolic and superior to the inferior mesen- ment (Figure 24-3). A 4-0 polyglycolic acid single layer
teric artery when transposed to the paracaval side, with- seromuscular running suture is used to restore the bowel
out kinking and free of tension. Ureteric anastomoses continuity in a cephalad position, as well as close its
should not be obstructive in nature and antireflux mesenteric window. Both ends of the isolated ileal seg-
ureteric reimplantation is not required in a low pressure ment are closed with a single layer 4-0 polyglycolic acid
ileal bladder substitute.21,22 seromuscular running suture. The distal 40- to 44-cm
end of the ileal segment is opened along its antimesen-
teric border (Figure 24-4).
Orthotopic Ileal Bladder Substitute
Ileal Segment Resection
Ureteroileal Anastomosis
The intraoperative epidural anesthesia (containing local
anesthetics) will cause bowel contraction and, therefore, The ureters are spatulated 1.5 to 2 cm and anastomosed
must be switched off at least 1 hour prior to the meas- by two 4-0 polyglycolic acid running sutures using the
urement of bowel length. This is to ensure accurate Nesbit technique in an end-to-side fashion to two longi-
bowel length measurement resulting in ideal reservoir tudinal 1.5- to 2-cm long incisions along the paramedian
dimension. A 54- to 56-cm segment of ileum is isolated antimesenteric border of the afferent tubular ileal seg-
25 cm proximal to the ileocecal valve. The length of the ment (Figure 24-5). This segment is 12 to 14 cm in
Chapter 24 Orthotopic Bladder Substitution in the Male and Female 447

Figure 24-3 Prepare 54- to 56-cm long ileal segment for the bladder substitute. Note the
different incision depths of the ileal mesentery proximally and distally, in order to preserve the
blood supply.

length. The distal ureteric adventitia is sutured to the acid seromuscular layer. The base of the “U” is folded
afferent ileal segment to remove tension on the anasto- over between the two limbs of the “U” (see Figure 24-6),
mosis and to cover it. The ureters are stented with 8 F resulting in a spherical reservoir consisting of four cross-
catheters. To prevent dislocation of the catheters, a rap- folded ileal segments. After closure of the lower half
idly absorbable 4-0 polyglycolic acid suture is placed of the anterior wall and most of the upper half, the
through the ureter and catheter together and loosely surgeon’s finger is introduced through the remaining
tied, not compromising the ureteric blood flow (see opening to determine the most caudal part of the reser-
Figure 24-5). The ureteric catheters are passed through voir. An 8- to 10-mm diameter hole is cut out of the
the wall of the most distal end of the afferent tubular seg- pouch wall avoiding the suture line (Figure 24-7). The anas-
ment, where it is covered by the fat of the mesentery tomoses must sit flat with the pelvic floor in the male
(Figure 24-6). This provides a “covered” canal in the and the pelvic floor/anterior vaginal wall in the female.
reservoir wall following withdrawal of the ureteric A funnel-shaped outlet, which would kink, must at all
catheters. times be avoided as it may result in outlet obstruction
(Figure 24-8).
Six 2-0 polyglycolic acid seromuscular sutures are
Construction of The Bladder Substitute
placed between the hole in the reservoir wall and the
and Urethral Anastomosis
membranous urethra (Figure 24-9). An 18 F urethral
For the reservoir construction, the two medial borders catheter is inserted before tying the six sutures. A 10 F
of the opened U-shaped distal part of the ileal segment cystostomy tube is placed into the reservoir through its
are oversewn with a single continuous 2-0 polyglycolic mesenteric fat prior to total closure of the reservoir
448 Part IV Bladder

Figure 24-4 Close the mesenteric window, avoiding deep sutures in the mesentery of the
terminal ileum to that of the mesentery of the bladder substitute, in order not to compromise
circulation. The distal 40- to 44-cm end of the ileal segment is opened along its
antimesenteric border.

(Figure 24-10). One 20 F silicon drain is placed into the a nasogastric tube, urethral catheter, suprapubic catheter,
pelvis and one near the ureteric anastomosis. wound drains, and two ureteric catheters. We use single
‘J’ or ureteric catheters (8 F) to stent the ureteric anasto-
moses that are exteriorized through the bladder substi-
POSTOPERATIVE MANAGEMENT
tute. This minimizes urine contact and absorption with
Good long-term results can only be achieved by vigilant the bladder substitution mucosa and allows accurate fluid
postoperative management and regular long-term follow- balance measurements. The suprapubic and transurethral
up. This will allow potential complications to be recog- catheters need to be gently flushed but vigorously aspi-
nized early and minimize morbidity to the patient and the rated with saline 0.9% every 6 hours. This is mandatory
bladder substitute. The in-hospital postoperative period to prevent any mucous clots and subsequent catheter
should be utilized for patient education and to help adjust blockages, which may lead to bladder substitution rup-
to their bladder substitute. A bladder substitute that func- ture. This risk is highest when bowel activity returns
tions well should have no infection, no incontinence, no whilst the urethral catheter is still in situ. Abdominal
acidosis, and no or minimal postvoid residual urine.7 bloating can be limited and bowel function return accel-
erated with subcutaneous neostigmine methylsulfate 3 to
6 × 0.5 mg/day commenced on the third day and addi-
Early Postoperative Period
tionally in smokers with nicotine patches. The ureteric
The patients are stationed in a high dependency ward for catheters are removed sequentially on the 5th to 8th
the first 24 to 48 hours. Postoperatively all patients have postoperative days. Total parenteral nutrition (without
Chapter 24 Orthotopic Bladder Substitution in the Male and Female 449

Figure 24-5 Perform a simple end-to-side Nesbit technique with a 4-0 polyglycolic acid
running suture for the ureteroileal anastomosis.

lipid supplementation) is commenced and tapered once of an alarm clock at night. The normal sensory voiding
an oral diet is reintroduced. Patients undergo regular reflex is lost following a cystectomy, requiring the patient
chest physiotherapy and are encouraged to mobilize on to initially wake up twice at night. Using two alarm
the first postoperative day. clocks allows for a less interrupted sleep. Voiding needs
to occur by relaxation of the pelvic floor, followed by
slight abdominal straining. This is aided by hand pres-
Late Postoperative Period
sure on the lower abdomen and bending forwards.
A cystogram is performed on the 8th to 10th postopera- Initially, effectiveness of reservoir emptying is monitored
tive day. If this excludes any urinary extravasations, the with in-out catheterization and ultrasound to detect any
suprapubic catheter is removed first, followed by the ure- postvoid residual urine. This can later then be monitored
thral catheter 2 days later. This allows for the puncture by suprapubic ultrasound alone. Effective sphincter
site from the suprapubic catheter in the bladder substi- training is taught by performing a digital rectal examina-
tute to seal. A urine sample should be sent for culture tion and requesting the contraction of the anal sphincter
when the urethral catheter is removed. A bladder substi- only and not the abdominal or gluteal muscles. The
tution is not colonized with bacteria like a urinary con- patient reproduces this task initially 10 times hourly,
duit, so any evidence of a bacteriuria or urinary tract maintaining contraction for 6 seconds and as continence
infection must be treated. Following removal of all is achieved on a regular daily basis. This seems to build
catheters, a quinolone antibiotic is implemented as pro- up resting tone and also with repetition, the patient
phylaxis for 5 days. learns to perform it by reflex prior to certain physical
The patient is instructed to void in a sitting position activities, thereby preventing any stress incontinence.
every 2 hours during the day and 3 hourly with the help Men may suffer from postmicturition dribble inconti-
450 Part IV Bladder

Figure 24-6 The two medial borders of the antimesenteric opened U-shaped distal ileum
segment is oversewn with a single layer seromuscular running suture. The base of the “U” is
folded over and tied between the two limbs of the “U” as shown by the arrow.

nence, which can be managed by instructing the patient


Metabolic Management
to “milk” the urethra empty at the end of a void.23 The
voiding interval is gradually increased from 2 to 4 hourly. As a metabolic acidosis is the commonest biochemical
This is performed in hourly aliquots and only when the abnormality following a bladder substitution, the urolo-
patient remains continent for the specified time period gist, as well as the patient, should be aware of the symp-
allocated. (That is, when the patient is continent for 2 toms it can cause in order to be able to rapidly implement
hours, the voiding period is increased to 3 hours. This is therapy (Table 24-1). The presence of fatigue, nausea,
only increased to 4 hours when continence at 3 hours is vomiting, or epigastric burning should alert the urologist
achieved.) The patient must try not to void before the to a metabolic acidosis. H2-blockers or proton pump
allotted time period even if dribble incontinence ensues. inhibitors should not be given for epigastric pain, as they
The aim is to achieve a gradually distended reservoir would worsen the metabolic acidosis. The base excess is
capacity of 500 ml, which will result in a low-pressure monitored with a venous blood gas analysis every second
system and therefore continence. This can be best under- day and immediately corrected if in a negative parameter.
stood by Laplace’s law (pressure = wall tension/radius), This is performed with sodium bicarbonate 2 to 6 g/day
where if the radius of the reservoir increases, the pressure in ileal bladder substitutions. In virtually all patients,
will decrease. As the reservoir pressure decreases below short-term treatment of their metabolic acidosis with
the urethral closing pressure, continence will be attained. NaHCO3 medication will be needed. This is ceased
The time to continence depends on good counseling within 3 months time, as the mucosa of the reservoir
with regular and effective sphincter training, surgical becomes more resilient to electrolyte and fluid exchange.
technique where nerve preservation to the urethra and In the early postoperative period, hypoosmolar urine
pelvic floor is achieved, and the age of the patient. remains in prolonged contact with the mucosa of the
Chapter 24 Orthotopic Bladder Substitution in the Male and Female 451

Figure 24-7 The caudal part of the reservoir is closed completely and the cranial part
almost completely by a single 2-0 polyglycolic acid running seromuscular suture. A finger is
introduced through the remaining opening of the reservoir and an 8- to 10-mm hole is cut
into the most caudal part of the reservoir, close to the mesentery and 2 to 3 cm away from
the suture edge that resulted from cross-folding the ileal segment.

Figure 24-8 The reservoir-urethral anastomosis must be flat and not funnel-shaped to
prevent subsequent kinking and outlet obstruction.
452 Part IV Bladder

Figure 24-9 Six seromuscular 2-0 polyglycolic acid sutures are placed between the
reservoir and the membranous urethra. These are tied after inserting an 18 F urethral
catheter. The catheter is used as a track to guide the reservoir onto the urethra. Cystostomy
and ureteric catheters are placed through the fat of the mesentery.

reservoir. The ileum reaches equilibrium by shifting salt the operative technique but also the meticulous long-
(NaCl) into the reservoir and simultaneously absorbing term regular follow-up that is instituted. Only this way
acid (H+) in the form of ammonium into the blood the optimal reservoir function and prevention of poten-
stream. This will result in a hypovolemic salt loosing tial delayed complications will be recognized and man-
state with an associated metabolic acidosis.8 These aged effectively. A suggested schema is illustrated (Table
patients have a rapid drop in body weight due to dehydra- 24-2). A total of 12% of patients will have positive uri-
tion and anorexia. Daily body weight monitoring is essen- nary cultures.24 Associated causes, such as postvoid resid-
tial in the postoperative recovery period. Patients need to ual urine, need to be identified, and presence of inguinal
consume 2 to 3 l of fluids per day that is supplemented or abdominal hernias that can prevent adequate reservoir
with increased salt intake in their diet to combat any salt- emptying needs to be treated. Protruding mucosa or
losing syndrome.5 stricture at the urethral-reservoir anastomosis may also
be the factors that can be easily managed transurethrally.
Diurnal continence of up to 83% in women and 90%
LONG-TERM FOLLOW-UP
in men with nocturnal rates over 80% at 12 months
The successful results of ileal low-pressure orthotopic should be achieved in both sexes.15 Incontinence rates
bladder substitution are not only due to the simplicity of tend to increase in the absence of a nerve-sparing cystec-
Chapter 24 Orthotopic Bladder Substitution in the Male and Female 453

Figure 24-10 After cystostomy tube placement into the reservoir, the cephalad part of the
pouch is closed completely; 20 F silicon drains are placed into the pelvis and near the
ureteric anastomosis.

Table 24-1 Symptoms of Metabolic Acidosis tomy, in the older patients and 5 years following a cys-
tectomy. The incontinence is generally minimal and not
Fatigue
debilitating with normal transurethral voiding achievable
Anorexia in up to 97% of patients.4
It is also important to recognize that the limited under-
Dyspepsia and heartburn standing of orthotopic bladder substitutions by other
medical professions may result in ignorant decisions
Nausea and vomiting
regarding patient care; therefore, direct referral or con-
Weight loss tact with the initial treating institution is recommended.
454 Part IV Bladder

Table 24-2 Follow-up Schema for Patients with Bladder Substitute


Months After Surgery 3 6 12 18 24 30 36 42 48 54 60

Clinical examination x x x x x x x x x x x

Urine culture x x x x x x x x x x x

Body weight x x x x x x x x x x x

Blood tests* x x x x x x x x x x x

Folic acid, vitamin B12 x x x x

Chest x-ray x x x x x x x

IVU x x x x

Renal ultrasound x x x x x x x

Bone scan (only if x x


≥pT3 and each N+)

Pelvic/abdominal x
CT-Scan (only if
≥pT3 and each N+)

Postvoid residual x x x x x x x x x x x

Urethral wash cytology x x x x x x x

*Hb, Na+, K+, Cl−, bicarb., creatinine, urea, ALP, gamma GT, AST, LDH, and venous blood gas analysis.

REFERENCES 9. Hofmann AF: Bile acid malabsorption caused by ileal


resection. Arch Intern Med 1972; 130:597.
1. Hobisch A, Tosun K, Kemmler G, et al: Life after 10. Proano M, Camilleri M, Phillips SF, Brown ML,
cystectomy and orthotopic neobladder versus ileal conduit Thomford GM: Transit of solids through the human
urinary diversion. Semin Urol Oncol 2001; 19(1):18–23. colon: regional quantification in the unprepared bowel.
2. Yoneda T, Igawa M, Shiina H, Shigeno K, Urakami S: Am J Physiol 1990; 258(6 Pt 1):G856–G862.
Postoperative morbidity, functional results and quality of 11. Freeman JA, Thomas A, Esrig D, et al: Urethral
life of patients following orthotopic neobladder recurrence in patients with orthotopic ileal neobladders.
reconstruction. Int J Urol 2003; 10(3):119–125. J Urol 1996; 156(5):1615–1619.
3. Doherty A, Burkhard F, Holliger S, Studer UE: Bladder 12. Yossepowitch O, Dalbagni G, Golijanin D, et al:
substitution in women. Curr Urol Rep 2001; 2(5):350–356 Orthotopic urinary diversion after cystectomy for
(review). bladder cancer: implication for cancer control and
4. Madersbacher S, Möhrle K, Burkhard F, Studer UE: patterns of disease recurrence. J Urol 2003;
Long-term voiding pattern of patients with ileal 169(1):177–181.
orthotopic bladder substitutes. J Urol 2002; 167(5):2052. 13. Hautmann RE, Miller K, Steiner U, Wenderoth U: The
5. Mills RD, Studer UE: Guide to patient selection and ileal neobladder: 6 years of experience with more than 200
follow-up for orthotopic bladder substitution. Contemp patients. J Urol 1993; 150(1):40–45.
Urol 2001; 2:35–40. 14. Studer UE, Danuser H, Hochreiter W, et al: Summary of
6. Skinner DG, Studer UE, Okada K, et al: Which patients 10 years’ experience with an ileal low-pressure bladder
are suitable for continent diversion or bladder substitution substitute combined with an afferent tubular isoperistaltic
following cystectomy or other definitive local treatment? segment. World J Urol 1996; 14:29–39.
Int J Urol 1995; 2(Suppl):105. 15. Turner WH, Danuser H, MoehrleK, Studer UE: The
7. Studer UE, Burkhard F, Danuser HJ, Thalmann G: Keys effect of nerve sparing cystectomy technique on
to success in orthotopic bladder substitution. Can J Urol postoperative continence after orthotopic bladder
1999; 6(5):876. substitution. J Urol 1997; 156(6):2118.
8. Mills RD, Studer UE: Metabolic consequences of 16. Hugonnet CL, Danuser HJ, Springer JP, Studer UE:
continent urinary diversion. J Urol 1999; 161:1057–1066. Urethral sensitivity and the impact on urinary continence
Chapter 24 Orthotopic Bladder Substitution in the Male and Female 455

in patients with an ileal bladder substitute after an afferent tubular segment detrimental to the upper
cystectomy. J Urol 2001; 165:1502–1505. urinary tract in the long term? J Urol 2002;
17. Burkhard FC, Studer UE: Orthotopic bladder 168(5):2030–2034.
substitution. Curr Opin Urol 2000; 10(4):343–349. 22. Studer UE, Siegrist T, Casanova GA, et al: Ileal bladder
18. Borirakchanyavat S, Aboseif SR, Carroll PR, Tanagho EA, substitute: antireflux nipple or afferent tubular segment?
Lue TF: Continence mechanism of the isolated female Eur Urol 1991; 20(4):315–326.
urethra: an anatomical study of the intrapelvic somatic 23. Bader P, Hugonnet CL, Burkhard F, Studer UE:
nerves. J Urol 1997; 158(3 Pt 1):822—826. Inefficient urethral milking secondary to urethral
19. Mills RD, Studer UE: Female orthotopic bladder dysfunction as an additional risk factor for incontinence
substitution: a good operation in the right circumstances. after radical prostatectomy. J Urol 2001;
J Urol 2000; 163(5):1501–1504. 166(6):2247–2252.
20. Doherty A, Burkhard F, Holliger S, Studer UE: Bladder 24. Varol C, Burkhard FC, Thalmann GN, Studer UE:
substitution in women. Curr Urol Rep 2001; Urethral recurrence following cystectomy for bladder
2(5):350–356. cancer; prevention and detection in patients with
21. Thoeny HC, Sonnenschein MJ, Madersbacher S, Vock P, orthotopic bladder substitutes. J Urol 2003; 169(Suppl
Studer UE: Is ileal orthotopic bladder substitution with 4):103.
C H A P T E R

25 Cancer of the Prostate: Detection


and Staging
Anthony V. D’Amico, MD, PhD,
and Michael W. Kattan, PhD

The goal of a staging system is to predict cancer-specific ing system is limited in its ability to stratify patients into
survival as accurately as possible using readily available clinically distinct groups based on cancer-specific out-
pretreatment parameters. These parameters define stages comes.
that correspond to rates of disease-specific survival fol-
lowing standard therapy that increase in a clinically sig-
PROGNOSTIC FACTORS
nificant manner as the clinical stage decreases. Validated
Combined Modality Staging
algorithms1–3 currently exist that provide accurate esti-
mates of prostate-specific antigen (PSA) failure based on Prior studies1,2 have confirmed that the serum PSA level,
pre-treatment clinical parameters following surgery or 2002 AJCC clinical T-category, and biopsy Gleason score
radiation therapy (RT) for patients with clinically local- are independent predictors of time to PSA failure follow-
ized disease. However, PSA failure may not translate into ing either radical prostatectomy (RP) or external beam RT.
death from prostate cancer for all patients because men Combined modality staging was developed to utilize these
with prostate cancer are generally over the age of 60 and independent and clinically significant predictors of PSA
often have competing causes of mortality.4 Therefore, outcome following definitive therapy to define prognostic
current efforts are aimed at defining a staging system groups. Risk groups based on the PSA, biopsy Gleason
based on pretreatment factors that accurately represent score, and the 2002 AJCC clinical T-category that stratify
rates of prostate cancer-specific mortality (PCSM) fol- the risk of PSA failure following RP or RT are illustrated
lowing current standard therapies. in Figures 25-1 and 25-2, respectively.3 This risk stratifi-
cation algorithm, which was originally defined based on
PSA failure rates, has also been shown to stratify time to
AMERICAN JOINT COMMISSION ON CANCER
PCSM following RT using simply the PSA and biopsy
STAGING SYSTEM
Gleason score as illustrated in Figure 25-3.7
Historically, staging defines the anatomic extent of a dis- Recently, a fourth parameter, the percent positive
ease and tumor volume remains the most important sin- prostate biopsies, has been shown in a validated prognos-
gle determinant of prognosis for many cancers. The tic factor analysis to provide clinically useful information
current 2002 American Joint Commission on Cancer regarding PSA outcome following either RP8 or external
(AJCC) staging system5 for patients with adenocarci- beam RT9 for intermediate risk patients as shown in
noma of the prostate is based solely on the findings of the Figures 25-4 and 25-5, respectively. This parameter has
digital rectal examination (DRE) or transurethral resec- also been shown to be predictive of time to PCSM fol-
tion of the prostate (TURP). However, the introduction lowing RT10 in the intermediate risk group, forming the
of the serum PSA has changed the presentation of basis for a 2-tiered risk group stratification as shown in
prostate cancer worldwide. The vast majority of patients Figure 25-6.
now present with nonpalpable PSA detected (i.e., clinical Molecular markers (such as p53, bcl-2, Ki-67, and
category T1c) disease.6 By grouping most of today’s c-erb-b2), neuroendocrine markers (such as neuron-specific
patients into a single T1c-category, the 2002 AJCC stag- enolase and chromogranin A), histopathologic markers

459
100 100

Prostate cancer specific survival


90 90
% bNED Survival 80 80
70 70
60 60
50 50
40 40
30 30
Low risk Low risk
20 Intermediate risk 20 Intermediate risk
10 High risk 10 High risk
0 0
0 1 2 3 4 5 6 7 8 9 10 0 1 2 3 4 5 6 7 8 9 10
Time in years Time in years

1020 704 422 156 51 13 110 106 92 72 56 38 28 15 4 2 1


693 407 203 99 29 6 155 147 125 93 66 44 25 12 4 3 3
414 198 91 35 9 1 150 144 130 107 71 49 28 12 7 5 4

Number at risk Number at risk

Figure 25-1. Prostate-specific antigen outcome following Figure 25-3. Prostate cancer-specific survival following
radical prostatectomy stratified by the pretreatment risk external beam radiation therapy stratified by the pretreatment
group for patients with clinically localized disease. Low risk: risk group for patients with clinically localized disease. Low
PSA ≤ 10 ng/ml and biopsy Gleason ≤ 6, and 2002 AJCC risk: PSA ≤ 10 ng/ml and biopsy Gleason ≤ 6. Intermediate
category T1c or T2a. Intermediate risk: PSA > 10 to 20 ng/ml risk: PSA > 10 to 20 ng/ml or biopsy Gleason 3 + 4. High
or biopsy Gleason 7 or 2002 AJCC category T2b. High risk: risk: PSA > 20 ng/ml or biopsy Gleason ≥ 4 + 3.
PSA > 20 ng/ml or biopsy Gleason 8 to 10 or 2002 AJCC
category T2c.

100
90
80
100
70
PSA 0utcome

90
60
80
50
% bNED survival

70
40
60
30
Low risk 50
20 Intermediate risk 40
10 High risk
30
0 < 34%
20 34−50%
0 1 2 3 4 5 6 7 8 9 10
10 > 50%
Time in years 0
90 88 70 46 34 26 16 5 3 3 2 0 1 2 3 4 5 6 7
173 167 112 69 45 24 10 4 3 2 1 Time in years
118 115 83 47 30 18 14 8 3 1 1
366 309 259 201 149 109 79 153
150 99 75 51 31 19 13 7
Number at risk 163 104 69 44 23 12 7 4
Figure 25-2. Prostate-specific antigen outcome following
external beam radiation therapy stratified by the pretreatment risk Number at risk
group for patients with clinically localized disease. Low risk: PSA
≤ 10 ng/ml and biopsy Gleason ≤ 6, and 2002 AJCC category Figure 25-4. Prostate-specific antigen outcome following
T1c or T2a. Intermediate risk: PSA > 10 to 20 ng/ml or biopsy radical prostatectomy stratified by the percent of positive
Gleason 7 or 2002 AJCC category T2b. High risk: PSA > 20 prostate biopsies for intermediate-risk patients with clinically
ng/ml or biopsy Gleason 8 to 10 or 2002 AJCC category T2c. localized disease.
Chapter 25 Cancer of the Prostate: Detection and Staging 461

Prostate cancer-specific survival (%)


100 100
90 90
80 80
% bNED survival

70 70
60 60
50 50
40 40
30 30
< 34% Iog rank p < 0.0001
20 34−50% 20 Low + Int  50%
10 > 50% 10 High + Int > 50%
0 0
0 1 2 3 4 5 0 1 2 3 4 5 6 7 8 9 10 11 12
Time in years Time in years

96 83 50 24 12 8 206 155 79 37 6 13
49 45 23 6 4 3 175 153 89 38 12 3
62 59 36 16 8 3

Number at risk
Number at risk
Figure 25-6. Prostate cancer-specific survival following
Figure 25-5. Prostate-specific antigen outcome following external beam radiation therapy stratified by the percent of
external beam radiation therapy stratified by the percent of positive prostate biopsies for intermediate-risk patients with
positive prostate biopsies for intermediate-risk patients with clinically localized disease.
clinically localized disease.

for pelvic lymphadenopathy. A bone scan is generally rec-


ommended to identify metastatic disease and is indicated
(such as microvessel density and ploidy), imaging for patients with either clinical stage T3 or stage T4 dis-
approaches (such as color Doppler), PSA derivatives ease, biopsy Gleason score of 4 + 3 or higher, a PSA level
(such as PSA velocity, PSA density, and free PSA), and greater than 20 ng/ml, or clinical symptoms.
reverse transcriptase polymerase chain reaction (rtPCR) The role of endorectal MRI (erMRI) has been evalu-
to examine PSA-expressing cells in the peripheral blood, ated for patients with clinical stage T1c or stage T2 dis-
bone marrow, and pelvic lymph nodes all have been ease to assess whether information regarding pathologic
examined11–15 to assess their ability to predict PSA out- stage and PSA outcome following RP was provided.18 In
come following definitive local therapy for patients with experienced hands, the erMRI finding of T3 versus T2
clinically localized disease. While many of these factors disease was 80% accurate in predicting pathologic stage.
have been predictive of PSA outcome on univariable However, the erMRI did not add clinically meaningful
analysis, they await testing in a multivariable model that information for the vast majority of the patients (low risk
accounts for the established prognostic factors to deter- and high risk) after accounting for the pretreatment PSA
mine their clinical significance. level, biopsy Gleason score, clinical T-category, and the
percent positive prostate biopsies. In the intermediate
risk patients, however, the erMRI provided a clinically
Radiologic Staging
relevant stratification of 5-year PSA outcome as shown in
The ability of computerized tomography, pelvic coil mag- Figure 25-7. At present, however, outside of high risk
netic resonance imaging (MRI), and transrectal ultra- localized of locally advanced prostate cancer, imaging of
sound (TRUS) to identify extracapsular extension (ECE) the pelvis with erMRI to assess for evidence of pelvic
and/or seminal vesicle invasion (SVI) for patients with lymphadenopathy or evidence of ECE or SVI remains
clinically localized disease based on the DRE is limited. under investigation.
The accuracy of these studies does not exceed 60%,16,17
and therefore none of these studies is recommended for
Pretreatment Nomograms
staging patients with clinical stage T1 or stage T2 disease.
Patients with more advanced disease (stage T3 or stage A popular tool for predicting outcomes in prostate can-
T4) as determined by DRE should have a computed cer is the nomogram. Strictly speaking, a nomogram is a
tomographic (CT) scan or MRI scan of the pelvis to assess series of lines with point values, which one manipulates
462 Part V Prostate Gland and Seminal Vesicles

100 nomograms have been developed and validated for pre-


90 dicting biochemical failure for patients treated with sur-
gery, brachytherapy, and external beam RT.3,21–7 They
80
are presented in Figures 25-8 to 25-10. They predict bio-
% bNED Survival

70
chemical failure, and future nomograms are necessary for
60 predicting more distant and clinically relevant endpoints,
50 such as metastasis and death. These prediction models
40 are available in software for the palm and desktop com-
30 puters from http://www.nomograms.org.
MR T2
20 MR T3
10 SUMMARY
0
The 2002 AJCC staging system is limited in its ability to
0 1 2 3 4 5 provide accurate information regarding time to PCSM
Time in years for individual patients who present with the most com-
mon clinical category of T1c disease. However, algo-
162 137 100 64 46 28 rithms for predicting PSA outcome following RP or
29 19 14 4 4 4 external beam RT that are based on pretreatment clini-
cal parameters that include the PSA level, biopsy
Number at risk Gleason score, and 2002 AJCC clinical T-category have
been validated.1–3 Nevertheless, given the competing
Figure 25-7. Prostate-specific antigen outcome following causes of mortality that exist in men undergoing defini-
radical prostatectomy stratified by the erMRI T-category for tive treatment for localized prostate cancer, many men
intermediate-risk patients with clinically localized disease. who sustain PSA failure will not live long enough to
develop clinical evidence of distant disease and far fewer
will die from the disease. Although pretreatment risk-
by drawing straight lines to obtain a prediction.19 The based staging systems predicting the endpoint of
term is attributed to Professor Maurice d’Ocagne in PCSM7,26 have been published, none has been validated
1889.20 The primary advantage of nomograms, relative in the PSA era. Studies are currently ongoing to define a
to other paper-based approach, such as tables, is that validated pretreatment staging system that can accu-
nomograms maintain a continuous prediction model, rately predict time to PCSM following surgery or RT
resulting in greater predictive accuracy. Pretreatment for prostate cancer.

Preoperative nomogram for PSA recurrence


0 10 20 30 40 50 60 70 80 90 100
Points

PSA
0.1 1 23 4 6 7 8 9 101 2 16 20 30 45 70 110
T2a T2c T3a
Clinical stage
T1c T1ab T2b

2+3 3+2 4+


Biopsy gleason sum
 2+2 3+3 3+4
Total points
0 20 40 60 80 100 120 140 160 180 200
60-month recurrence free prob.
.96 .93 .9 .85 .8 .7 .6 .5 .4 .3 .2 .1 .05

Figure 25-8. Pretreatment nomogram for patient considering surgery. (Adapted with
permission from Kattan MW, Eastham JA, Stapleton AMF, et al: J Natl Cancer Inst 1998;
90(10):766–771.)
Chapter 25 Cancer of the Prostate: Detection and Staging 463

3D conformal radiation therapy nomogram for PSA recurrence

0 10 20 30 40 50 60 70 80 90 100
Points

Pretreatment PSA
0.3 1 2 3 4 5 6 7 910 25 50 100
T2a T3ab T3c
Clinical stage
T1c T2b T2c
357 9
Bx. gleason sum
24 6 8 10

Dose (Gy)
86.4 72 68 64.8
No
Hormones
Yes
Total points
0 20 40 60 80 100 120 140 160 180

60 month Rec. free prob.


0.99 0.98 0.95 0.9 0.8 0.7 0.5 0.3 0.1 0.01

Figure 25-9. Pretreatment nomogram for patient considering external beam radiation.
(Adapted with permission from Kattan MW, Zelefsky, Kupelian PA, et al: J Clin Oncol
2000; 18:3352–3359.

Brachytherapy nomogram for PSA recurrence


0 10 20 30 40 50 60 70 80 90 100
Points
0.8
Pretreatment PSA
0.6 1 2 3 4 5 6 8 10 15 20 30 40 60 80 100

42 7
Biopsy GI.Sum
536 8
T2a
97 clinical stage
T2b T1c
No
XRT
Yes
Total points
0 10 20 30 40 50 60 70 80 90 100 110 120 130

60-month Rec. free prob.


0.99 0.98 0.96 0.93 0.9 0.8 0.7 0.6 0.5 0.4 0.25 0.12

Figure 25-10. Pretreatment nomogram for patients considering brachytherapy. (Adapted


with permission from Kattan MW, Potters, L, Blasko JC, et al: Urology 2001;
58(3):393–399.)
464 Part V Prostate Gland and Seminal Vesicles

REFERENCES predictive indicators of prostate carcinoma recurrence


after surgery. Cancer 1998; 82:168–174.
1. Kattan MW, Eastham JA, Stapleton AMF, Wheeler TM, 14. Waltregny D, de Leval L, Menard S, de Leval J,
Scardino PT: A preoperative nomogram for disease Castronovo V: Independent prognostic value of the 67-kd
recurrence following radical prostatectomy for prostate laminin receptor in human prostate cancer. J Natl Cancer
cancer. JNCI 1998; 90:766–771. Inst 1997; 89:1224–1228.
2. D’Amico AV: Combined-modality staging for localized 15. Berruti A, Dogliotti L, Mosca A, et al: Circulating
adenocarcinoma of the prostate. Oncology 2001; neuroendocrine markers in patients with prostate
15:1049–1059. carcinoma. Cancer 2000; 88:2590–2597.
3. Graefen M, Karakiewicz PI, Cagiannos I, et al: A 16. Platt JF, Bree RL, Schwab RE: The accuracy of CT in the
validation of two preoperative nomograms predicting staging of prostatic carcinoma. Am J Radiol 1987;
recurrence following radical prostatectomy. Urol Oncol 149:315–321.
2002; 7:141–146. 17. Rifkin MD, Zerhouni A, Gatsonis CA, et al:
4. From the National Center for Health Statistics: National Comparison of magnetic resonance imaging and
Vital Statistics 2002; 50:1–120. ultrasonography in staging early prostate cancer. Results
5. Greene FL, Page DL, Fleming ID, et al: American Joint of a multi-institutional cooperative trial. NEJM 1990;
Committee on Cancer, Manual for staging cancer, 6th 323:621–629.
edition, pp 337–346. New York, Springer, 2002. 18. D’Amico AV, Whittington R, Malkowicz SB, et al:
6. Catalona WJ, Smith DS, Ratliff TL, Basler JW: Detection Endorectal magnetic resonance imaging as a predictor of
of organ-confined prostate cancer is increased through biochemical outcome following radical prostatectomy for
prostate-specific antigen-based screening. JAMA 1993; men with clinically localized prostate cancer. J Urol 2000;
270:948–954. 164:759–763.
7. D’Amico AV, Cote K, Loffredo M, Renshaw AA, Chen 19. Hankins TL: Blood, dirt and nomograms. Hist Sci Soc
MH: Pre-treatment predictors of time to cancer specific 1999; 90:50–80.
death following prostate specific antigen failure. J Urol 20. Banks J. Nomograms, Vol 6. New York, Wiley, 1985.
2003; 169(4):1320–1324. 21. Kattan MW, Eastham JA, Stapleton AMF, Wheeler TM,
8. D’Amico AV, Whittington R, Malkowicz SB, et al: Scardino PT: A preoperative nomogram for disease
Clinical utility of the percentage of positive prostate recurrence following radical prostatectomy for prostate
biopsies in defining biochemical outcome after radical cancer. J Natl Cancer Inst 1998; 90(10):766–771.
prostatectomy for patients with clinically localized 22. Kattan MW, Potters L, Blasko JC, et al: Pretreatment
prostate cancer. J Clin Oncol 2000; 18:1164–1172. nomogram for predicting freedom from recurrence after
9. D’Amico AV, Schultz D, Silver B, et al: The clinical utility permanent prostate brachytherapy in prostate cancer.
of the percent positive prostate biopsies in predicting Urology 2001; 58(3):393–399.
biochemical outcome following external beam radiation 23. Kattan MW, Zelefsky MJ, Kupelian PA, et al:
therapy for patients with clinically localized prostate Pretreatment nomogram for predicting the outcome of
cancer. Int J Radiat Oncol Biol Phys 2001; 49:679–684. three-dimensional conformal radiotherapy in prostate
10. D’Amico AV, Keshaviah A, Manola J, et al: The clinical cancer. J Clin Oncol 2000; 18:3352–3359.
utility of the percent of positive prostate biopsies in 24. Graefen M, Karakiewicz P, Cagiannos I, et al: A validation
predicting prostate cancer specific and overall survival of two preoperative nomograms predicting recurrence
following radiation therapy for patients with localized following radical prostatectomy in a cohort of European
prostate cancer. Int J Radiat Oncol Biol Phys 2002; men. Urol Oncol 2002; 7(4):141–146.
53:581–587. 25. Graefen M, Karakiewicz PI, Cagiannos I, et al:
11. Ismail MT, Petersen RO, Alexander AA, Newschaffer C, International validation of a preoperative nomogram for
Gomella LG: Color Doppler imaging in predicting the prostate cancer recurrence following radical
biologic behavior of prostate cancer: correlation with prostatectomy. J Clin Oncol 2002; 20(15):3206–3212.
disease-free survival. Urol 50:906–912. 26. Roach M, Lu J, Pilepich MV, et al: Four prognostic
12. Yang RM, Naitoh J, Murphy M, et al: Low P27 groups predict long-term survival from prostate cancer
expression predicts poor disease-free survival in patients following radiotherapy alone on radiation therapy
with prostate cancer. J Urol 1998; 159:941–945. oncology group clinical trials. Int J Radiat Oncol Biol
13. Stapleton AM, Zbell P, Kattan MW, et al: Assessment of Phys 2000; 47:609–615.
the biologic markers p53, Ki-67, and apoptotic index as
C H A P T E R

26AClinically Localized (Stage T1a-T2c)


Adenocarcinoma of the Prostate:
Surgical Management and Prognosis
Maxwell V. Meng, MD, and Peter R. Carroll, MD

Natural History of Prostate Cancer


Prostate cancer remains a major health concern in the
U.S. and throughout the world. The institution of Ultimately, the role and necessity of surgical removal of
screening protocols, based on the combination of dig- the prostate from men with cancer can only be deter-
ital rectal examination and serum prostate specific mined in appropriately designed clinical trials. The most
antigen (PSA) testing, has increased the opportunity relevant endpoint of these studies is the impact of prosta-
for cancer cure by allowing earlier diagnosis at lower tectomy on overall survival. Prior published reports eval-
stages of disease. Over the past decade, advances in uating various prostate cancer treatments are not
both surgical and therapeutic radiation therapy tech- applicable to contemporary patients. Currently, there are
niques have revolutionized the ability to adequately several ongoing randomized trials seeking to compare
treat prostate cancer while simultaneously reducing active therapy with watchful waiting.3 The results of a
treatment-related morbidity. This chapter focuses on recent study from the Scandinavian International Union
the management and outcomes of patients with clini- against Cancer provide evidence that radical prostatec-
cally localized (stages T1a to T2c) cancer of the tomy significantly reduces disease-specific mortality.4
prostate. A total of 695 men with clinical stages T1b, T1c, or T2
prostate cancer were randomized to either watchful wait-
ing or prostatectomy. During a median follow-up of 6.2
RADICAL PROSTATECTOMY
years, there was no difference between the two groups
Rationale for Treatment
with respect to overall survival; however, death due to
Proponents of screening for prostate cancer cite the prostate cancer in the surveillance cohort (8.9%) was
disease prevalence and mortality, ability to effectively greater than that observed in the surgery cohort (4.6%, p
treat localized cancers, and inability to cure metastatic = 0.02). In addition, men after surgery had a lower risk of
disease as compelling reasons for this practice.1 Despite distant metastases (hazard ratio 0.63). Although there
the numerous factors supporting early prostate cancer was no reduction in overall mortality, a difference is
detection, controversy continues to surround the need likely to be noted with longer follow-up given the signif-
for, and specific choice of, intervention.2 This is partic- icant reduction in metastases in those managed with sur-
ularly true for radical prostatectomy. Definitive surgery gery. This important study supports the utility of early
is not only curative for most organ-confined tumors detection and treatment of prostate cancer in selected
(stages T1 to T2) but is also associated with limited patients.
morbidity and is amenable to selective use with adju- Although it appears that surgery for localized prostate
vant therapy in high-risk patients. Nevertheless, the cancer impacts disease-specific outcomes, the rational
potential for operative and postoperative morbidity selection of therapy remains complex, and examination of
exists and surgery may not be necessary in men with other data provides information regarding the natural
lower-risk disease who may be candidates for surveil- history of untreated prostate cancer. Chodak et al.5
lance alone. analyzed the pooled data of 823 men (cT1-2) from six

465
466 Part V Prostate Gland and Seminal Vesicles

nonrandomized studies treated prior to the era of PSA the work of Chodak et al. and calculated survival benefit
testing. The risk of metastases was significant and from surgery. In contrast to the study of Fleming et al.,
dependent on tumor grade—2.1% per year, 5.4% per the estimated survival advantage in men with well differ-
year, and 13.5% per year for well differentiated, moder- entiated, moderately differentiated, and poorly differen-
ately differentiated, and poorly differentiated tumors, tiated tumors increased to 1.0, 2.4, and 2.7 years,
respectively. At 15 years, the fraction of patients with respectively.
metastatic disease was 40%, 70%, and 85% in these
groups, respectively (Table 26A-1).
Selection of Patients for Radical Prostatectomy
Albertsen et al.6 analyzed the long-term outcome of
451 men with clinical stage T1-2 tumors managed with Despite the disparate result of the various publications
immediate or delayed hormonal therapy. Again, tumor and controversy regarding the “best” treatment, it is clear
grade was the important factor determining patient out- that selection of therapy needs to be based on several
come. The cancer-specific mortalities in men with patient and tumor factors. In general, radical prostatec-
Gleason sums 2 to 4, 5 to 7, and 8 to 10 were 9%, 28%, tomy is considered in men with a life expectancy greater
and 51%, respectively. Overall, 46% of men treated expec- than 10 years, a duration during which an untreated can-
tantly living 15 years or more will die of prostate cancer cer may progress and/or metastasize. In addition, the
and lose approximately one-third of their remaining life patients should be free of serious comorbidities and be
expectancy, even at older age of diagnosis (i.e. >75 years).6,7 able to tolerate a major operation. With respect to cancer
Fleming et al.8 attempted to address questions regard- variables, the tumor should be both clinically significant
ing the selection of surveillance or definitive therapy and at a stage where surgical extirpation is likely to be cur-
using decision analysis. In their Markov model, they ative. These aspects of tumor behavior and biology are
incorporated estimates of progression to metastatic dis- often difficult to assess based on traditional measures, such
ease with watchful waiting from review of the literature, as digital rectal examination, serum PSA, and Gleason
efficacy of radical prostatectomy based on pathologic grade. Thus, more accurate determinations of outcome
stage, and arbitrary reductions in survival after surgery have been developed and are currently commonly applied
due to impact on quality of life to determine which strat- to aid in the appropriate selection of men for radical
egy resulted in a greater survival advantage. Based on the prostatectomy, as well as other definitive treatments.
data selected for their model, the authors reported that
surgical intervention for prostate cancer provided limited
Pretreatment Risk Stratification
benefit to the patient, relative to expectant management,
with well-differentiated tumors. In men with moderate The most commonly used risk assessment criteria
or poorly differentiated tumors, they stated that treat- include serum PSA, clinical tumor stage (T stage), and
ment offered less than 1-year improvement in quality- Gleason grade on biopsy. Recently, the extent of disease,
adjusted life expectancy in those men 60 to 65 years old assessed by systematic biopsy (e.g., percent positive biop-
and that treatment was harmful in men over age 70. sies), has been shown to have prognostic significance and
Thus, they conclude that selection of watchful waiting in such information may be incorporated into the assess-
men with localized prostate cancer is a feasible alterna- ment of pretreatment risk. In general, the role of imag-
tive to radical prostatectomy. Application of the model by ing modalities, such as transrectal ultrasonography,
other investigators, using other estimates of disease pro- computed tomography (CT), and magnetic resonance
gression and efficacy of treatment, resulted in much dif- imaging/spectroscopy, is limited in initial risk stratifica-
ferent results. Beck et al.9 utilized progression data from tion, except in those with advanced disease.
Due to widespread screening efforts, men are increas-
ingly diagnosed with early stage disease (i.e., T1c). The
Table 26A-1 Development of Metastases in Men with
Localized Prostate Cancer Treated Conservatively
risk for disease recurrence after radical prostatectomy
increases with higher clinical stage. In men with stage T1c,
PERCENT WITH METASTASES 5-year PSA-free survival after radical prostatectomy is
Histologic
greater than 85%, while those with palpable but localized
Grade 10 Years 15 Years
(cT2) have disease-free rates between 70% and 80%.10
1 19 40 Serum PSA levels can vary within any clinical stage
but usually correlate with tumor volume and, therefore,
2 42 70 pathologic stage and outcome after surgery. As reported
by Ohori and Scardino,11 progression-free survival 5
3 74 85
years after prostatectomy in men with PSA ≤ 4.0 ng/ml,
From Chodak GW, Thisted RA, Gerber GS, et al: N Engl J Med 4.1 to 10 ng/ml, 10.1 to 20 ng/ml, and >20 ng/ml was
2002; 330: 781–789. 94%, 85%, 66%, and 38%, respectively. These values are
Chapter 26A Clinically Localized (Stage T1a-T2c) Adenocarcinoma of the Prostate 467

comparable to those reported by others. In the study (cT1c or T2a and Gleason sum 2 to 6 and PSA <10 ng/ml),
from Catalona et al.,12 7-year progression-free survival intermediate-risk (cT2b, Gleason sum 7, or PSA 10 to
was 93% in men with serum PSA ≤ 2.5 ng/ml, 80% with 20 ng/ml), or high-risk (cT2c or Gleason sum 8 to 10 or
PSA 2.5 to 4.0 ng/ml, 76% with PSA 4.1 to 10 ng/ml, PSA >20 ng/ml).20 In low-risk patients, it should be men-
and 40% with PSA >10 ng/ml. Walsh et al.13,14 reported tioned that excellent outcomes might be achieved with a
10-year rates of 87%, 75%, 30%, and 28% for PSA variety of modalities, including watchful waiting in
groups of <4 ng/ml, 4.1 to 10 ng/ml, 10.1 to 20 ng/ml, selected patients. Although progression occurs slowly
and >20 ng/ml, respectively. within this population, eventual active treatment is likely
Gleason grade, as determined by biopsy, is a significant in men who are young or have elevated PSA levels.21
predictor of outcomes after radical prostatectomy. With High-risk patients can undergo radical prostatectomy
increasing tumor grade, the likelihood of disease recur- with acceptable morbidity and reasonable rates of local
rence increases. Five and 10-year disease-free survival in control; however, long-term cure is less likely and if sur-
patients with Gleason sum 2 to 4 are approximately 90% gery is selected, the patient must realize that adjuvant or
but drop to approximately 60% for those men with sum secondary treatment may be necessary. Although catego-
of 7. Over half of men with Gleason sum 8 to 10 will rization of patients into fewer (e.g., 3 or 4) risk groups
develop PSA recurrence at 5 years.10 simplifies the situation, it is important to point out that
In addition to clinical factors, pathologic information prognostic power can be reduced. This is particularly
obtained from the prostatectomy specimen can provide true for those patients with intermediate- and high-risk
prognostic information. This includes variables, such as disease, where significant overlap occurs and discrimina-
pathologic stage, total tumor volume, surgical margin tion of clinical outcome is less accurate.22,23 The develop-
status, and tumor grade in the specimen. The develop- ment of additional models incorporating novel and/or
ment of predictive nomograms and models has allowed molecular determinants of tumor behavior will allow
the prediction of both pathologic stage and clinical out- identification of patients most likely to benefit from
come (i.e., PSA-free survival), based on standard pre- prostatectomy, as well as those who may benefit from
treatment variables, and may increase the accuracy of risk adjuvant treatments.
assessment over the use of clinical stage, PSA level, and
Gleason score alone.15–17 The nomograms published by
Pelvic Lymph Node Dissection
Partin et al.18 predict the pathologic stage of disease, thus
helping one decide the role of surgery based on the cate- Contemporary series of patients undergoing radical
gorical estimation of organ-confined, established capsu- prostatectomy demonstrate that the risk of pelvic lymph
lar penetration, seminal vesical invasion, and lymph node node metastases is low, typically between 4% and 9%.24,25
involvement.15,16 In applying the Partin nomograms, it is This is largely due to the earlier detection of cancers at
important to point out that adverse pathologic features lower stage, as well as the refined selection of patients
alone, such as seminal vesicle invasion or extracapsular undergoing surgery with reduced likelihood of nodal
disease, may not preclude surgical therapy. From the involvement. Thus, pelvic lymphadenectomy may not be
experience of Catalona et al.,12 over 70% of patients with routinely indicated for all patients undergoing radical
unconfined disease and 35% of those with seminal vesi- prostatectomy. However, the detection of positive lymph
cle invasion have long-term (10 years) cancer-free sur- nodes provides important prognostic information and
vival with prostatectomy alone (no adjuvant therapy). should be performed in those patients at higher risk, as
Kattan and colleagues, rather than estimating patho- determined by nomogram or risk-group stratification.
logic stage as an outcome, created a continuous scale, Traditionally, it was thought that all patients with lymph
based on PSA, clinical stage, and Gleason grade, to cal- node metastases experience recurrence after prostatec-
culate the probability that a patient remains free of bio- tomy and therefore identification would spare patients an
chemical recurrence at 5 years after surgery.17 They ineffective, and potentially morbid, operation. Recent
have created a similar postoperative nomogram, pre- data question the paradigm and may renew interest in
dicting freedom from progression 7 years after surgery both pelvic lymphadenectomy and more aggressive sur-
based on PSA, Gleason sum in the prostatectomy spec- gical therapy. In addition, at the time of radical retropu-
imen, and individual pathologic features.19 These mod- bic prostatectomy, the pelvic lymph nodes are easily
els can be easily applied in the clinical setting using a accessible and removal can be performed with minimal
web-based (www.mskcc.org/nomograms/prostate) or morbidity; in general, unless the lymph nodes appear
computer calculation. grossly involved, frozen section analysis is unnecessary.
In order to simplify pretreatment risk stratification, a Heidenreich et al.26 extended the boundaries of the
three-group system has been developed based on PSA, pelvic lymphadenectomy in 103 patients, including the
clinical stage, and biopsy Gleason score. One that is com- external and internal iliac, obturator, common iliac, and
monly applied categorizes patients as low-risk patients presacral lymph nodes. They found a high rate of nodal
468 Part V Prostate Gland and Seminal Vesicles

metastases (26%) in patients undergoing increased sam- evaluation. Not only does this support the concept of
pling, suggesting that if lymph nodes are to be sampled, immediate androgen deprivation in those men with
the standard limits may be inadequate. node-positive prostate cancer but also raises the issue of
Nearly all men with proven pelvic lymph node metas- whether complete excision via prostatectomy and lymph
tases will have biochemical relapse with 5 to 7 years after node dissection plays a role in improving cancer out-
surgery. Thus, radical prostatectomy with distant disease comes even in those with regional metastases.
has been avoided because of limited benefit to the patient.
In a retrospective, nonrandomized study, Cadeddu et al.27
Results of Radical Prostatectomy
compared 10-year survival in men with proven lymph
node involvement who did and did not undergo radical The techniques of radical retropubic and perineal prosta-
prostatectomy. Overall, men fared better if the prostate tectomy are covered elsewhere in this book. Therefore,
had been removed and there was a suggestion of we describe cancer outcomes and morbidity related to
improved survival in this cohort. Similarly, Ghavamian such surgery in this section. Due to an improved under-
et al.28 demonstrated an overall survival advantage at 10 standing of periprostatic anatomy, increased surgeon
years in men with pTxN+ prostate cancer undergoing experience, and refinement in anesthesia and periopera-
prostatectomy and orchiectomy, compared with those tive patient care, morbidity from prostatectomy has been
with orchiectomy alone. More recently, data from reduced from that seen in previous decades.
Messing et al.29 indirectly address this question. In a
prospective randomized trial of 98 men undergoing rad-
Early and Intraoperative Complications
ical prostatectomy and pelvic lymph node dissection, an
improvement in survival was observed in men receiving Table 26A-2 summarizes perioperative data from con-
immediate hormonal therapy for microscopic lymph temporary series of patients undergoing radical retropu-
node disease compared to those men receiving delayed bic prostatectomy. Operative mortality, defined as death
hormonal therapy at the time of disease progression. within 30 days of surgery is exceedingly rare (<0.3%) but
This difference, at a median follow-up of 7.1 years, was is more common with increasing age and comorbid-
statistically significant ( p = 0.02) with 77% of men in the ity.30,32,35–38 In the 2001 report from Lepor et al.,32 a sin-
immediate-therapy group alive without disease at last gle death occurred in 1000 consecutive operations

Table 26A-2 Perioperative Morbidity and Mortality of Radical Retropubic Prostatectomy


Washington
University Mayo Clinic NYU Toulouse Baylor
(n = 1870)30 (n = 1000)31 (n = 1000)32 (n = 620)33 (n = 472)34

Complication Number % Number % Number % Number % Number %

Mortality 3 0.2 0 0 1 0.1 1 0.2 2 0.4

Rectal injury 3 0.2 6 0.6 5 0.5 3 0.5 3 0.6

Colostomy — — 0 0 0 0 — — 0 0

Ureteral injury — — — — 1 0.1 0 0 1 0.2

Myocardial infarction 9 0.7 7 0.7 5 0.5 1 0.2 2 0.4

Pulmonary embolism 22 1.7 6 0.6 2 0.2 5 0.8 5 1

DVT 8 0.6 14 1.4 2 0.2 14 2.3 6 1.3

Sepsis — — — — — — 1 0.2 3 0.6

Wound problem 17 1.3 9 0.9 9 0.9 6 1 14 2.9

Lymphocele — — — — 1 0.1 14 2.3 10 2.1

Anastomotic stricture — — 87 8.7 — — 3 0.5 42 9.0


Chapter 26A Clinically Localized (Stage T1a-T2c) Adenocarcinoma of the Prostate 469

(0.1%). The perioperative mortality rate at UCSF is and need for blood transfusion have been reported, with
0.13%. Similarly, complications, including myocardial the perineal approach associated with lower reported val-
infarction (0.1% to 0.4%), deep venous thrombosis ues (Tables 26A-3 and 26A-4).28,29,35,38 In general, the
(1.1%), and pulmonary embolism (0.75%) are less fre- average blood loss in recent series is less than 1000 ml
quent with modern perioperative care and are likely to and the use of nonautologous blood transfusion is less
decline further with continued experience and refine- than 5%; our experience with current mean blood loss
ments in technique.30,32,36,37 Begg et al.39 examined varia- and transfusion rate is 486 ml and 1%, respectively, in
tions in morbidity after prostatectomy using the our last 400 patients. However, the individual surgeon or
SEER-Medicare database. In examining the record of institutional experience dictates the need to donate autol-
11,522 patients who underwent prostatectomy, neither ogous blood. Postoperative bleeding requiring explo-
hospital volume nor surgeon volume was significantly ration is rare (<0.5%).35
associated with surgery-related death. However, postop- Rectal injury is also uncommon, estimated to occur in
erative morbidity, as well as late urinary complications 0.5% of men.35,39 These can be typically managed with
(stricture or fistula), was lower when surgery was per- primary closure, omental coverage, dilation of the anal
formed in very-high-volume hospitals ( p = 0.03) and by sphincter, and antibiotics even without preoperative
very-high-volume surgeons ( p < 0.001). bowel preparation. A routine diverting colostomy is rarely
The most common intraoperative problem is hemor- performed (0% to 0.06%) but may be necessary in those
rhage. Significant variations in the estimated blood loss with prior radiation exposure or extensive contamination.

Table 26A-3 Estimated Blood Loss During Radical Retropubic Prostatectomy


Institution Number Mean EBL (ml) Range (ml)

Mayo Clinic40 316 1020 100–4320

Washington University41 65* 1420 200–2500

65† 1605 250–3500

Toulouse33 220 300 100–1500

Mayo Clinic35 1728 600 —

Baylor42 954 800 150–5000

NYU32 1000 819 SD 14.9

UCSF 400 486 100–1200

*
With internal iliac artery occlusion.

Without internal iliac artery occlusion.

Table 26A-4 Summary of Radical Perineal Prostatectomy Series


Hospitali- Positive PSA-Free Follow-Up
Blood zation Continence Potency Margins Survival (Mean
Series Number Loss (ml) (days) (%) (%) (%) (%) Months)

Frazier43 122 565 12 96 77 29 — —

Lance44 190 802 — 65 8 43 82 42.9

Sullivan45 138 416 4.5 72 — 9 70 30

Weldon46 200 — — — — 44 79 35

Weldon47 110 645 — 95 70 — — —

Parra48 500 270 1.5 94 47 16 — —


470 Part V Prostate Gland and Seminal Vesicles

Injuries to the obturator nerve and ureter are rare tors, and the use of ultrasensitive or hypersensitive PSA
(both <0.1%) and can be managed intraoperatively by assays improves the lead-time for early detection of
direct reanastomosis and ureteroneocystostomy, recurrence.55,56 Amling et al.57 analyzed the effect of
respectively.11,32,49 using various PSA cut points defining biochemical failure
after radical prostatectomy in a cohort of 2782 men with
clinically localized disease (stages cT1 to T2). Five- and
Late Complications
10-year rates of PSA recurrence varied greatly, depend-
The majority of patients experience an uneventful post- ing on whether the threshold was 0.2, 0.3, 0.4, or 0.5
operative course, resuming oral intake and ambulation ng/ml. Clinical progression was directly related to the
the first day after surgery. In the U.S., men are safely dis- maximum PSA reached within 3 years of surgery. Using
charged between 1 and 5 days after surgery with the ure- definitions requiring multiple increases in PSA, such as
thral catheter in place.32,38 With the interest in earlier that proposed by American Society for Therapeutic
removal of the catheter (<10 days), rates of urinary reten- Radiology and Oncology for biochemical failure after
tion have increased and may approach 20%.50 radiation therapy, gave misleading results with overesti-
Development of an anastomotic stricture occurs in mation of short-term failure but improved long-term
approximately 1% to 10% of patients.11,32,38 Other com- recurrence-free outcomes. They conclude that a PSA cut
plications, including incisional hernia, lymphocele, ileus, point of 0.4 ng/ml may be most reasonable, with the lack
and inguinal hernia, are all less than 1%, while rates of of continuation of PSA progression in many of those with
rehospitalization are less than 5%.11,32,38 PSA values between 0.2 and 0.4 ng/ml; this may balance
the identification of patients with actual cancer recur-
rence at risk for subsequent clinical progression with
Outcomes After Radical Prostatectomy
sparing unnecessary adjuvant therapy in men with low
Cancer Control
but detectable PSA levels.
Contemporary methods of radical prostatectomy for Radical retropubic prostatectomy is associated with
patients with clinically localized disease are generally overall 5- and 10-year actuarial biochemical progression-
associated with very good outcomes. Several endpoints free survival rates ranging from 59% to 84% and from
can be evaluated—overall survival, cause-specific sur- 47% to 75%, respectively (Table 26A-5).35,59,60,63
vival, and biochemical relapse-free survival. Paulson Biochemical relapse-free survival following radical per-
et al.51 reported a 90% cancer-specific survival at 13.5 ineal prostatectomy ranges from 70% to 82% with mean
years after radical perineal prostatectomy for patients follow-up of 30 to 43 months (see Table 26A-4).43–45
with organ-confined and specimen-confined diseases. Biochemical relapse typically occurs 8 years prior to any
In an analysis of men treated with radical retropubic evidence of clinical disease recurrence; this is reflected by
prostatectomy at the Mayo clinic, Zincke et al.35,52 higher clinical disease-free rates at 5 (84% to 86%) and 10
reported crude and cause-specific survival rates of 75% (72% to 78%) years. The variability in outcomes across
and 90%, respectively. The crude survival rates at 10 and different reports and techniques likely reflects differences
15 years after surgery were similar to those of age- in patient selection, definition of biochemical failure, and
matched men from the general population without to a lesser extent variations in surgical technique.
prostate cancer. In two contemporary radical prostatectomy series
Despite the excellent results of radical prostatectomy, from the Cleveland Clinic and The Johns Hopkins
between 22% and 50% of patients thought to have Hospital, totaling more than 2000 men, no patient
organ-confined cancer are found to have disease beyond showed signs of clinical disease in the absence of PSA
the prostate on careful pathologic examination of the sur- failure.56,60 Following PSA recurrence, up to 68% of men
gical specimen.18,20,53,54 Given the protracted natural his- progressed to clinical disease at a median follow-up of 19
tory of prostate cancer and the fact that residual and/or months. The use of adjuvant therapy, such as radiation or
recurrent disease may respond to secondary therapy, androgen deprivation, at the time of PSA recurrence
postoperative follow-up with serum PSA testing is an reduced the rate of clinical disease progression (21%).
important endpoint in subsequent evaluation. Nearly all Without adjuvant or salvage therapy, metastatic disease
recurrent clinical and metastatic disease is preceded by a developed in 34% of patients at a median actuarial time
rising PSA, with only a few sporadic reported cases of of 8 years after PSA failure.
recurrence in the absence of a detectable serum PSA Han et al.61 recently reported the long-term biochem-
level. Biochemical failure is defined as either the persist- ical disease-free and cancer-specific survival in 2404
ence of a detectable PSA after surgery or the develop- patients managed with radical prostatectomy at The
ment of a detectable PSA in those with a previously Johns Hopkins Hospital. Mean follow-up was 6.3 years
undetectable postoperative level. The threshold consti- (range 1 to 17) and 26% of patients (n = 621) were fol-
tuting a detectable serum PSA varies among investiga- lowed at least for 10 years. Overall actuarial PSA progres-
Chapter 26A Clinically Localized (Stage T1a-T2c) Adenocarcinoma of the Prostate 471

Table 26A-5 Biochemical Relapse-Free Survival Following Radical Retropubic Prostatectomy


5-Year 10-Year 15-Year Follow-Up
Institution Number (%) (%) (%) Outcome (Years)

Washington University58 1778 78* — — PSA < 0.6 ng/ml Mean 4.0

Baylor20 1000 78 75 75† PSA < 0.4 ng/ml Mean 4.4

UCLA59 601 69 47 — PSA < 0.4 ng/ml Median 2.8

86 78 — Clinically free of disease

Mayo Clinic35 3170 70 52 — PSA < 0.2 ng/ml Mean 5.0

85 72 — Clinically free of disease

Cleveland Clinic60 423 59 — — PSA < 0.2 ng/ml Median 4.3

84 — — Clinically free of disease

Johns Hopkins61 2404 84 74 66 PSA < 0.2 ng/ml Mean 6.3

UCSF62 666 78 — — PSA < 0.2 ng/ml Median 3.1

*7-year results.

14-year results.

sion-free, metastasis-free, and cancer-specific survivals Pathologic stage is one of the most important prog-
were: 84%, 96%, and 99% at 5 years; 74%, 90%, and nostic factors, with increasing rates of recurrence with
96% at 10 years; 66%, 82%, and 90% at 15 years, respec- extraprostatic extension with negative surgical margins,
tively. These data are similar to those reported by positive surgical margins, seminal vesicle invasion, and
Catalona et al.58 and Hull et al.20 (see Table 26A-5). lymph node metastases. Actuarial recurrence-free sur-
Clinical and pathologic stage, pretreatment PSA, and vival in patients with organ-confined disease (i.e., ≤pT2)
pathologic Gleason score are all predictors of progres- is 97% at 5 years, 93% at 10 years, and 84% at 15 years.
sion after surgery (Table 26A-6).11 Catalona et al.12 Comparable data were reported by Hull et al.20 At 10
reported 79% and 66% 7-year disease-free survivals in years, those with organ-confined and nonorgan-confined
those with stage cT1c and stage cT2, respectively. From tumors experienced 92% and 53% progression-free sur-
the Johns Hopkins series, patients with clinical stage ≤ T2 vivals, respectively. Men with pathologic stages T1-2,
had 5-, 10-, and 15-year recurrence-free survival rates of T3a/b, T3c, and N+ had 10-year actuarial PSA-free sur-
86%, 76%, and 71%, respectively, compared to 60% and vivals of 92%, 71%, 37%, and 7%, respectively. At
49% in patients with T3a disease (15-year data unavail- UCSF, patients with pathologically confined disease
able).61 At 10 years, Hull et al.20 observed >80% and 67% (pT2, all grades) have a 5-year PSA progression-free
PSA-free survival in those with clinical stages T1 and T2, actuarial survival rate of 80%, compared with 57% in
respectively. those with higher stages.
Seven-year progression-free survival was 93%, 80%, The incidence of positive surgical margins has varied
76%, and 40% in PSA ranges of <2.5, 2.5 to 4.0, 4.1 to greatly, ranging between 10% and 68%.64–66 The finding
10, and >10 ng/ml, respectively.12 The 10-year recur- of a positive margin on the prostatectomy specimen
rence-free data from Johns Hopkins are comparable: would suggest incomplete removal of the tumor and is
91% for ≤ 4 ng/ml, 79% for 4.1 to 10 ng/ml, 57% for associated with a 2- to 4-fold increased risk of progres-
10.1 to 20 ng/ml, and 48% for >20 ng/ml.61 sion.20 Nevertheless, those men with a positive margin
Higher Gleason grade, both on diagnostic biopsy and may experience reasonable long-term recurrence-free
in the surgical specimen, is associated with increased risk survival (36% at 10 years) without adjuvant therapy; the
of disease recurrence. Seven-year progression-free sur- reported 10-year results for those with negative margins
vival was 84% in patients with Gleason sum 2 to 4, 68% is 81%. In an attempt to reduce the incidence of positive
with Gleason sum 5 to 7, and 48% with Gleason sum 8 surgical margins while performing nerve-sparing radical
to 10.12 Han et al.61 reported 10-year recurrence-free retropubic prostatectomy, we examined the utility of
rates of 91%, 79%, 57%, and 48% for Gleason sums ≤ 6, intraoperative frozen section analysis.67 In 101 men,
3 + 4, 4 + 3, and 8 to 10, respectively. intraoperative frozen section was performed on the surgical
472 Part V Prostate Gland and Seminal Vesicles

Table 26A-6 Risk Factors for Progression After Radical Prostatectomy (Multivariate Analysis)
Variable Relative Risk (95% CI) p Value

Preoperative parameter

Clinical stage 0.0071

T1a,b versus T1c 0.6

T1c versus T2a 0.1

T1c versus T2b 2.47 (1.52–4.03) 0.0003

T1c versus T2c 1.91 (1.06–3.42) 0.0304

Biopsy Gleason sum

2–4 versus 5–6 0.15

5–6 versus 7 2.6 (1.75–3.87) <0.0001

5–6 versus 8–10 3.21 (1.72–5.97) 0.0002

Clinical and pathologic parameters

Clinical stage 0.15

Biopsy Gleason sum 0.12

Specimen Gleason sum 0.0008

2-4 versus 5-6 2.48 (1.34–4.58) 0.0038

5-6 versus 7 2.48 (1.34–4.58) 0.0038

5-6 versus 8-10 4.55 (2.19-9.42) <0.0001

Extracapsular extension 0.0019

Focal versus none 2.17 (1.20–3.92) 0.011

Established versus none 2.72 (1.56–4.74) 0.0004

Focal versus established 0.13

Surgical margin

Positive versus negative 4.37 (2.90–6.58) <0.0001

Seminal vesicle involvement

Present versus absent 2.61 (1.70–4.01) <0.0001

Lymph node metastases

Present versus absent 3.31 (2.11–5.20) <0.0001

margin thought to be at risk of tumor involvement. If the group, 12 (80%) had no evidence of tumor in addition-
margin was positive, additional tissue was excised. ally resected tissue. Overall, the positive and negative
Findings of intraoperative examination were identical to predictive values for intraoperative frozen section analy-
those of final permanent section in 91% of cases. Of the sis were 73% and 94%, respectively. The risk of recur-
15 patients with a positive frozen section, 11 had identi- rence in patients with either positive or negative
cal findings on permanent pathologic sections. In this intraoperative frozen section findings was similar. These
Chapter 26A Clinically Localized (Stage T1a-T2c) Adenocarcinoma of the Prostate 473

data suggest that intraoperative frozen section analysis Table 26A-7 Risk Factors for Urinary Incontinence
may be applied during radical prostatectomy to spare the After Radical Prostatectomy
neurovascular bundles in selected patients. Variable p Value*
In order to improve simplify, as well as increase the
accuracy of assessment of postoperative prognosis, clini- Patient weight (continuous) 0.0002
cal and pathologic features have been combined. A mul-
tivariate analysis from Johns Hopkins found that Obstructive urinary symptoms 0.004
pathologic stage, Gleason score, and surgical margin sta-
TURP 0.001
tus were the best predictors of the probability of cancer
recurrence.68 The risk stratification scheme, discussed Blood loss (continuous) 0.016
above, is able to categorize patients into low-, intermedi-
ate-, and high-risk groups with 5-year recurrence-free Nerve resection 0.001 (0.015)
survival rates of 98%, 73%, and 65%, respectively.20 Anastomotic stricture 0.001 (0.015)
Although radical prostatectomy is fairly effective even in
higher-risk patients, early identification of those at risk Patient age (continuous) 0.001 (0.0001)
for eventual primary treatment failure help select men
appropriate for adjuvant therapy and enrollment in clin- Anastomotic method 0.001 (0.0001)
ical trials.
*In univariate analysis; numbers in parenthesis represent p values in
multivariate analyses.
Urinary Function
After radical prostatectomy, immediate postoperative
incontinence may occur in up to 80% of patients. More within 18 months; more recently, half of patients have
contemporary series report a significantly lower rate of immediate recovery of urinary control.
immediate incontinence.30,32,69 The wide variability in In a national survey including 1796 men undergoing
incontinence rates reported in the literature is due to radical prostatectomy who were continent before the
improvements in surgical technique, as well as differing operation, 19% required pads daily and 3.6% were
definitions of what constitutes urinary continence. totally incontinent.73 The National Institutes of Health-
Approximately 90% of men who undergo radical sponsored Prostate Cancer Outcomes study found that
prostatectomy will be continent at 1 year, when the 8.5% of men reported “severe” incontinence.74 It is
definition is no regular use of pads and/or no leakage important to note that despite the presence of urinary
with moderate exercise. Severe and persistent inconti- incontinence, most patients were minimally bothered
nence, defined as leakage with normal activity or the and highly satisfied with their treatment. This has been
need for ≥ 3 pads per day, occurs in 1% to 6% of confirmed in a recent randomized study comparing radi-
patients.70 cal prostatectomy with watchful waiting.75 Despite a
Most major centers report urinary incontinence higher incidence of urinary leakage with radical prostate-
rates less than 10%. In 593 patients undergoing sur- ctomy (49%), overall well being and subjective quality-
gery by Walsh, some degree of stress urinary inconti- of-life assessment were not significantly different from
nence was present in 8%; the remaining 92% of the watchful waiting group.
patients were completely continent.71 The group from We recently examined the impact of even minimal uri-
Baylor reported the median time to recovery of conti- nary leakage after radical prostatectomy on health-
nence was 1.5 months, with 92% and 95% of patients related quality of life.76 Postoperative assessment of
continent at 1 and 2 years, respectively.72 Factors asso- urinary health-related quality of life, using the urinary
ciated with incontinence after radical prostatectomy function and bother subscales of the University of
are listed in Table 26A-7. At UCSF, a contemporary California Los Angeles Prostate Cancer Index, American
cohort of patient undergoing radical retropubic prosta- Urologic Association Symptom Index, and a single ques-
tectomy (n = 217) was surveyed anonymously using a tion assessing satisfaction, was performed in 168 men at
detailed, validated questionnaire, including the a mean of 75 weeks after surgery. Overall, 87% reported
University of California, Los Angeles/Rand Prostate no pad use while 12% used one pad daily. Statistically
Cancer Index quality-of-life urinary domain questions. significant differences were observed between these
At 1 year after surgery, overall continence (defined groups with respect to mean function score ( p < 0.0001),
above) was 98%, with immediate continence in 23% mean bother score ( p < 0.0001), and change in AUA
and 55% of patients continent by 1 month. Catalona quality-of-life score ( p < 0.0001). Overall satisfaction was
et al.12 noted that 96% and 87% of men younger and higher in the no pad group (87.1) compared to the single-
older than 70 years, respectively, recovered continence pad group (50.9; p < 0.0001).
474 Part V Prostate Gland and Seminal Vesicles

Erectile Function treatment of localized prostate cancer. J Urol 1994;


152:1910–1914.
Until the description of the anatomy and location of the 4. Holmberg L, Bill-Axelson A, Helgesen F, et al: A
autonomic branches of the pelvic plexus innervating the randomized trial comparing radical prostatectomy with
corpora cavernosa, men were expected to be impotent watchful waiting in early prostate cancer. N Engl J Med
after radical prostatectomy.77 Identification of these 2002; 347:781–789.
nerves allowed modification of the surgical technique 5. Chodak GW, Thisted RA, Gerber GS, et al: Results of
and attempted preservation of potency in men undergo- conservative management of clinically localized prostate
ing removal of the prostate.78 Quinlan et al.79 reported cancer. N Engl J Med 1994; 330:242–248.
outcomes in 600 men aged 34 to 72 years undergoing 6. Albertsen PC, Fryback DG, Storer BE: Long-term
radical prostatectomy. Of 503 men with preoperative survival among men with conservatively treated localized
prostate cancer. J Am Med Assoc 1995; 274:626–631.
sexual function followed at least 18 months, 68%
7. Albertsen PC, Hanley JA, Gleason DF, Barry MJ:
remained potent after surgery. Studies have found that Competing risk analysis of men aged 55 to 74 years at
multiple factors are involved in the preservation of diagnosis managed conservatively for clinically localized
potency.30,79,80 In the Johns Hopkins’ experience, these prostate cancer. J Am Med Assoc 1998; 280:975–980.
included age, stage, and preservation or excision of the 8. Fleming C, Wasson JH, Albertsen PC et al: A decision
neurovascular bundles.79 Potency was preserved in 91% analysis of alternative treatment strategies for
younger than 50 years, 75% in men 50 to 60 years, 58% clinically localized prostate cancer. Prostate Patient
in men 60 to 70 years, and 25% in men older than 70 Outcomes Research Team. J Am Med Assoc 1993;
years. With increasing age, preservation of both neu- 269:2650–2658.
rovascular bundles was more important in potency. After 9. Beck JR, Kattan MW, Miles BJ: A critique of the decision
controlling for other variables, the presence of more analysis for clinically localized prostate cancer. J Urol
1994; 152:1894–1899.
advanced stage (nonorgan-confined, seminal vesicle inva-
10. Walsh PC: The natural history of localized prostate
sion) was associated with increased risk of impotence. cancer: a guide to therapy. In Campbell’s Urology.
Similarly, Rabbani et al.80 identified patient age, preoper- Philadelphia, WB Saunders, 1998.
ative potency, and extent of neurovascular bundle preser- 11. Ohori M, Scardino PT: Localized prostate cancer. Curr
vation as predictors of potency recovery. At 3 years Probl Surg 2002; 39:833–957.
following surgery, 76%, 56%, and 47% of those aged 12. Catalona WJ, Ramos CG, Carvalhal GF: Contemporary
<60, 60 to 65, and >65 years were potent. Men with only results of anatomic radical prostatectomy. CA Cancer J
partial preoperative erection and unilateral neurovascular Clin 1999; 49:282–296.
preservation were less likely (47% and 25%, respectively) 13. Walsh PC, Partin AW, Epstein JI: Cancer control and
to recover potency. Potency after radical perineal prosta- quality of life following anatomical radical retropubic
tectomy has been documented in smaller series, with prostatectomy: results at 10 years. J Urol 1994;
152:1831–1836.
rates near 70%.43,51
14. Pound CR, Partin AW, Epstein JI, Walsh PC: Prostate-
As with urinary function after prostatectomy, a wide specific antigen after anatomic radical retropubic
range of success has been reported for postoperative prostatectomy: patterns of recurrence and cancer control.
potency (3% to 46%). This variability is due to patient Urol Clin North Amer 1997; 24:395–406.
selection, surgeon expertise, definition of potency, and 15. Partin AW, Yoo J, Carter HB, et al: The use of prostate
means of assessing sexual function. In men with erectile specific antigen, clinical stage and Gleason score to
dysfunction after a bilateral nerve-sparing prostatectomy, predict pathological stage in men with localized prostate
sildenafil may be efficacious.81–83 Between 43% and 75% cancer. J Urol 1993; 150:110–114.
of such patients may respond to sildenafil; conversely, 16. Partin AW, Mangold LA, Lamm DM, et al:
few men benefit if both neurovascular bundles have been Contemporary update of prostate cancer staging
excised or ligated. nomograms (Partin tables) for the new millennium.
Urology 2001; 58:843–848.
17. Kattan MW, Eastham JA, Stapleton AM: A preoperative
nomogram for disease recurrence following radical
REFERENCES prostatectomy for prostate cancer. J Natl Cancer Inst
1998; 90:766–771.
1. Walsh PC: Prostate cancer kills: strategy to reduce death. 18. Partin AW, Kattan MW, Subong EN, et al: Combination
Urology 1994; 44:463–466. of prostate-specific antigen, clinical stage, and Gleason
2. Thompson IM: Counseling patients with newly diagnosed score to predict pathological stage of localized prostate
prostate cancer. Oncology (Huntingt) 2000: 14:119–126. cancer: a multi-institutional update. J Am Med Assoc
3. Wilt TJ, Brawer MK: Prostate cancer intervention 1997; 277:1445–1451.
versus observation trial: a randomized trial comparing 19. Kattan MW, Wheeler TM, Scardino PT: Postoperative
radical prostatectomy versus expectant management for nomogram for disease recurrence after radical
Chapter 26A Clinically Localized (Stage T1a-T2c) Adenocarcinoma of the Prostate 475

prostatectomy for prostate cancer. J Clin Oncol 1999; 34. Dillioglugil O, Leibman BD, Leibman NS, et al: Risk
17:1499–1507. factors for complications and morbidity after radical
20. Hull GW, Rabbani F, Abbas F, et al: Cancer control with retropubic prostatectomy. J Urol 1997; 157:1760–1767.
radical prostatectomy alone in 1,000 consecutive patients. 35. Zincke H, Oesterling JE, Blute ML, et al: Long-term
J Urol 2002; 167:528–534. (15 years) results after radical prostatectomy for clinically
21. Koppie TM, Grossfeld GD, Miller D, et al: Patterns of localized prostate (stage T2c or lower) prostate cancer.
treatment of patients with prostate cancer initially J Urol 1994; 152:1850–1857.
managed with surveillance: results from the CapSure 36. Lu-Yao GL, Albertsen P, Warren J, Yao SL: Effect of age
database. Cancer of the prostate strategic urological and surgical approach on complications and short-term
research endeavor. J Urol 2000; 164:81–88. mortality after radical prostatectomy: a population-based
22. Grossfeld GD, Latini DM, Lubeck DP, et al: Predicting study. Urology 1999; 54:301–307.
disease recurrence in intermediate and high-risk patients 37. Thompson IM, Middleton RG, Optenberg SA, et al:
undergoing radical prostatectomy using percent positive Have complication rates decreased after treatment for
biopsies: results from CapSure. Urology 2002; localized prostate cancer? J Urol 1999; 162:107–112.
59:560–565. 38. Hu JC, Gold KF, Pashos CL, et al: Temporal trends in
23. Moul JW, Connelly RR, Lubeck DP, et al: Predicting risk radical prostatectomy complications from 1991 to 1998.
of prostate specific antigen recurrence after radical J Urol 2003; 169:1443–1448.
prostatectomy with the center for prostate disease 39. Begg CB, Riedel ER, Bach PB, et al: Variations in
research and cancer of the prostate strategic urologic morbidity after radical prostatectomy. N Engl J Med
research endeavor databases. J Urol 2001; 166:1322–1327. 2002; 346:1138–1144.
24. Bishoff JT, Reyes A, Thompson IM, et al: Pelvic 40. Rainwater LM, Segura JW: Technical consideration in
lymphadenectomy can be omitted in selected patients with radical retropubic prostatectomy: blood loss after ligation
carcinoma of the prostate: development of a system of of dorsal venous complex. J Urol 1990; 143:1163–1165.
patient selection. Urology 1995; 45:270–274. 41. Kavoussi LR, Myers JA, Catalona WJ: Effect of
25. Danella JF, deKerion JB, Smith RB, Steckel J: The temporary occlusion of hypogastric arteries on blood loss
contemporary incidence of lymph node metastases in during radical retropubic prostatectomy. J Urol 1991;
prostate cancer: implications for laparoscopic lymph node 146:362–365.
dissection. J Urol 1993; 149:1488–1491. 42. Eastham JA, Scardino PT: Radical prostatectomy. In
26. Heidenreich A, Varga Z, von Knobloch R: Extended Campbell’s Urology. Philadelphia, WB Saunders, 1998.
pelvic lymphadenectomy in patients undergoing radical 43. Frazier HA, Robertson JE, Paulson DF: Radical
prostatectomy: high incidence of lymph node metastasis. prostatectomy: the pros and cons of the perineal versus
J Urol 2002; 167:1681–1686, 2002. retropubic approach. J Urol 1992; 147:888–890.
27. Cadeddu JA, Partin AW, Epstein JI, Walsh PC: Stage D1 44. Lance RA, Freidrichs PA, Kane CJ, et al: A comparison of
(T1-3, N1-3, M0) prostate cancer: a case-controlled radical retropubic with perineal prostatectomy for
comparison of conservative treatment versus radical localized prostate cancer within the uniformed services
prostatectomy. Urology 1997; 50:251–255. urology research group. BJU Int 2001; 87:61–65.
28. Ghavamian R, Bergstralh EJ, Blute ML, et al: Radical 45. Sullivan LD, Weir MJ, Kinahan JF, Taylor DL: A
retropubic prostatectomy plus orchiectomy versus comparison of the relative merits of radical perineal and
orchiectomy alone for pTxN+ prostate cancer: a matched radical retropubic prostatectomy. BJU Int 2000;
comparison. J Urol 1999; 161:1223–1227. 85:95–100.
29. Messing EM, Manola J, Sarosdy M, et al: Immediate 46. Weldon VE, Tavel FR, Neuwirth H, Cohen R: Patterns
hormonal therapy compared with observation after radical of positive specimen margins and detectable prostate
prostatectomy and pelvic lymphadenectomy in men with specific antigen after radical perineal prostatectomy.
node-positive prostate cancer. N Engl J Med 1999; J Urol 1995; 153:1565–1569.
341:1781–1788. 47. Weldon VE, Tavel FR, Neuwirth H: Continence, potency
30. Catalona WJ, Carvalhal GF, Mager DE, Smith DS: and morbidity after radical perineal prostatectomy. J Urol
Potency, continence, and complication rates in 1870 1997; 158:1470–1475.
consecutive radical retropubic prostatectomies. J Urol 48. Parra RO: Analysis of an experience with 500 radical
1999; 162:433-438. perineal prostatectomies in localized prostate cancer.
31. Lerner SE, Blute ML, Lieber MM, Zincke H: Morbidity J Urol 2000; 163A:284–285.
of contemporary radical retropubic prostatectomy for 49. Walsh PC: Anatomic radical retropubic prostatectomy.
localized prostate cancer. Oncology (Huntingt) 1995; In Campbell’s Urology. Philadelphia, WB Saunders, 1998.
9:379–382. 50. Patel R, Lepor H: Removal of urinary catheter on
32. Lepor H, Nieder AM, Ferrandino MN: Intraoperative postoperative day 3 or 4 after radical retropubic
and postoperative complications of radical retropubic prostatectomy. Urology 2003; 61:156–160.
prostatectomy in a consecutive series of 1000 cases. J Urol 51. Paulson DF: Impact of radical prostatectomy in the
2001; 166:1729–1733. management of clinically localized disease. J Urol 1994;
33. Leandri P, Rossignol G, Gautier JR, Ramon J: Radical 152:1826–1830.
retropubic prostatectomy: morbidity and quality of life. 52. Zincke H, Bergstralh EJ, Blute ML, et al: Radical
J Urol 1992; 147:883–887. prostatectomy for clinically localized prostate cancer:
476 Part V Prostate Gland and Seminal Vesicles

long-term results of 1143 patients from a single 67. Goharderakhshan RZ, Sudilovsky D, Carroll LA, et al:
institution. J Clin Oncol 1994; 12:2254–2263. Utility of intraoperative frozen section analysis of surgical
53. Grossfeld GD, Chang JJ, Broering JM, et al: Does margins in region of neurovascular bundles at radical
the completeness of prostate sampling predict outcome prostatectomy. Urology 2002; 59:709–714.
for patients undergoing radical prostatectomy? Data 68. Epstein JI, Partin AW, Sauvageot J, et al: Prediction of
from the CapSure database. Urology 2000; progression following radical prostatectomy: a
56:430–435. multivariate analysis of 721 men with long-term follow-
54. Han M, Partin AW, Zahurak M et al: Biochemical up. Am J Surg Pathol 1996; 20:286–292.
(prostate specific antigen) recurrence probability following 69. Goluboff ET, Saidi JA, Mazer S, et al: Urinary continence
radical prostatectomy for clinically localized prostate after radical prostatectomy: the Columbia experience.
cancer. J Urol 2003; 169:517–523. J Urol 1998; 159:1276–1280.
55. Yu H, Diamandis EP, Prestigiacomo AF, Stamey TA: 70. Wahle GR: Urinary incontinence after radical
Ultrasensitive assay of prostate-specific antigen used for prostatectomy. Semin Urol Oncol 2000; 18:66–70.
early detection of prostate cancer relapse and estimation 71. Steiner MS, Morton RA, Walsh PC: Impact of anatomical
of tumor-doubling time after radical prostatectomy. Clin radical prostatectomy on urinary continence. J Urol 1991;
Chem 1995; 41:430–434. 145:512–515.
56. Pound CR, Partin AW, Eisenberger MA, et al: Natural 72. Eastham JA, Goad JR, Rogers E, et al: Risk factors for
history of progression after PSA elevation following urinary incontinence after radical prostatectomy. J Urol
radical prostatectomy. J Am Med Assoc 1999; 1996; 156:1707–1713.
281:1591–1597. 73. Murphy GP, Mettlin C, Menck H, et al: National patterns
57. Amling CL, Bergstralh EJ, Blute ML, et al: Defining of prostate cancer treatment by radical prostatectomy:
prostate specific antigen progression after radical results of a survey by the American College of Surgeons
prostatectomy: what is the most appropriate cut point? Commission on Cancer. J Urol 1994; 152:1817–1819.
J Urol 2001; 165:1146–1151. 74. Stanford JL, Feng Z, Hamilton AS, et al: Urinary and
58. Catalona WJ, Smith DS: Cancer recurrence and survival sexual function after radical prostatectomy for clinically
rates after anatomic radical retropubic prostatectomy for localized prostate cancer. J Am Med Assoc 2000;
prostate cancer: intermediate-term results. J Urol 1998; 283:354–360.
160:2428–2434. 75. Steineck G, Helgesen J, Adolfsson J, et al: Quality of life
59. Trapasso JG, deKerion JB, Smith RB, Dorey F: The after radical prostatectomy or watchful waiting. N Engl
incidence and significance of detectable levels of serum J Med 2002; 347:790–706.
prostate specific antigen after radical prostatectomy. 76. Cooperberg MR, Master VA, Carroll PR: Health related
J Urol 1994; 152:1821–1825. quality of life significance of single pad urinary
60. Kupelian PA, Katcher J, Levin HS, et al: Stage T1-2 incontinence following radical prostatectomy. J Urol
prostate cancer: a multivariate analysis of factors affecting 2003; 170:512–515.
biochemical and clinical failures after radical 77. Walsh PC, Donker PJ: Impotence following radical
prostatectomy. Int J Radiat Oncol Biol Phys 1997; prostatectomy: insight into etiology and prevention.
37:1043–1052. J Urol 1982; 128:492–497.
61. Han M, Partin AW, Pound CR, et al: Long-term 78. Walsh PC: Anatomic radical prostatectomy: evolution of
biochemical disease-free and cancer-specific survival the surgical technique. J Urol 1998; 160:2418–2424.
following anatomic radical retropubic prostatectomy. The 79. Quinlan DM, Epstein JI, Carter BS, et al: Sexual function
15-year Johns Hopkins experience. Urol Clin North Am following radical prostatectomy: influence of preservation
2001; 28:555–565. of neurovascular bundles. J Urol 1991; 145:998–1002.
62. Carroll PR, Meng MV, Downs TM, Grossfeld GD: 80. Rabbani F, Stapleton AM, Kattan MW, et al: Factors
Radical retropubic prostatectomy. In Prostate Cancer. predicting recovery of erections after radical
Hamilton, Decker, 2002. prostatectomy. J Urol 2000; 164:1929–1934.
63. Catalona WJ, Smith DS: 5-year tumor recurrence rates 81. Zippe CD, Jhaveri FM, Klein EA, et al: Role of
after anatomical radical retropubic prostatectomy for Viagra after radical prostatectomy. Urology 2000;
prostate cancer. J Urol 1994; 152:1837–1842. 55:241–245.
64. Wieder JA, Soloway MS: Incidence, etiology, location, 82. Zippe CD, Kedia S, Kedia AW, Pasqualotto F: Sildenafil
prevention and treatment of positive surgical margins citrate (Viagra) after radical retropubic prostatectomy:
after radical prostatectomy for prostate cancer. J Urol pro. Urology 1999; 54:583–586 (editorial).
1998; 160:299–315. 83. Blander DA, Sanchez-Ortiz RF, Wein AJ, Broderick GA:
65. Blute ML, Bostwick DG, Seay TM et al: Pathologic Efficacy of sildenafil in erectile dysfunction after radical
classification of prostate carcinoma: the impact of margin prostatectomy. Int J Impot Res 2000; 12:165–168.
status. Cancer 1998; 82:902–908.
66. Grossfeld GD, Tigrani VS, Nudell D, et al: Management
of a positive surgical margin after radical prostatectomy:
decision analysis. J Urol 2000; 164:93–99.
C H A P T E R

26BClinically Localized Adenocarcinoma


of the Prostate: Radiation Therapy
W. Robert Lee, MD, MS

Management of men with clinically localized (T1-2) is to affect the natural history of the disease, especially in
prostate cancer is one of the most controversial areas in men with favorable disease, it will not be apparent for
all of oncology. Treatment options include expectant several years following treatment.
management, radical prostatectomy (RP), external beam In addition, many men with prostate cancer are
radiation therapy (EBRT), and interstitial brachytherapy beyond the age of 65 at diagnosis and have competing co-
(IB), with or without additional EBRT.1 The results of morbidities. In a report including more than 1400 men
newer approaches, including high intensity focused ultra- treated with EBRT on Radiation Therapy Oncology
sound (HIFU) and cryotherapy, have demonstrated some Group (RTOG) trials between 1975 and 1992, more than
promise but they will not be discussed here. 50% of men died of other causes.5 Other reports of men
The gold standard for treatment efficacy is consid- managed with expectant management illustrate the same
ered to be the randomized clinical trial (RCT). To date phenomenon.6 The competing risk from other causes of
the number of RCT comparing definitive treatments in mortality (cerebrovascular disease, cardiovascular dis-
men with T1-2 prostate cancer is very few, and in some ease) makes it difficult to demonstrate that treating
cases the trials have methodologic flaws that preclude prostate cancer reduces overall mortality.
interpretation.2,3 Given the relative paucity of level I evi- Finally, temporal trends confound any statements
dence, clinicians and their patients rely on retrospective, about treatment efficacy. With the advent of PSA screen-
nonrandomized historic case series for treatment recom- ing, significant stage migration has been documented.7,8
mendations. The potential for bias in these historic Men treated in 2003 in general have less disease than
series is well known. Several characteristics of prostate men treated in the late 1980s. Treatment techniques have
cancer make interpretation of available information also evolved in the past two decades. RP has been refined,
difficult. and morbidity has been reduced. Techniques of radiation
In the first place, the long natural history of clinically therapy (EBRT or IB) have also evolved. Thus, the
localized prostate cancer makes it very difficult to results reported from men treated 15 years ago may not
demonstrate a beneficial effect of any therapeutic proce- be representative of what is achievable with current
dure. D’Amico4 has quantified prostate cancer-specific methods.
mortality during the first decade in men with T1-2 The preceding paragraphs should emphasize the
prostate cancer treated with RP or EBRT in the prostate- difficulties inherent in interpreting the available liter-
specific antigen (PSA) era. This report included more ature as it relates to the management of men with T1-2
than 7000 men (4946 treated with RP and 2370 treated prostate cancer. In this chapter, biochemical relapse-
with EBRT) from 44 treating institutions. The rate of free survival (BRFS) (incorporating PSA into the
prostate cancer specific mortality (PCSM) was dependent definition of disease-free survival) will be emphasized.
on pretreatment risk grouping and primary treatment. In The use of PSA as an endpoint has a number of advan-
men treated with EBRT, the rate of PCSM at 10 years tages and disadvantages, which are discussed in a pre-
was <5% for men with low-risk disease, rising to approx- vious section. Patient-reported endpoints, including
imately 10% in the intermediate-risk group and nearly health-related quality of life (HRQOL) after definitive
25% in the high-risk group. If aggressive local treatment treatment is considered elewhere.

477
478 Part V Prostate Gland and Seminal Vesicles

RISK STRATIFICATION
A number of variables have been reported to be inde-
pendent predictors of BRFS following radiation therapy
(EBRT or IB) for clinically localized prostate cancer.
These variables include Gleason grade, pretreatment
PSA level, and clinical T stage.9–15 In hopes of accurately
predicting biochemical outcome following definitive
treatment, a number of investigators have combined
these variables to produce risk groupings.10,16,17 One of
the more commonly used risk stratifications was first
described by D’Amico.10 In a single-institution setting
with BRFS as the endpoint, the D’Amico risk stratifica-
tion system successfully classified patients into three
groups (low, intermediate, and high) regardless of treat-
ment received (RP, EBRT, or IB). A recent report has
demonstrated that the D’Amico risk groupings also pre-
dict prostate cancer-specific survival.4 The results with
radiation therapy should be presented according to risk
grouping.

HISTORIC PERSPECTIVE, EXTERNAL BEAM


RADIATION THERAPY
Megavoltage EBRT for clinically localized prostate can-
cer has been in use since the late 1950s following the
development of Co60 teletherapy and linear accelera-
tors.18 Over the ensuing years a number of advances in
technology have allowed for modifications in the treat-
ment planning process. As late as the early 1990s most
men treated with EBRT were treated with conventional
methods. In this paradigm, the treatment planning
process relied on two-dimensional plain radiographs for
target localization and shielding of normal tissues. This
method did not allow direct visualization of the prostate
gland. The location of the prostate was estimated based
on bony landmarks and radio-opaque contrast in nearby
normal tissues. Figure 26B-1 Example of beam-shaping used for 3DCRT. A,
With the development of more sophisticated treat- Antero-posterior beam. B, Right lateral beam. C, Oblique
ment planning software, three-dimensional conformal beam
radiation therapy (3DCRT) was born. In 3DCRT, the
target organ and any relevant nearby normal tissues are
outlined on sequential computed tomography (CT)
images. These outlines are then reconstructed to allow
shaping of the radiation portal to conform to the shape of
the target organ (Figure 26B-1). As outlined later, dose gradient that results in reduced total dose to nearby
3DCRT techniques allow for an increase in dose to the normal tissues.19 IMRT is gradually being introduced
prostate gland without a significant increase in normal into community practice although the availability
tissue complications. remains somewhat limited at this time.
Intensity-modulated radiation therapy (IMRT) is a
further refinement of 3DCRT. With IMRT the fluence
TECHNIQUE OF EBRT
of linear accelerator photon beams are modulated to cre-
ate more ideal dose distributions. Potential advantages of Widespread availability of the necessary equipment
IMRT versus 3DCRT include a greater conformality of should permit all men treated with EBRT for prostate
radiation dose about the target organ and a much steeper cancer to be approached using 3DCRT. CT simulators
Chapter 26B Clinically Localized Adenocarcinoma of the Prostate: Radiation Therapy 479

Table 26B-1 Volume Recommendations of the lymph nodes above 15%, elective pelvic nodal irradiation
International Commission of Radiation Units should be strongly considered.
and Measurements (ICRU Report 50)
Parameters Definition BIOCHEMICAL RELAPSE-FREE SURVIVAL:
EXTERNAL BEAM RADIATION THERAPY
GTV Palpable or visible extent of tumor
Soon after the discovery of PSA in the serum of men with
CTV GTV plus margins for subclinical disease prostate cancer this protein began to be used to monitor
PTV CTV plus margins for organ motion the results of therapy.29 Early reports indicated that meas-
and daily set-up error ured serum PSA would decrease in the first several
months for the majority of men with T1-2 prostate can-
cer treated with EBRT.30–32 Soon thereafter radiation
are becoming commonplace in radiation therapy depart- oncologists began to incorporate PSA into the definition
ments, but even if conventional simulators are used, the of disease-free survival. In the early 1990s, a number of
information obtainable from CT or magnetic resonance different definitions of BRFS following EBRT were
(MR) should be incorporated into the treatment plan- developed and reported in the peer-reviewed literature.
ning process. These newer techniques should be The multiple definitions led to considerable difficulties in
approached in a systematic fashion. deciding when biochemical relapse had occurred and
With the introduction of 3DCRT, a new terminology made comparisons and interpretation of published reports
is required (Table 26B-1).20 In this vocabulary, the gross difficult. In an attempt to reconcile the numerous defini-
tumor volume (GTV) represents the tumor which is pal- tions of biochemical relapse, the American Society of
pable or visible on imaging. The clinical target volume Therapeutic Radiology and Oncology (ASTRO) con-
(CTV) includes the GTV plus any additional structures vened a conference to discuss the role of PSA measure-
that are felt to be at risk for microscopic involvement ments after radiation therapy. The result of this
(seminal vesicles or pelvic lymph nodes). The planning conference was the publication of the ASTRO Consensus
target volume (PTV) includes the CTV plus a margin Definition (ACD) of biochemical failure.33 The ACD
that accounts for organ motion and set-up error. In most defined biochemical failure as three consecutive rises
cases of prostate cancer, the CTV to PTV margin is 5 to from a nadir value, and the time to failure was the mid-
10 mm. Many clinicians will use nonuniform margins point between the nadir value and the first rise. To date,
with the smallest CTV to PTV margins posteriorly in the utilization of the ACD is not universal.34 Weaknesses
the vicinity of the rectum.21 of the present definition have been identified.35–37 Despite
A variety of beam arrangements have been described these problems many investigators continue to report
ranging from a coplanar, four-field technique to a non- their results using the ACD, and the results reported in
coplanar seven-field approach. Megavoltage energies (>6 this chapter will rely on the ACD unless otherwise stated.
MV) are recommended. Immobilization devices are com- A number of series providing external beam radiation
monly used to decrease set-up error, reducing the CTV results according to prognostic grouping are outlined in
to PTV margin.20,22,23 In an attempt to reduce the CTV Table 26B-2.12,16,17,38 The definition of the low-risk
to PTV margin further, a number of investigators have group is relatively consistent between series but there are
localized the prostate daily prior to delivering treatment. differences in the higher risk categories across series.
Common techniques include transabdominal ultrasound This likely explains the consistent results observed in the
localization of the prostate24 and the use of implanted low-risk group, whereas the BRFS results in the higher
fiducial markers.25 risk groups are slightly different. A brief summary of each
Elective irradiation of the pelvic lymph nodes has report follows.
been debated for more than 30 years. In men with a low Shipley et al.38 and others published a pooled analysis
risk of pelvic lymph node involvement according to the examining BRFS following EBRT in 1999. In this report,
Partin tables26 or the Roach formula,27 it is difficult to data from 1765 patients with T1-2 prostate cancer
justify treating the lymph nodes. In men with more treated between 1988 and 1995 at six separate institutions
advanced disease, a recent RTOG trial argues in favor of were collected and analyzed. None of these patients
including the pelvic lymph nodes.28 Patients with a risk of were treated with ADT. Fifty-one percent of patients were
pelvic lymph nodes of >15% were enrolled in this four- treated with 3DCRT. The median follow-up was 4.1
arm trial. Preliminary results indicate that elective radia- years and the minimum follow-up was 24 months. The
tion therapy to the pelvic lymph nodes when combined ACD was used to calculate BRFS. The 5-year estimate of
with neoadjuvant androgen deprivation improves BRFS. BRFS for all patients was 65.8% (95% CI 62.8 to 68).
In the absence of compelling data to argue the contrary, Recursive partitioning analysis of pretreatment PSA
in patients receiving radiation therapy with a risk of level, clinical T-stage, and Gleason score produced four
480 Part V Prostate Gland and Seminal Vesicles

Table 26B-2 Five-year BRFS Following EBRT for T1-2 Prostate Cancer According to Prognostic Group
No. Median Median Prognostic 5-Year Reference
Authors Patients Tx Dates Follow-Ups Dose (Gy) Group BRFS No.

Shipley et al. 1765 1988–1995 4.1 years 69.4 I 81 38

II 69

III 47

IV 29

Zelefsky et al. 743 1988–1995 3 years 75.6 Favorable 85 17

Intermediate 65

Unfavorable 35

Kupelian et al. 628 1990–1998 51 months 70.2 Favorable 90* 12

Unfavorable 59*

D’Amico et al. 381 1988–2000 3.8 years 70.4 Low 78 16

Intermediate <34% 65

Intermediate >34% 35

High 40

*Eight-year BRFS.

prognostic groups. According to prognostic group the Kupelian et al.12 have published the results of 628 men
5-year BRFS ranged from 81% to 29%. Only 5% of bio- with T1-2 treated with EBRT at the Cleveland Clinic
chemical relapses occurred beyond 5 years. This paper between 1990 and 1998. Patients were treated to doses
provides a good benchmark of the results achievable with between 68 and 78 Gy (median 70.2 Gy). Conformal
EBRT in the early 1990s. techniques were used in 321 (51%) of patients. Twenty-
Zelefsky et al.17 have reported on 743 men treated at three percent of patients were treated with ADT for a
the Memorial Sloan-Kettering Cancer Center between median of 6 months. The median follow-up was 51
1988 and 1995. These men were treated according to a months and an average of 8.7 PSA levels were obtained
phase I dose-escalation study to assess the toxicity of per patient in follow-up.
high-dose 3DCRT. Ninety-six men were treated to 68.4 The 8-year rate of BRFS for all patients was 70%. On
Gy, 266 treated to 70.2 Gy, 320 to 75.6 Gy, and 61 multivariable analysis T stage, pretreatment PSA,
patients treated to 81 Gy. One hundred and ninety-five Gleason score, and neoadjuvant ADT were all independ-
(26%) patients were in clinical stage T3. Two hundred ent predictors of BRFS. The authors divided patients
and thirteen patients (29%) received neoadjuvant andro- into a favorable group (T1-2a, Gleason score < 6, and
gen deprivation therapy (ADT) to reduce the size of the pretreatment PSA < 10) and an unfavorable group (T2b
prostate gland and reduce the total dose to the bladder or T2c, or Gleason score > 6, or pretreatment PSA > 10).
and rectum. The median follow-up was 36 months with The 8-year BRFS were 90% and 59% for the favorable
22% of patients followed for 4 to 8 years. and unfavorable groups respectively. In the unfavorable
The 5-year rate of BRFS was dependent on T stage group, total doses above 72 Gy were associated with
(T1-2 versus T3), pretreatment PSA (<10 versus = 10), and improved BRFS, 75% versus 41% (p < 0.001).
Gleason score (= 6 versus >6). Patients without poor risk D’Amico and colleagues have reported a retrospective
features (T1-2, PSA < 10, Gleason score = 6) experienced a cohort study of 381 men treated with EBRT between
BRFS of 85%. The 5-year BRFS in men with a single poor 1988 and 2000.16 All men were treated with conformal
risk feature was 65% and 35% in men with more than one methods. The median central axis dose was 70.4 Gy. No
poor risk feature. Prescription dose of greater than or equal patient received ADT. The median follow-up is 3.8 years
to 75.6 Gy was also an independent predictor of BRFS. (range 1 to 12.9 years).
Chapter 26B Clinically Localized Adenocarcinoma of the Prostate: Radiation Therapy 481

The estimates of BRFS at 8 years following treatment a pretreatment PSA above 10 ng/ml, the BRFS was 62%
were dependent on prognostic group. The 8-year BRFS in the 78-Gy arm versus 42% in the 70-Gy arm, p = 0.03
in the low-risk group (T1-2a, Gleason score < 7, and pre- (Figure 26B-2B). In patients with a PSA below 10 ng/ml,
treatment PSA < 10) was 78% (95% CI 72 to 83). The there was no evidence of a treatment effect according to
intermediate-risk group (PSA > 10 but <20, Gleason score radiation dose with BRFS of 75% in both arms, p = non-
7, or T-2b) was divided further according to the percent significant (NS) (Figure 26B-2C).
positive biopsies (<34% versus >34%). The 8-year BRFS As the total dose to the prostate gland increases, the
in the low volume intermediate-risk group was 65% (95% possibility of increased rectal or urinary morbidity has
CI 58 to 72) compared to 35% (95% CI 12 to 55) in the been examined in a number of studies. One prospective
high volume intermediate-risk group. The 8-year BRFS trial has concluded that 3DCRT produces fewer late
was 40% (95% CI 28 to 52) in the high-risk group. effects than conventional techniques when similar doses
Summarizing this information, it is clear that pretreat- are used.44 The dose-limiting toxicity in most reports of
ment variables can be combined to create prognostic dose escalation appears to be rectal bleeding.45–47 The
groups. In men with favorable-risk disease, EBRT is time course of the development of gastrointestinal com-
associated with 5-year BRFS of 78% to 90%. Men with plications is shorter than the time course of the develop-
intermediate- and high-risk features experience 5-year ment of genitourinary complications so that late bladder
BRFS at 5 years of approximately 65% and 35%, respec- toxicity may not yet be evident. In the randomized trial
tively. EBRT alone to conventional doses (70 Gy) from M.D. Anderson Hospital of 78 Gy versus 70 Gy, the
appears insufficient in men with T1-2 disease and inter- rate of Grade 2 or greater rectal complications was
mediate- or high-risk features. higher in the 78-Gy arm, 26% versus 12% (p = 0.001).43
The results from this trial and other reports indicate that
rectal injury following 3DCRT is a function of dose and
THE EVIDENCE FOR DOSE ESCALATION
volume.43,45–47
Several reports from patients treated in the pre-PSA era The RTOG has recently completed a multi-institu-
suggested that improved results were possible in men tional phase I/phase II dose-escalation trial of 3DCRT
with locally advanced and/or high-grade disease with (RTOG 94-06). Late toxicity of the first three dose lev-
doses above 70 Gy.39,40 In fact, a randomized trial from els (68.4, 73.8, and 79.2 Gy) has been published.48,49 To
Shipley and investigators at the Massachusetts General date no Grades 4 to 5 toxicities have been observed and
Hospital demonstrated improved local control in patients the rate of Grade 3 toxicity is less than 5%. The rates of
with high-grade disease when a proton boost [to 75.6 GI/GU morbidity are lower than observed with historic
cobalt Gray equivalent (CGE)] was added to photon irra- controls to lower dose levels using conventional
diation.41 approaches. This observation has led to the development
A number of contemporary series provide evidence and activation of a randomized phase III trial comparing
that doses above 72 Gy may benefit patients with T1-2 79.2 to 70.2 Gy (RTOG P-0126).
prostate cancer.12,17,42 Results from these series are out- The improved conformality of IMRT compared to
lined in Table 26B-3. All of these series are nonrandom- 3DCRT offers the possibility of further dose escalation
ized and the follow-up is generally shorter in men treated without an increase in normal tissue injury. One of the
to higher doses. The ASTRO definition is very sensitive largest experiences with this new approach has been
to length of follow-up making the conclusions from these described by Zelefsky et al.50 In this report, the early tox-
nonrandomized reports less than definitive. icity and biochemical results from 772 men treated at
The investigators at M.D. Anderson Hospital have Memorial Hospital between 1996 and 2001 are outlined.
reported 5-year results from a randomized trial compar- Ninety percent of these men were treated to a dose of 81
ing conventional dose (70 Gy) EBRT to high-dose (78 Gy and 10% were treated to 86.4 Gy. The late rectal and
Gy) EBRT.43 Three hundred and one men were random- urinary toxicity was much lower than expected. The actu-
ized and are eligible for analysis. Patients were treated arial probability of Grade 2 or greater rectal toxicity was
between 1993 and 1998. The primary endpoint of the 4%, while the analogous rate of Grade 2 or greater uri-
trial was BRFS. The two arms were well matched with nary toxicity was 15% with most of the urinary toxicity
respect to known prognostic factors. manifest as urethra symptoms requiring medication.
With a median follow-up of 60 months, the 6-year Although the follow-up is relatively short (24 months),
BRFS was improved in the 78-Gy arm versus the 70-Gy the BRFS is encouraging. The 3-year rates of BRFS is
arm, 70% versus 64%, p = 0.03 (Figure 26B-2A). A sub- 92%, 86%, and 81% according to the low-, intermedi-
set analysis using a stratification variable included in the ate-, and high-risk groups, respectively. Preliminary
randomization process indicated that the benefit from results from other institutions are available51 but longer
the high-dose treatment was restricted to patients with a follow-up is required to assess 5 to 10-year BRFS and
pretreatment PSA above 10 ng/ml. In those patients with long-term morbidity.
Figure 26B-2 A, BRFS curves of M.D. Anderson phase III randomized trial, including all
patients. B, BRFS curves of M.D. Anderson phase III randomized trial, including only
patients with a pretreatment PSA greater than or equal to 10 ng/ml. C, BRFS curves of M.D.
Anderson phase III randomized trial, including only patients with a pretreatment PSA less
than 10 ng/ml.

Table 26B-3 Five-Year BRFS Following EBRT for Prostate Cancer According to Prescription Dose
No. Median Dose Prognostic 5-year Reference
Authors Patients Tx Dates Follow-ups Group (Gy) Group BRFS No.

Hanks et al. 618 1988–1997 53 month Low (<72.5) PSA <10, favorable 77 42

High (=72.5) PSA <10, favorable 89

Low (<76) PSA <10, unfavorable 70

High (=76) PSA <10, unfavorable 92

Low (<76) PSA 10–19.9, favorable 72

High (=76) PSA 10–19.9, favorable 86

Low (<76) PSA 10–19.9, unfavorable 51

High (=76) PSA 10–19.9, unfavorable 82

Low (<76) PSA >20, favorable 23

High (=76) PSA >20, favorable 63

Low (<76) PSA >20, unfavorable 29

High (=76) PSA >20, unfavorable 26

Zelefsky et al. 530 1988–1995 36 months Low (64–70) Low risk 83 17

High (75–81) Low risk 95

Low (64–70) Intermediate risk 55

High (75–81) Intermediate risk 78

Low (64–70) High risk 23

High (75–81) High risk 53

Kupelian et al. 738 1986–1999 45 months Low (<72) Favorable 81 12

High (=72) Favorable 98

Low (<72) Unfavorable 41

High (=72) Unfavorable 75


Chapter 26B Clinically Localized Adenocarcinoma of the Prostate: Radiation Therapy 483

These capsules were far smaller than the radium capsules


ADT AND EXTERNAL RADIATION THERAPY FOR
and allowed for easier placement. In the 1960s,
T1-2 PROSTATE CANCER
Whitmore and Carlton popularized the open retropubic
The results from nonrandomized historic comparisons of approach using permanent I-125 sources.59,60 The mod-
patients treated with EBRT with or without ADT have ern closed IB procedure was originally described by
suggested a benefit to ADT,12,52 although this observa- Holm et al.61 and was further developed by Blasko et al.62
tion has not been universal.17 It is possible that the effect The present widespread utilization of permanent
of ADT is not observed in the Memorial series because prostate brachytherapy (PPB) is the result of prostate
of the consistently high prostate doses. These results that screening and improved technology that currently allows
demonstrate a benefit to ADT combined with EBRT for an outpatient procedure that generally can be accom-
in T1-2 prostate cancer must be considered hypothesis plished in 1 to 2 hours.
generating as often the follow-up is shorter in the ADT The earliest prostate brachytherapy procedures using
group that will lead to a perceived improvement in BRFS radium or radon involved temporary sources. In the
using the ACD. 1970s low-dose rate Ir-192 was used as a temporary
The results from four phase III randomized trials prostate brachytherapy (TPB) source.63,64 This proce-
comparing EBRT alone to EBRT combined with ADT dure required surgical exposure of the prostate. The
in men with locally advanced disease have been pub- open procedure allowed for guidance of transperineal,
lished.53–56 In all studies, the addition of ADT has pro- template aided after-loading needles into the prostate
duced improved local control, decreased rates of distant gland. The needles were manually directed by way of a
metastases, and improved disease-free survival. Overall suprapubic surgical incision. After the needles were posi-
survival was improved in two studies.53,54 Although the tioned, Ir-192 ribbons were manually loaded. The typical
inclusion criteria are different for each of these three dose-rate ranged from 0.5 to 1.0 Gy per hour delivering
studies, most patients included in these studies had a total dose of 30 to 35 Gy over 2 to 3 days. EBRT (35 to
locally advanced disease. For instance, only 9% of 45 Gy) would follow. The initial results with this proce-
patients in the EORTC study had T1-2 tumors and none dure were encouraging but several disadvantages, includ-
of the patients in RTOG 86-10 had nonpalpable disease. ing the necessity for an open procedure and radiation
The magnitude of benefit for the addition of ADT to exposure from manual loading, have limited the use of
EBRT in men with T1-2 prostate cancer is unknown. temporary LDR prostate brachytherapy to very few cen-
Randomized trials from the RTOG and the Dana-Farber ters at present.
Cancer Center investigating the impact of short-term
(4 to 6 months) neoadjuvant ADT in men with T1-2
TECHNIQUE OF PERMANENT PROSTATE
prostate have been completed but have yet to be
BRACHYTHERAPY
reported.
The modern technique of PPB includes three compo-
nents: (1) treatment planning, (2) placement of the
HISTORIC PERSPECTIVE, INTERSTITIAL
sources, and (3) an evaluation of the implant quality. In
BRACHYTHERAPY
the early work of Ragde and Blasko, the three compo-
Brachytherapy is a general term used to describe the nents were separated in time but advances in imaging and
placement of a radioactive source(s) into or near (Gk. treatment planning software have led to these compo-
“Brachy” = near) a tumor. The source can be placed per- nents being compressed. In some centers, all three com-
manently or reside temporarily. Brachytherapy is used in ponents are completed in the operating room at the same
the treatment of many solid tumors, including breast, sitting.65 Brachytherapy treatment planning determines
head and neck, gynecologic, and thoracic neoplasms. the optimal radiation dose distribution for an individual
The history of prostate brachytherapy extends nearly 100 patient depending on the size and shape of the prostate
years beginning in the first decade of the 20th century in gland. A number of different dose distribution philoso-
Paris. A number of renowned urologists have contributed phies have been described. Most practitioners will use
to the history of prostate brachytherapy. some sort of peripheral loading scheme to reduce the
The first technique was described by Pasteau and dose to the urethra and maximize dose to the peripheral
Degrais in 1911.57 Their approach relied on temporary zone. In patients with very low-risk features, some authors
intraurethral placement of radium capsules. Soon there- have used MR guidance to target only the peripheral
after, Hugh Hampton Young obtained some radium and zone of the prostate gland.66 Transrectal ultrasound
modified the Paris technique by designing his own device images are generally used for the treatment planning but
to hold the radium. A surgeon at the Memorial Hospital other imaging modalities have been described.
in New York, Benjamin Barringer, was the first to Two radioisotopes are in common use in the United
describe an interstitial approach using radon capsules.58 States: I-125 and Pd-103. Each of these isotopes possesses
484 Part V Prostate Gland and Seminal Vesicles

Table 26B-4 Prescription Doses (Gy) for PPB


According to Isotope
Dose (Gy), Dose (Gy),
Radioisotope PPB Alone PPB plus EBRT*

I-125 144 100–110

Pd-103 115–125 80–90

*Combined with EBRT (40–50 Gy).

a low-dose rate and low energy relative to the isotope


used for temporary brachytherapy (Ir-192). The radiation
dose prescribed differs according to isotope and whether
brachytherapy is used alone (monotherapy) or combined
with EBRT (combined modality therapy, CMT) (Table
26B-4).67 The low energy of the sources simplifies radia-
tion protection precautions and patients can be dis-
charged from the hospital immediately.
The radioactive sources are usually placed transper-
ineally using some form of perineal template and ultra-
sound guidance (although MR- and CT-guided
approaches have been described). Two techniques of
source deposition are commonly employed. In the pre-
loaded technique, brachytherapy needles with sources
and spacers are prepared beforehand and with each nee-
dle placement multiple sources are deposited. In the
afterloading approach, a specially designed brachyther-
apy “gun” is used to deposit one source at a time. In
either case, the needle or gun can be visualized and
placed in the desired location. Close monitoring of the
source deposition process allows the operator to recog-
nize and adjust for changes that may occur intraopera-
tively (prostate gland movement, prostate gland swelling,
and source movement).
Some form of postprocedure dosimetric evaluation is B
mandatory.68 Most investigators use CT for this assess- Figure 26B-3 Postimplant CT scan images through the mid-
ment but other methods have been described. With com- gland (A) with resulting dose-volume histogram (B). From the
mercially available software it is possible to import CT dose-volume histogram it is clear that 94% of the prostate is
images and outline the prostate gland and nearby struc- encompassed by the 100% isodose line, while the vast
tures (urethra, rectum, penile bulb, etc.). Automated majority of the urethra is receiving less than 150% of the
algorithms have been designed to identify the sources. prescription dose.
Based on the delineation of the prostate and the location
of the sources, isodose distributions can be calculated and
dose-volume histograms created (Figure 26B-3). The emerging, acceptable doses to normal tissues (urethra,
accurate delineation of prostate volumes on CT images rectum, penile bulb) are much less clear.
following brachytherapy can be difficult. Postoperative
prostate swelling and degradation of the image secondary
TECHNIQUE OF TEMPORARY PROSTATE
to the metallic sources can lead to disagreement amongst
BRACHYTHERAPY
reviewers.69
A standard definition of a “good implant” has yet to be TPB also benefitted from the advances in ultrasound and
universally accepted by the brachytherapy community. treatment planning computers in the 1980s. The simul-
There is early evidence that delivering at least 90% of the taneous development of high-dose rate afterloading
prescription dose to 90% of the prostate gland results in machines stimulated a number of investigators across the
improved BRFS.70,71 Although preliminary work is globe to refine the methods of TPB.
Chapter 26B Clinically Localized Adenocarcinoma of the Prostate: Radiation Therapy 485

High-dose rate (HDR) afterloading machines are


BIOCHEMICAL RELAPSE-FREE SURVIVAL
devices that contain a single high-intensity Ir-192 source
FOLLOWING PROSTATE BRACHYTHERAPY
(5 to 10 Ci). The source is attached to the end of a wire
that is controlled by the machine. The machine can pre- As with patients treated with EBRT, serum PSA levels
cisely position the source and keep it there for a specified decline soon after treatment with PPB. The ACD is
period of time (usually measured in seconds). The source commonly used to define BRFS following PPB, although
can thus be moved within needles to a number of loca- the ACD was not originally intended for this purpose.
tions allowing the treating physician to create a dose dis- Some advocate the use of a nadir definition following
tribution of virtually any shape or size. PPB instead of the ACD.74 The results outlined in this
Most HDR prostate brachytherapy techniques utilize section use a number of different definitions to define
transrectal ultrasound to guide the needles into place BRFS making comparisons problematic.
within and around the prostate.72 A variety of perineal Compared to men treated with EBRT, men treated
templates have been devised to assist needle placement. with PPB are more likely to manifest a sudden increase in
In some centers, ultrasound-based treatment planning serum PSA to be followed by a decline. Authors have
allows treatment to be delivered at the time of the needle described this phenomenon as PSA bounce or PSA
placement.73 Most sites use CT images for the treatment spike.75–77 The incidence of this phenomenon ranges
planning process. HDR needles are identified, and the from 17% to 35% depending on the definition utilized.
prostate gland is contoured on the CT images. Nearby The median time to PSA spike is 18 to 24 months and
structures (urethra, rectum) are identified, and a the mostly occurs within the first 5 years. The magnitude of
desired dose is delivered to the prostate while limiting PSA bounce has been as large as 15 ng/ml, although most
the dose to the urethra and rectum. bounces are less than 3 ng/ml. Two reports find PSA
Once a plan has been generated, the afterloading spikes to be more common in younger men, which may
machine delivers the treatment by moving the source to reflect an increased level of sexual activity.76,77 To date in
different locations within the HDR catheters in a three separate reports, there has been no correlation
sequential manner. Typical treatment times range from between PSA bounce and subsequent BRFS.75–77
5 to 10 minutes. Typically patients receive multiple treat- Clinicians should be aware of this phenomenon and exer-
ments over the course of 6 to 36 hours. Due to the high cise caution prior to instituting salvage therapy.
energy of the Ir-192 source, a specially shielded room is The results described later are divided according to
required for treatment. whether patients are treated with monotherapy (PPB
Most clinicians have combined HDR brachytherapy alone) or CMT (PPB plus EBRT). CMT has been rec-
with 4 to 5 weeks of EBRT although HDR monotherapy ommended over monotherapy for a number of reasons:
is taking place in a few centers. Dose-fractionation increased intraprostatic dose; treatment of extracapsular
schedules differ between institutions and despite prostate cancer; and as a form of insurance to “fill in” the
attempts, no consensus has been reached as to the best areas of reduced dose that may result from a suboptimal
treatment schedule. implant. Some institutions use CMT selectively in men

Table 26B-5 BRFS Following PPB for T1-2 Prostate Cancer


No. Percentage Percentage of Median Follow-ups BRFS
Authors Patients P/I of T1/T2 Gleason Score > 6 PSA (months) (years)

Stock86 97 I/P 13/87 18 NS 18 76 (2)

Grado et al.83 392 I/P 6/92 20 7.3 30 80 (5)

D’Amico16 66 P 23/77 20 NS 41 0–85 (5)

Blasko9 230 P 30/70 40 7.3 41.5 83.5 (9)

Ragde85 147 I 22/78 0 8.8* 93 66 (12)

Zelefsky15 248 I 58/42 25 7 48 71 (5)

Brachman et al.82 695 I/P 17/83 15 NS 51 64 (7)

Grimm84 125 I 24/76 0 5.1 ~78 85 (10)


486 Part V Prostate Gland and Seminal Vesicles

with high-risk features,78 whereas others use CMT in all PSA. The 5-year estimate of disease-free survival for all
patients regardless of T stage, PSA, and Gleason score.79 patients was 71%. The 5-year estimates of disease-free
The necessity of supplemental EBRT in patients survival according to prognostic risk groups were as fol-
treated with PPB has yet to be demonstrated. No ran- lows: 88% for favorable risk; 77% for intermediate risk;
domized trials have been reported. The addition of sup- and 38% for high risk (p < 0.0001).
plemental EBRT to PPB increases patient’s and Grimm et al.87 recently published an update of the
physician’s time commitment with a resulting increase in Seattle experience with prostate brachytherapy as
costs.80 There is also the possibility that combination monotherapy. The authors reported on 125 men treated
therapy will increase morbidity and decrease HRQOL between January 1998 and December 1990 with I-125
compared to PPB or EBRT alone.81 The hypothesis that prostate brachytherapy alone. These men were carefully
supplemental EBRT improves results is currently being selected and had very favorable pretreatment characteris-
tested in a recently activated randomized trial of the tics. The clinical T stage was less than or equal to T2a in
RTOG (RTOG P-0232). This trial includes measure- 85% of men, and all men had a Gleason score of 6 or less.
ments of patient-reported HRQOL. Forty-three percent of men had a pretreatment PSA
below 4 ng/ml and more than 75% had a pretreatment
PSA below 10 ng/ml. Patients were routinely followed
BIOCHEMICAL RELAPSE-FREE SURVIVAL:
with physical examination and serum PSA determina-
PPB ALONE
tions at 3 to 6-month intervals during the first 5 years
Table 26B-5 summarizes the results achieved with PPB and yearly thereafter. The median follow-up was 81.4
alone in men with clinically localized prostate can- months. These authors used two consecutive PSA rises as
cer.9,10,15,82–86 PSA-based disease-free survival is 63% to evidence of failure. Disease-free survival was estimated
93% at 5 years in selected patients treated with PB alone. by the Kaplan–Meier method. Eight men were docu-
Two published reports with the longest follow-up report mented to have clinical failures (4 with positive prostate
10-year actuarial estimates of BRFS to be 66% to biopsy and 4 with positive bone scan). All clinical failures
85%.85,87 Brief descriptions of a few selected series are were diagnosed within 5 years. Eight additional patients
provided later. were found to have biochemical evidence of disease
Blasko et al.9 have reported on a consecutive series of recurrence. The Kaplan–Meier estimate of biochemical
230 men with T1-2 prostate cancer treated with Pd-103 disease-free survival in this cohort is 85.1% (95% CI 79.3
alone at the Seattle Prostate Institute between 1988 and to 90.9) at 10 years. The only variable that predicted for
1995. The majority of patients (56%) presented with pal- BDFS in this cohort was pretreatment PSA.
pable T2a disease and 40% were classified as having a The authors then provided a comparison to a cohort
Gleason score of 7 or higher. The median PSA was of 97 men treated by the same authors early in their
7.3 ng/ml. Seventy-five percent of men had a pretreat- prostate brachytherapy experience. The authors reported
ment PSA below 10 ng/ml. Two consecutive PSA eleva- on 97 men treated between January 1986 and December
tions were required for biochemical recurrence. The 1987. The demographic characteristics of this earlier
median follow-up was 41.5 months. The estimate of cohort were similar to the later cohort, but the BDFS was
BRFS was 83.5% (95% CI 78.3 to 88.7) at 9 years. significantly worse in the men treated in 1986 and 1987
Pretreatment PSA and Gleason score were found to be (10 Yr BDFS 65% versus 87%, p = 0.0002). The authors
predictive of BRFS on univariate analysis. Grouping concluded that the superior results in the later patients
patients into three risk categories according to T stage, were the result of refinements in the prostate brachyther-
Gleason score and pretreatment PSA value demonstrated apy technique.
significant differences in BRFS. In weighing the available evidence about the efficacy
Zelefsky et al.15 have provided an update of the expe- of PPB, several important points should be considered.
rience at Memorial Hospital. These authors reported on Each and every report represents a retrospective, single-
248 men treated with prostate brachytherapy alone institution experience. The extent to which the results
between 1989 and 1996. All men were treated with I-125. achieved at centers of excellence are generalizable to
The median age was 65. Most men (58%) had nonpalpa- general community practice is unknown. The Seattle
ble disease. Twenty-four percent of men had a Gleason group has provided the best evidence that a learning
score of 7 or higher. The median pretreatment PSA was curve exists for the PB procedure in that the BRFS rates
7 ng/ml, and 78% of men had a PSA of less than 10 are inferior in the group of patients treated early in their
ng/ml. Three rises in PSA was defined as evidence of experience despite similar pretreatment characteristics.
recurrence, and the median follow-up was 48 months Differences in patient selection criteria, brachytherapy
(range 12 to 126 months). The authors categorized techniques, experience of the brachytherapists, and def-
patients into previously published prognostic groups initions of biochemical relapse preclude any definitive
according to T stage, Gleason score, and pretreatment statements comparing techniques, isotopes, etc. From an
Chapter 26B Clinically Localized Adenocarcinoma of the Prostate: Radiation Therapy 487

Table 26B-6 Five-Year BRFS Following PPB for T1-2 Prostate Cancer
According to Prognostic Groups
Authors No. Follow-Ups 5-Year
Patients (Months) Isotope Risk Category BRFS (%)

Blasko9 103 48.9 Pd-103 Low 94

107 39.5 Intermediate 82

20 45.5 High 65

Zelefsky15 112 I-125 Favorable 88

92 Intermediate 77

22 Unfavorable 38

Grimm84 97 I-125 Low 87*

27 Intermediate 79*

D’Amico16 32 41.0 Pd-103 Low 88

15 Intermediate 32

19 High NA

*Ten-year BRFS.

Table 26B-7 BRFS Following PPB Combined with EBRT for T1-2 Prostate Cancer
No. Implant EBRT EBRT Follow-Ups BRFS (%)
Author Patients Isotope(s) MPD* (Gy) Dose (Gy) Fields (Years) (Years)

Ragde85 82 125-I 110 45 4-field: P-SV 10 79 (10)

Blasko78 231 125-I/103-Pd 110/90 45 4-field: P-SV 5 79 (9)

Lederman et al.89 348 125-I/103-Pd 110/90 45 4-field 4 77 (6)

Critz79 689 125-I 120 45 P-SV-PPT 4 88 (6)

Dattoli et al.88 124 103-Pd 90 41 Limited Pelvic 3.8 76 (4)

*Matched peripheral dose.

evidence-based perspective, the value of these results have 5-year BRFS of 32% to 65%, suggesting that PPB
should be considered limited; multi-institutional, alone is not adequate in these higher risk patients. This
prospective trials are required. observation has led many authors to recommend supple-
mental EBRT or ADT (or both) in these men. Results
with CMT are outlined in the following section. The use
PSA OUTCOME STRATIFIED BY RISK GROUP
of ADT with PPB is discussed as follows.
Just as in patients treated with EBRT, results following
PPB according to prognostic grouping have been pub-
BIOCHEMICAL RELAPSE-FREE SURVIVAL: PPB
lished. Table 26B-6 summarizes the available information
COMBINED WITH EBRT
from those authors who reported results with PPB alone
according to risk groups.9,10,15,87 The 5-year BRFS ranges The results from five centers with a significant experi-
from 88% to 94% in men with low-risk disease treated ence with CMT (PPB plus EBRT) are outlined in Table
with PPB alone. Patients with more unfavorable features 26B-7.78,79,85,88,89 All patients in these reports were treated
488 Part V Prostate Gland and Seminal Vesicles

after 1986 and have had continuous PSA follow-up avail- and eighty-nine men were treated between 1992 and
able. The closed transperineal method of PPB was used 1996 and received an I-125 implant (120 Gy) followed in
in all cases. Three reports originate from centers where 3 weeks by EBRT (45 Gy). The median follow-up was
CMT is used in all prostate cancer cases,79,88,89 and two 4 years. Twenty-eight (4%) men had a Gleason score
are from institutions that select CMT for higher risk above 7 and 44 (6%) had a pretreatment PSA above 20.
patients, reserving PPB alone for patients with low-risk No patient received ADT. As is the custom with these
disease.78,85 investigators, the ACD was not used to calculate BRFS.
Ragde et al.85 have described the results in 82 men Instead, an absolute value of = 0.2 was used to define cure.
treated with PPB (I-125, 110 Gy maximum permissible Patients are censored if the PSA is declining (but not
dose [MPD]) and EBRT (45 Gy). All men were treated =0.2) at the time of analysis. The 5-year estimate of
between January 1987 and December 1989 so that the BRFS is 88%.
follow-up is quite long (median 122 months). These Dattoli et al.88 have recently updated their experience
patients were selected for CMT based on high-risk fea- with CMT in patients with high-risk features. They have
tures. The vast majority of men (84%) had palpable dis- reported on 161 men with T1-3 prostate cancer treated
ease, although only four men were felt to have clinical with PPB (Pd-103, 80 Gy) and EBRT (41.4 Gy). The PPB
extracapsular disease (T3). The mean PSA in this group was performed for 4 weeks following EBRT. The median
was 14.7 ng/ml (sd 21.2), and 18 men (24%) had a follow-up for nonfailing patients was 7 years.
Gleason score above 6. ADT was not used. The ACD Approximately 25% of patients had a pretreatment PSA
was used to calculate BRFS. The 10-year estimate of above 20. Approximately 20% of the patients were assigned
BRFS was 79%. a Gleason score above 7. The ACD was not used calculate
Blasko et al.78 have reported on 231 men treated with BRFS. Instead, a PSA level of =0.2 ng/ml defined the
PPB (I-125, 110 Gy; Pd-103, 90 Gy) and EBRT (45 Gy). absence of disease. The 10-year estimate of BRFS is 79%.
The median follow-up was 58 months. During this The BRFS from two series that provide results
period, some men were treated with monotherapy, and according to prognostic grouping are summarized in
the use of supplemental EBRT was at the discretion of Table 26B-8.78,89 Although the amount of information is
the physician. The mean PSA was 15.6 ng/ml, and 35% small, it appears that patients with favorable (and perhaps
of patients presented with a Gleason score above 7. No intermediate-) risk disease do well with CMT, but
patients received ADT. The ACD was used to calculate patients with high-risk features do not fare nearly as well,
BRFS. The results of BRFS were provided according to suggesting that they may benefit from effective systemic
the prognostic groupings of Zelefsky. Ten-year BRFS therapy combined with local therapy.
estimates were 87%, 85%, and 62% for low-, intermedi-
ate-, and high-risk groups, respectively. In this report,
ADT AND PERMANENT PROSTATE
the authors did compare the results with CMT to
BRACHYTHERAPY FOR T1-2 PROSTATE
monotherapy according to risk group and found no
CANCER
group that benefitted from the addition of EBRT.
Lederman et al.89 have reported on 348 patients with The use of ADT combined with PPB is common. In
T1-3a disease treated with PPB (I-125, 110 Gy; Pd-103, recent reports, ADT has been combined with PPB in 7%
90 Gy) and EBRT (45 Gy). In this report, PPB was per- to 69% of cases.90 ADT may be utilized for three pur-
formed prior to EBRT. No patient received ADT. The poses: (1) to downsize the prostate to facilitate PPB; (2)
median follow-up was 44 months. One hundred and to decrease morbidity; and (3) to improve cure rates. It is
sixty-four (50%) men had a pretreatment PSA above 10 clear that 3 to 6 months of ADT will lead to a 30% to
ng/ml and 119 (34%) were assigned a Gleason score 40% reduction in prostate volume on transrectal US
between 7 and 10. Only 17 (4.6%) men had T3 disease. measurements.91 It is not clear that ADT reduces mor-
The ACD was not used to define BRFS. Instead, bio- bidity. In fact there at least is an evidence that ADT prior
chemical failure was defined as the use of ADT anytime to PPB may increase acute urinary morbidity and
following treatment, or a serum PSA > 5.0 ng/ml at last decrease sexual function.92,93 The question whether ADT
follow-up, or two consecutive rises in serum PSA > 0.5 improves cure rates is more complicated.
ng/ml. The estimated BRFS at 6 years for the entire Whether the results of randomized trials showing a
group was 77% (95% CI 72% to 82%). The authors benefit for the addition of ADT to EBRT in patients with
divided patients into risk groups according to pretreat- locally advanced disease can be extrapolated to lower risk
ment PSA, T stage, and Gleason score. The estimates of patients treated with PPB is not known. The results from
BRFS were 88%, 75%, and 51% for the low-, intermedi- nonrandomized historic comparisons of patients treated
ate-, and high-risk groups, respectively. with PPB with or without ADT have been inconsistent.
The results from the radiation therapy clinics of Some reports indicate an improvement in BRFS94,95 while
Georgia have been summarized recently.79 Six hundred others do not.96 Given that most authors use the ACD in
Chapter 26B Clinically Localized Adenocarcinoma of the Prostate: Radiation Therapy 489

Table 26B-8 BRFS Following PPB Combined with EBRT for T1-2 Prostate Cancer According to Prognostic Groups
No. BRFS Time Low (%) Intermediate High (%)
Authors Patients Point (years) (N) (%) (N) (N)

Blasko78 231 10 87 (75) 85 (104) 62 (52)

Lederman et al.89 348 5 88 (165) 75 (124) 51 (59)

Table 26B-9 BRFS Following HDR Brachytherapy and EBRT


No. Percentage Median Implant No. EBRT BRFS (%)
Authors Patients of T1-2 Follow-ups Dose (Gy) Fractions Dose (Gy) (years)

Borghede et al.97 50 76 45 months 20 2 50 84*

Dinges et al.98 82 26 24 months 18–20 2 45 53 (2)

Kovacs and Galalae99 144 68 8 years 30† 2 40 73 (8)‡

Mate et al.72 104 90 45 months 12–16 4 50.4 84 (5)§

Martinez100 207 91 4.4 years 16.5–23 2–3 46 74 (5)

*Eighteen-month crude rate.



Prescription dose to the peripheral zone.

Three consecutive PSA rises, all above 1.0 ng/ml.
§
For patients with pretreatment PSA < 20 ng/ml.

these reports, any results favoring the addition of ADT PARTICLE BEAM THERAPY
should be questioned. In all series, the follow-up is Particle beams used in the treatment of prostate cancer
shorter in patients receiving ADT (representing a tempo- include neutrons and photons. Early randomized trials
ral trend seen nationwide), and the ACD is extraordinar- suggested a benefit to neutron therapy in men with locally
ily sensitive to length of follow-up. Randomized trials advanced disease but an increase in complications was
examining the value of ADT combined with PPB are noted.103 The complications were attributed to inadequate
required to make conclusive statements. collimation and inability to shield normal tissues near the
prostate. It is now possible to use conformal methods with
BIOCHEMICAL RELAPSE-FREE SURVIVAL: HDR neutrons, and the results remain encouraging.104 For the
TEMPORARY PROSTATE BRACHYTHERAPY time being, however, the lack of treatment facilities with
neutron capabilities limit the use of this modality.
To date most reports of HDR brachytherapy have The number of proton facilities in the United States
focused on men with T1-2 with intermediate- or high- is similarly small, although many new sites are under
risk features (>T2b, Gleason score > 6, pretreatment PSA construction. The physical characteristics of proton
>10), and the number of men with favorable disease beams are fundamentally different than those of pho-
included in these reports is small. The BRFS results of ton beams. The dose gradient outside of the PTV is
several series are summarized in Table 26B-9.72,97–100 The much steeper with proton beams and may result in
patients included in these series are very heterogeneous. lower morbidity. The largest experience to date is from
BRFS is defined very differently with some series using investigators at Loma Linda.105 The early results
an absolute PSA level to define cure and others relying appear similar to the results achieved with 3DCRT.
on a rising PSA to define recurrent disease; only two of Comparison trials of proton beam versus photon beam
the series use the ACD to calculate BRFS. In some series, therapy are ongoing.
pretreatment ADT is given, making interpretation
difficult.
A small number of investigators have developed phase REFERENCES
II protocols to examine the efficacy of HDR brachyther-
apy alone (without EBRT).101,102 Follow-up is too short 1. Millikan R, Logothetis C: Update of the NCCN
to report BRFS, but the acute morbidity appears to be guidelines for treatment of prostate cancer. Oncology
tolerable. (Huntingt) 1997;11(11A):180–193.
490 Part V Prostate Gland and Seminal Vesicles

2. Hanks GE, More on the Uro-Oncology Research Group patients with localized prostate cancer. Int J Radiat
Report of radical surgery vs. radiotherapy for Oncol Biol Phys 2000;47(5; 7-15-2000):1261–1266.
adenocarcinoma of the prostate. Int J Radiat Oncol Biol 16. D’amico AV, Whittington R, Malkowicz SB, et al:
Phys 1988;14(5):1053–1054. Biochemical outcome after radical prostatectomy or
3. Paulson DF, Lin GH, Hinshaw W, et al: Radical surgery external beam radiation therapy for patients with
versus radiotherapy for adenocarcinoma of the prostate. clinically localized prostate carcinoma in the prostate
J Urol 1982;128(3):502–504. specific antigen era. Cancer 2002;95(2; 7-15-2002):
4. D’amico AV, Moul J, Carroll PR, et al: Cancer-specific 281–286.
mortality after surgery or radiation for patients with 17. Zelefsky, M. J., Leibel, S. A., Gaudin, P. B., et al: Dose
clinically localized prostate cancer managed during the Escalation With Three-Dimensional Conformal
prostate-specific antigen era. J Clin Oncol 2003;21(11; Radiation Therapy Affects the Outcome in Prostate
6-1-2003):2163–2172. Cancer. Int J Radiat Oncol Biol Phys 6-1-1998;41(3):
5. Roach M III, Lu J, Pilepich MV, et al: Long-term 491-500.
survival after radiotherapy alone: Radiation Therapy 18. Ray, G. R., Cassady, J. R., and Bagshaw, M. A. Definitive
Oncology Group prostate cancer trials. J Urol Radiation Therapy of Carcinoma of the Prostate.
1999;161(3):864–868. A Report on 15 Years of Experience. Radiology
6. Albertsen PC, Hanley JA, Gleason DF, et al: Competing 1973;106(2):407-18.
risk analysis of men aged 55 to 74 years at diagnosis 19. Reinstein, L. E., Wang, X. H., Burman, C. M., et al:
managed conservatively for clinically localized prostate A Feasibility Study of Automated Inverse Treatment
cancer. JAMA 1998;280(11; 9-16-1998):975–980. Planning for Cancer of the Prostate. Int J Radiat Oncol
7. Jhaveri FM, Klein EA, Kupelian PA, et al: Declining Biol Phys 1-1-1998;40(1):207-14.
rates of extracapsular extension after radical 20. Prescribing, recording, and reporting photon beam
prostatectomy: evidence for continued stage migration. therapy. ICRU Report 50. Bethesda, MD; 1993. Report
J Clin Oncol 1999;17(10):3167–3172. No.: 9.
8. Ung JO, Richie JP, Chen MH, et al: Evolution of the 21. Pickett, B., Roach, M., III, Verhey, L., et al: The Value
presentation and pathologic and biochemical outcomes of Nonuniform Margins for Six-Field Conformal
after radical prostatectomy for patients with clinically Irradiation of Localized Prostate Cancer. Int J Radiat
localized prostate cancer diagnosed during the PSA era. Oncol Biol Phys 4-30-1995;32(1):211-8.
Urology 2002;60(3):458–463. 22. Fiorino, C., Reni, M., Bolognesi, A., et al: Set-Up
9. Blasko JC, Grimm PD, Sylvester JE, et al: Palladium-103 Error in Supine-Positioned Patients Immobilized With
brachytherapy for prostate carcinoma. Int J Radiat Oncol Two Different Modalities During Conformal
Biol Phys 2000;46(4; 3-1-2000):839–850. Radiotherapy of Prostate Cancer. Radiother Oncol
10. D’amico AV, Whittington R, Malkowicz SB, et al: 1998;49(2):133-41.
Biochemical outcome after radical prostatectomy, 23. Malone, S., Szanto, J., Perry, G., et al: A Prospective
external beam radiation therapy, or interstitial radiation Comparison of Three Systems of Patient Immobilization
therapy for clinically localized prostate cancer. JAMA for Prostate Radiotherapy. Int J Radiat Oncol Biol Phys
1998;280(11; 9-16-1998):969–974. 10-1-2000;48(3):657-65.
11. D’amico AV, Whittington R, Malkowicz SB, et al: 24. Lattanzi, J., McNeeley, S., Hanlon, A., et al:
Pretreatment nomogram for prostate-specific antigen Ultrasound-Based Stereotactic Guidance of Precision
recurrence after radical prostatectomy or external-beam Conformal External Beam Radiation Therapy in
radiation therapy for clinically localized prostate cancer. Clinically Localized Prostate Cancer. Urology
J Clin Oncol 1999;17(1):168–172. 2000;55(1):73-8.
12. Kupelian PA, Elshaikh M, Reddy CA, et al: Comparison 25. Pouliot, J., Aubin, M., Langen, K. et al: (Non)-Migration
of the efficacy of local therapies for localized prostate of Radiopaque Markers Used for on-Line Localization
cancer in the prostate-specific antigen era: a large of the Prostate With an Electronic Portal Imaging
single-institution experience with radical prostatectomy Device. Int J Radiat Oncol Biol Phys 7-1-2003;56(3):
and external-beam radiotherapy. J Clin Oncol 862-6.
2002;20(16; 8-15-2002):3376–3385. 26. Partin, A. W., Kattan, M. W., Subong, E. N., et al:
13. Merrick GS, Butler WM, Galbreath RW, et al: Combination of Prostate-Specific Antigen, Clinical
H. Five-year biochemical outcome following permanent Stage, and Gleason Score to Predict Pathological Stage
interstitial brachytherapy for clinical T1–T3 prostate of Localized Prostate Cancer. A Multi-Institutional
cancer. Int J Radiat Oncol Biol Phys 2001;51 Update. JAMA 5-14-1997;277(18):1445-51.
(1; 9-1-2001):41–48. 27. Roach M III, Marquez C, Yuo HS, et al: Predicting the
14. Zagars GK, Pollack A, von Eschenbach AC: Prognostic risk of lymph node involvement using the pre-
factors for clinically localized prostate carcinoma: treatment prostate specific antigen and gleason score in
analysis of 938 patients irradiated in the prostate specific men with clinically localized prostate cancer. Int J
antigen era. Cancer 1997;79(7; 4-1-1997):1370–1380. Radiat Oncol Biol Phys 1-1-1994;28(1; 1-1-
15. Zelefsky MJ, Hollister T, Raben A, et al: Five-year 1994):33–73.
biochemical outcome and toxicity with transperineal 28. Roach M III, DeSilvio M, Lawton C, et al: Phase III
CT-planned permanent I-125 prostate implantation for trial comparing whole-pelvic versus prostate-only
Chapter 26B Clinically Localized Adenocarcinoma of the Prostate: Radiation Therapy 491

radiotherapy and neoadjuvant versus adjuvant combined 42. Hanks GE, Hanlon AL, Pinover WH, et al: Dose
androgen suppression: Radiation Therapy Oncology selection for prostate cancer patients based on dose
Group 9413. J Clin Oncol 2003;21(10; 5-15-2003): comparison and dose response studies. Int J Radiat
1904–1911. Oncol Biol Phys 2000;46(4; 3-1-2000):823–832.
29. Stamey TA, Yang N, Hay AR., et al: Prostate-specific 43. Pollack A, Zagars GK, Starkschall G, et al: Prostate
antigen as a serum marker for adenocarcinoma of the cancer radiation dose response: results of the M. D.
prostate. N Engl J Med 1987;317(15; 10-8-1987): Anderson phase III randomized trial. Int J Radiat Oncol
909–916. Biol Phys 2002;53(5; 8-1-2002):1097–1105.
30. Kaplan I, Prestidge BR, Cox RS, et al: Prostate specific 44. Dearnaley DP, Khoo VS, Norman AR, et al:
antigen after irradiation for prostatic carcinoma. J Urol Comparison of radiation side-effects of conformal and
1990;144(5):1172–1175. conventional radiotherapy in prostate cancer: a
31. Kaplan ID, Cox RS, Bagshaw MA: A model of prostatic randomised trial. Lancet 1999;353(9149; 1-23-1999):
carcinoma tumor kinetics based on prostate specific 267–272.
antigen levels after radiation therapy. Cancer 1991;68(2; 45. Hanlon AL, Schultheiss TE, Hunt MA, et al: Chronic
7-15-1991):400–405. rectal bleeding after high-dose conformal treatment of
32. Zagars GK, Pollack A: Kinetics of serum prostate-specific prostate cancer warrants modification of existing
antigen after external beam radiation for clinically morbidity scales. Int J Radiat Oncol Biol Phys 1997;38
localized prostate cancer. Radiother Oncol 1997;44(3): (1; 4-1-1997):59–63.
213–221. 46. Lee WR, Hanks GE, Hanlon AL, et al: Lateral rectal
33. Consensus Statement: Guidelines for PSA following shielding reduces late rectal morbidity following high
radiation therapy. American Society for Therapeutic dose three-dimensional conformal radiation therapy for
Radiology and Oncology Consensus Panel. Int J Radiat clinically localized prostate cancer: further evidence for a
Oncol Biol Phys 1997;37(5):1035–1041. significant dose effect. Int J Radiat Oncol Biol Phys
34. McMullen KP, Lee WR: A structured literature review 1996;35(2; 5-1-1996):251–257.
to determine the use of the American Society for 47. Storey MR, Pollack A, Zagars G, et al: Complications
Therapeutic Radiology and Oncology Consensus from radiotherapy dose escalation in prostate cancer:
Definition of Biochemical Failure. Urology preliminary results of a randomized trial. Int J Radiat
2003;61(2):391–396. Oncol Biol Phys 2000;48(3; 10-1-2000):635–642.
35. Horwitz EM, Uzzo RG, Hanlon AL, et al: Modifying 48. Michalski JM, Purdy JA, Winter K, et al: Preliminary
the American Society for Therapeutic Radiology and report of toxicity following 3D radiation therapy for
Oncology Definition of Biochemical Failure to minimize prostate cancer on 3DOG/RTOG 9406. Int J Radiat
the influence of backdating in patients with prostate Oncol Biol Phys 2000;46(2; 1-15-2000):391–402.
cancer treated with 3-dimensional conformal radiation 49. Ryu JK, Winter K, Michalski JM, et al: Interim report of
therapy alone. J Urol 2003;169(6):2153–2157. toxicity from 3D conformal radiation therapy (3D-CRT)
36. Kattan MW, Fearn PA, Leibel S, et al: The definition of for prostate cancer on 3DOG/RTOG 9406, level III
biochemical failure in patients treated with definitive (79.2 Gy). Int J Radiat Oncol Biol Phys 2002;54
radiotherapy. Int J Radiat Oncol Biol Phys 2000;48(5; (4; 11-15-2002):1036–1046.
12-1-2000):1469–1474. 50. Zelefsky MJ, Fuks Z, Hunt M, et al: High-dose intensity
37. Vicini FA, Kestin LL, Martinez AA: The importance modulated radiation therapy for prostate cancer: early
of adequate follow-up in defining treatment success toxicity and biochemical outcome in 772 patients. Int J
after external beam irradiation for prostate cancer. Radiat Oncol Biol Phys 2002;53(5; 8-1-2002):
Int J Radiat Oncol Biol Phys 1999;45(3; 10-1-1999): 1111–1116.
553–561. 51. Mohan DS, Kupelian PA, Willoughby TR: Short-course
38. Shipley WU, Thames HD, Sandler HM, et al: intensity-modulated radiotherapy for localized prostate
Radiation therapy for clinically localized prostate cancer with daily transabdominal ultrasound localization
cancer: a multi-institutional pooled analysis. JAMA of the prostate gland. Int J Radiat Oncol Biol Phys
1999;281(17; 5-5-1999):1598–1604. 2000;46(3; 2-1-2000):575–380.
39. Hanks GE, Martz KL, Diamond JJ: The effect of dose 52. D’amico AV, Schultz D, Loffredo M, et al: Biochemical
on local control of prostate cancer. Int J Radiat Oncol outcome following external beam radiation therapy with
Biol Phys 1988;15(6):1299–1305. or without androgen suppression therapy for clinically
40. Perez CA, Walz BJ, Zivnuska FR, et al: Irradiation of localized prostate cancer. JAMA 2000;284(10; 9-13-2000):
carcinoma of the prostate localized to the pelvis: analysis 1280–1283.
of tumor response and prognosis. Int J Radiat Oncol Biol 53. Bolla M, Collette L, Blank L, et al: Long-term results
Phys 1980;6(5):555–563. with immediate androgen suppression and external
41. Shipley WU, Verhey LJ, Munzenrider JE, et al: irradiation in patients with locally advanced prostate
Advanced prostate cancer: the results of a randomized cancer (an EORTC study): a phase III randomised trial.
comparative trial of high dose irradiation boosting with Lancet 2002;360(9327; 7-13-2002):103–106.
conformal protons compared with conventional dose 54. Granfors T, Modig H, Damber JE, et al: Combined
irradiation using photons alone. Int J Radiat Oncol Biol orchiectomy and external radiotherapy versus
Phys 1995;32(1; 4-30-1995):3–12. radiotherapy alone for nonmetastatic prostate cancer
492 Part V Prostate Gland and Seminal Vesicles

with or without pelvic lymph node involvement: a 67. Nag S, Beyer D, Friedland J, et al: American
prospective randomized study. J Urol 1998;159(6): Brachytherapy Society (ABS) recommendations for
2030–2034. transperineal permanent brachytherapy of prostate
55. Lawton CA, Winter K, Murray K, et al: Updated results cancer. Int J Radiat Oncol Biol Phys 1999;44
of the phase III Radiation Therapy Oncology Group (4; 7-1-1999):789–799.
(RTOG) trial 85-31 evaluating the potential benefit of 68. Nag S, Bice W, DeWyngaert K, et al: The American
androgen suppression following standard radiation Brachytherapy Society recommendations for permanent
therapy for unfavorable prognosis carcinoma of the prostate brachytherapy postimplant dosimetric analysis.
prostate. Int J Radiat Oncol Biol Phys 2001;49 Int J Radiat Oncol Biol Phys 2000;46(1; 1-1-2000):
(4; 3-15-2001):937–946. 221–230.
56. Pilepich MV, Winter K, John MJ, et al: Phase III 69. Lee WR, Roach M III, Michalski J, et al: Interobserver
Radiation Therapy Oncology Group (RTOG) trial variability leads to significant differences in quantifiers of
86-10 of androgen deprivation adjuvant to definitive prostate implant adequacy. Int J Radiat Oncol Biol Phys
radiotherapy in locally advanced carcinoma of the 2002;54(2; 10-1-2002):457–461.
prostate. Int J Radiat Oncol Biol Phys 2001;50 70. Potters L, Cao Y, Calugaru E, et al: A comprehensive
(5; 8-1-2001):1243–1252. review of CT-based dosimetry parameters and
57. Aronowitz JN: Dawn of prostate brachytherapy: biochemical control in patients treated with permanent
1915–1930. Int J Radiat Oncol Biol Phys 2002;54 prostate brachytherapy. Int J Radiat Oncol Biol Phys
(3; 11-1-2002):712–718. 2001;50(3; 7-1-2001):605–614.
58. Aronowitz JN: Benjamin Barringer: originator of the 71. Stock RG, Stone NN, Tabert A, et al: A dose-response
transperineal prostate implant. Urology 2002; study for I-125 prostate implants. Int J Radiat Oncol
60(4):731–734. Biol Phys 1998;41(1; 4-1-1998):101–108.
59. Eastham JA, Kattan MW, Groshen S, et al: Fifteen-year 72. Mate TP, Gottesman JE, Hatton J, et al: High dose-rate
survival and recurrence rates after radiotherapy for afterloading 192iridium prostate brachytherapy:
localized prostate cancer. J Clin Oncol 1997; feasibility report. Int J Radiat Oncol Biol Phys
15(10):3214–3222. 1998;41(3; 6-1-1998):525–533.
60. Zelefsky MJ, Whitmore WF Jr: Long-term results of 73. Martinez AA, Kestin LL, Stromberg JS, et al: Interim
retropubic permanent 125iodine implantation of the report of image-guided conformal high-dose-rate
prostate for clinically localized prostatic cancer. J Urol brachytherapy for patients with unfavorable prostate
1997;158(1):23–29. cancer: the William Beaumont phase II dose-escalating
61. Holm HH, Juul N, Pedersen JF, et al: Transperineal trial. Int J Radiat Oncol Biol Phys 2000;47(2; 5-1-2000):
125iodine seed implantation in prostatic cancer guided 343–352.
by transrectal ultrasonography. J Urol 1983; 74. Critz FA: A standard definition of disease freedom is
130(2):283–286. needed for prostate cancer: undetectable prostate specific
62. Blasko JC, Ragde H, Grimm, PD, et al: Prostate antigen compared with the American Society of
brachytherapy: importance of technique. J Clin Oncol Therapeutic Radiology and Oncology Consensus
1996;14(6):1965–1967. Definition. J Urol 2002;167(3):1310–1313.
63. Martinez A, Benson RC, Edmundson GK, et al: Pelvic 75. Cavanagh W, Blasko JC, Grimm PD, et al:
lymphadenectomy combined with transperineal Transient elevation of serum prostate-specific antigen
interstitial implantation of iridium-192 and external following (125)I/(103)Pd brachytherapy for
beam radiotherapy for locally advanced prostatic localized prostate cancer. Semin Urol Oncol
carcinoma: technical description. Int J Radiat Oncol Biol 2000;18(2):160–165.
Phys 1985;11(4):841–847. 76. Critz FA, Williams WH, Benton JB, et al: Prostate
64. Puthawala AA, Syed AM, Austin PA, et al: Long-term specific antigen bounce after radioactive seed
results of treatment for prostate carcinoma by staging implantation followed by external beam radiation for
pelvic lymph node dissection and definitive irradiation prostate cancer. J Urol 2000;163(4):1085–1089.
using low-dose rate temporary iridium-192 interstitial 77. Stock RG, Stone NN, Cesaretti JA: Prostate-specific
implant and external beam radiotherapy. Cancer antigen bounce after prostate seed implantation for
2001;92(8; 10-15-2001):2084–2094. localized prostate cancer: descriptions and implications.
65. Nag S, Ciezki JP, Cormack R, et al: Intraoperative Int J Radiat Oncol Biol Phys 2003;56(2; 6-1-2003):
planning and evaluation of permanent prostate 448–453.
brachytherapy: report of the American Brachytherapy 78. Blasko JC, Grimm PD, Sylsvester JE, et al: The role of
Society. Int J Radiat Oncol Biol Phys 2001;51 external beam radiotherapy with I-125/Pd-103
(5; 12-1-2001):1422–1430. brachytherapy for prostate carcinoma. Radiother Oncol
66. D’amico AV, Cormack R, Tempany CM, et al: Real-time 2000;57(3):273–278.
magnetic resonance image-guided interstitial 79. Critz FA, Williams WH, Levinson AK, et al:
brachytherapy in the treatment of select patients with Simultaneous irradiation for prostate cancer:
clinically localized prostate cancer. Int J Radiat Oncol intermediate results with modern techniques. J Urol
Biol Phys 1998;42(3; 10-1-1998):507–515. 2000;164(3 Pt 1):738–741.
Chapter 26B Clinically Localized Adenocarcinoma of the Prostate: Radiation Therapy 493

80. Brandeis J, Pashos CL, Henning JM, et al: A nationwide adverse sexual health-related quality-of-life outcome
charge comparison of the principal treatments for early after prostate brachytherapy. Urology 2002;
stage prostate carcinoma. Cancer 2000;89(8; 10-15-2000): 59(4):480–484.
1792–1799. 94. Merrick GS, Butler WM, Galbreath RW, et al:
81. Brandeis JM, Litwin MS, Burnison CM, et al: Quality of Five-year biochemical outcome following permanent
life outcomes after brachytherapy for early stage prostate interstitial brachytherapy for clinical T1–T3 prostate
cancer. J Urol 2000;163(3):851–857. cancer. Int J Radiat Oncol Biol Phys 2001;51(1; 9-1-2001):
82. Brachman DG, Thomas T, Hilbe J, et al: Failure-free 41–84.
survival following brachytherapy alone or external beam 95. Stone NN, Stock RG: Prostate brachytherapy: treatment
irradiation alone for T1-2 prostate tumors in 2222 strategies. J Urol 1999;162(2):421–426.
patients: results from a single practice. Int J Radiat 96. Potters L, Torre T, Ashley R, et al: Examining the role
Oncol Biol Phys 2000;48(1; 8-1-2000):111–117. of neoadjuvant androgen deprivation in patients
83. Grado GL, Larson TR, Balch CS, et al: Actuarial undergoing prostate brachytherapy. J Clin Oncol
disease-free survival after prostate cancer brachytherapy 2000;18(6):1187–1192.
using interactive techniques with biplane ultrasound and 97. Borghede G, Hedelin H, Holmang S, et al: Irradiation of
fluoroscopic guidance. Int J Radiat Oncol Biol Phys localized prostatic carcinoma with a combination of high
1998;42(2; 9-1-1998):289–298. dose rate iridium-192 brachytherapy and external beam
84. Grimm PD, Blasko JC, Sylvester JE, et al: 10-year radiotherapy with three target definitions and dose levels
biochemical (prostate-specific antigen) control of inside the prostate gland. Radiother Oncol 1997;
prostate cancer with (125)I brachytherapy. Int J Radiat 44(3):245–250.
Oncol Biol Phys 2001;51(1; 9-1-2001):31–40. 98. Dinges S, Deger S, Koswig S, et al: High-dose rate
85. Ragde H, Korb LJ, Elgamal AA, et al: Modern prostate interstitial with external beam irradiation for localized
brachytherapy. prostate specific antigen results in 219 prostate cancer—results of a prospective trial. Radiother
patients with up to 12 years of observed follow-up. Oncol 1998;48(2):197–202.
Cancer 2000;89(1; 7-1-2000):135–341. 99. Kovacs G, Galalae R: Fractionated perineal high-
86. Stock RG, Stone NN, DeWyngaert, et al: Prostate dose-rate temporary brachytherapy combined with
specific antigen findings and biopsy results following external beam radiation in the treatment of localized
interactive ultrasound guided transperineal prostate cancer: Is lymph node sampling necessary?
brachytherapy for early stage prostate carcinoma. Cancer Cancer Radiother 2003;7(2):100–106.
1996;77(11; 6-1-1996):2386–2392. 100. Martinez AA, Gustafson G, Gonzalez J, et al: Dose
87. Grimm PD, Blasko JC, Sylvester JE, et al: 10-year escalation using conformal high-dose-rate
biochemical (prostate-specific antigen) control of brachytherapy improves outcome in unfavorable
prostate cancer with (125)I brachytherapy. Int J Radiat prostate cancer. Int J Radiat Oncol Biol Phys 2002;53(2;
Oncol Biol Phys 2001;51(1; 9-1-2001):31–40. 6-1-2002):316–327.
88. Dattoli M, Wallner K, True L, et al: Long-term 101. Martinez AA, Pataki I, Edmundson G, et al: Phase II
outcomes after treatment with external beam radiation prospective study of the use of conformal high-dose-rate
therapy and palladium 103 for patients with higher risk brachytherapy as monotherapy for the treatment of
prostate carcinoma: influence of prostatic acid favorable stage prostate cancer: a feasibility report. Int J
phosphatase. Cancer 2003;97(4; 2-15-2003):979–983. Radiat Oncol Biol Phys 2001;49(1; 1-1-2001):61–69.
89. Lederman GS, Cavanagh W, Albert PS, et al: 102. Yoshioka Y, Nose T, Yoshida K, et al: High-dose-rate
Retrospective stratification of a consecutive cohort of interstitial brachytherapy as a monotherapy for localized
prostate cancer patients treated with a combined prostate cancer: treatment description and preliminary
regimen of external-beam radiotherapy and results of a phase I/II clinical trial. Int J Radiat Oncol
brachytherapy. Int J Radiat Oncol Biol Phys 2001;49(5; Biol Phys 2000;48(3; 10-1-2000):675–681.
4-1-2001):1297–1303. 103. Russell KJ, Caplan RJ, Laramore GE, et al: Photon
90. Lee WR: The role of androgen deprivation therapy versus fast neutron external beam radiotherapy in the
combined with prostate brachytherapy. Urology treatment of locally advanced prostate cancer: results of a
2002;60(3, Suppl 1):39–44. randomized prospective trial. Int J Radiat Oncol Biol
91. Gleave ME, Goldenberg SL, Chin JL, et al: Randomized Phys 1994;28(1; 1-1-1994):47–54.
comparative study of 3 versus 8-month neoadjuvant 104. Lindsley KL, Cho P, Stelzer KJ, Koh WJ, Austin-
hormonal therapy before radical prostatectomy: Seymour M, Russell KJ, Laramore GE, Griffin TW:
biochemical and pathological effects. J Urol Fast neutrons in prostatic adenocarcinomas: worldwide
2001;166(2):500–506. clinical experience. Recent Results Cancer Res.
92. Crook J, McLean M, Catton C, et al: Factors influencing 1998;150:125–136.
risk of acute urinary retention after TRUS-guided 105. Slater JD, Rossi CJ Jr, Yonemoto LT, et al: Conformal
permanent prostate seed implantation. Int J Radiat proton therapy for early-stage prostate cancer. Urology
Oncol Biol Phys 2002;52(2; 2-1-2002):453–460. 1999;53(5):978–984.
93. Hollenbeck BK, Dunn RL, Wei JT, et al: Neoadjuvant
hormonal therapy and older age are associated with
C H A P T E R

27 Regionally Advanced Adenocarcinoma


of the Prostate (T3-4N+M0):
Management and Prognosis
Mary Frances McAleer, MD, PhD,
and Richard K. Valicenti, MD

Adenocarcinoma of the prostate is considered to be apy in conjunction with external beam radiation therapy
regionally advanced once malignant cells are identified (EBRT) to enhance local control and disease-free sur-
outside the prostatic capsule (T3a), have invaded the vival, and as adjuvant systemic treatment for manage-
seminal vesicle(s) (T3b), or involve adjacent bladder, rec- ment of occult metastatic disease.9,10
tum, musculature, or pelvic wall (T4). Also, included in
this category are tumors with nodal metastases (N1-3).1
ANDROGEN DEPRIVATION THERAPY
While the overall incidence of prostate cancer in the U.S.
Rationale
rose with the widespread institution of prostate-specific
antigen (PSA) screening in the early 1990s, the percent- Androgens have been shown to stimulate the growth and
age of patients presenting with regionally advanced dis- proliferation of neoplastic prostate cells both in vitro and
ease has decreased.2,3 Using pretreatment PSA level, in vivo by interacting with androgen receptors on these
Gleason score and clinical stage, Partin et al.4 were the cells.11–14 Inhibition of this interaction by hormone ther-
first to demonstrate that the risk of regional spread of apy has been shown to reduce gross tumor volume sig-
prostate cancer can be estimated. Although the actual nificantly.15 This theoretically leads to increased tumor
diagnosis of regionally advanced prostate cancer requires blood flow and decreased tumor hypoxia, which, in turn,
biopsy or pathology demonstrating adenocarcinoma with may allow for improved treatment outcomes with locore-
extra-prostatic spread, multimodality staging (combined gional treatment. Androgen deprivation may additionally
use of tumor stage, pretreatment PSA, and Gleason improve therapeutic results by eradicating distant
score) is an important and useful method to identify men micrometastatic disease not controlled by surgery or
at high risk of subclinical regionally advanced disease.5 radiation therapy alone. The mechanism of these effects
is thought to involve the induction of apoptosis in the
targeted prostate cells, which may have an additive or
GENERAL MANAGEMENT OF REGIONALLY
even synergistic effect when combined with the DNA-
ADVANCED PROSTATE CANCER
damaging action of radiation.16,17
Locoregional therapy for T3 or greater adenocarcinoma
of the prostate, including radical prostatectomy (RP)
Methods of Androgen Deprivation
(with or without lymphadenectomy) and radiation ther-
Orchiectomy
apy alone, have not resulted in improved disease-specific
or overall survival (OS) in these patients, with the major- The most commonly used method of androgen depriva-
ity demonstrating clinically overt metastases by 10 tion worldwide is orchiectomy. Surgical castration has
years.6–8 Given these poor outcomes with monotherapy, been shown to remove 95% of circulating testosterone,
the recent NCCN guidelines and patterns of care with long-lasting reduction of serum levels of this hor-
prostate cancer decision tree advocate the use of andro- mone. Orchiectomy has both economic and compliance
gen deprivation both as neoadjuvant cytoreductive ther- advantages over the other methods of androgen deprivation

494
Chapter 27 Regionally Advanced Adenocarcinoma of the Prostate (T3-TN + M0) 495

discussed later.18 The psychologic ramifications of this


Antiandrogens
approach, however, have limited its widespread use in the
U.S. Antiandrogens function as competitive inhibitors of
testosterone by binding to the androgen receptors on
prostate cells. The two classes of antiandrogens include
Estrogen Supplementation
steroidal and nonsteroidal agents. The former include
Estrogens are thought to act either indirectly by suppress- aminoglutethimide, cyproterone acetate (Androcur), and
ing pituitary gonadotropin, which, in turn, inhibits testic- megestrol, which additionally suppress testosterone by
ular testosterone synthesis, or by directly binding to hypothalamic and pituitary negative feedback mecha-
prostate cell hormone receptors and thereby competitively nisms. The nonsteroidal antiandrogens, namely, flu-
blocking receptor binding by androgens. Use of the syn- tamide, bicalutamide (Casodex) and nilutamide, block
thetic estrogen diethylstilbestrol (DES) by the Veterans androgen uptake and function in prostate cells primarily
Administration Cooperative Urologic Research Group at the level of the androgen receptor. In contrast to the
(VACURG) was the first exogenous hormone therapy steroidal antiandrogens, the nonsteroidal agents do not
given for prostate cancer. DES at doses of 5 mg/day decrease gonadotropin or testosterone levels, and thus
showed similar survival to orchiectomy and improved can- have been shown to maintain libido and potency. Of the
cer-specific survival; however, cardiovascular mortality was nonsteroidal antiandrogens, bicalutamide has higher
increased with DES treatment.19 Reducing the DES dose binding affinity and longer area under the curve (half-life
to 1 mg/day resulted in equivalent survival as with the of 7 to 10 days) with a dose of 50 mg daily being equiva-
higher dose, but castrate levels of testosterone were not lent to 750 mg/day (250 mg every 8 hours) of flutamide
achieved.19,20 Side effects of estrogenic therapy, such as (half-life of 5 to 6 hours).28 Diarrhea and, less frequently,
fluid retention, gynecomastia, and decreased libido, have but more serious hepatic toxicity are side effects of flu-
resulted in its infrequent use in clinical practice. tamide therapy.29 Bicalutamide is associated with more
breast tenderness and gynecomastia than castration.30
Preliminary results of two multi-institutional random-
Progesterone Supplementation
ized trials comparing bicalutamide monotherapy (150 mg
Medroxyprogesterone (Provera) and megestrol (Megace), daily) to surgical or medical (goserelin 3.6 mg every 28
two progestins, also suppress gonadotropin release and days) castration in patients with nonmetastatic prostate
thus interfere with testosterone production.21 These agents cancer demonstrated equivalent survival with median fol-
have not been found to inhibit serum androgen levels com- low-up of over 200 weeks.30 Although longer follow-up is
pletely or for any prolonged period of time, however, and needed, bicalutamide monotherapy may provide a quality
therefore they are not used as monotherapeutic agents. of life advantage over castration with the preservation of
sexual function in patients treated with the antiandrogen.
Luteinizing Hormone Releasing Hormone Analogs
ANDROGEN DEPRIVATION AND RADIATION
Luteinizing hormone releasing hormone (LHRH)
THERAPY
analogs, such as goserelin (Zoladex) and leuprolide
(Lupron), function by initially inducing luteinizing hor- Multivariate analysis of data from a prospective trial of
mone (LH) and follicle stimulating hormone (FSH) the Radiation Therapy Oncology Group (RTOG study
release by the pituitary followed by a gradual inhibition 75-06) investigating pretreatment factors associated with
(after 1 to 2 weeks) of these hormones. This activity disease outcome revealed that hormonal therapy either
results in an initial increase (1 to 2 days) then decrease of prior to or during radiation therapy for prostate carci-
testosterone to castrate levels (by 20 to 28 days).22 noma correlated with improved local tumor control.31 As
LHRH agonists have been shown in randomized trials to a result, androgen deprivation has been studied prospec-
be as efficacious as orchiectomy or DES in the treatment tively in conjunction with radiation therapy in two con-
of advanced prostate cancer.23,24 The initial transient ele- texts: (1) neoadjuvantly as cytoreductive therapy in
vation of testosterone associated with LHRH agonist use patients with bulky primary tumors; (2) adjuvantly in
has resulted in exacerbation of pain in approximately patients at increased risk for micrometastatic disease.
10% of patients with metastatic disease and is thought to Each of these approaches is presented in detail later.
be due to a flare reaction of tumor cell proliferation.25,26
To avoid this flare reaction, antiandrogens (see later) are
Radiation Therapy with Neoadjuvant Hormonal
frequently coadministered either temporarily or continu-
Therapy
ously with the LHRH analogues.27 The principle side
effects of LHRH agonists are impotence, decreased Based on the promising results of RTOG 75-06 noted
libido, testicular atrophy, and hot flashes. earlier, as well as on the observation of >94% primary
496 Part V Prostate Gland and Seminal Vesicles

tumor clearance in both a phase II randomized study neoadjuvant or prolonged hormonal therapy in these
examining the efficacy and toxicity of megestrol and DES patients.
with definitive EBRT for locally advanced prostate can-
cer (RTOG 83-07), and a similar phase II trial (RTOG
Radiation Therapy with Adjuvant Hormonal Therapy
85-19) investigating flutamide and goserelin cytoreduc-
tion with EBRT the RTOG conducted a phase III trial The use of androgen deprivation as adjuvant systemic
(86-10) comparing EBRT with and without hormonal therapy with EBRT in patients with regionally advanced
therapy given prior to and during the radiation treat- adenocarcinoma of the prostate has been studied
ment.31–34 The patients enrolled in RTOG 86-10 had prospectively by several groups. One of the earliest
bulky T26-T4 prostate cancers, with 91% being lymph reports of adjuvant hormonal therapy and radiation ther-
node negative.35 The androgen deprivation method used apy in patients with T3-4NxM0 prostate cancer was a 15-
in this phase III trial consisted of two months of goserelin year follow-up by Zagars et al.37 Patients in this study
(3.6 mg subcutaneously every 28 days) and flutamide (250 were randomized either to EBRT alone or to EBRT plus
mg orally 3 times daily) prior to EBRT (45 to 50 Gy DES until relapse or death. Significant improvement in
pelvic RT plus 20 to 25 Gy prostate boost) with contin- actuarial disease-free survival was seen in patients receiv-
ued hormonal therapy during the radiation treatment. ing the DES with EBRT (63%) in contrast to those
There were 456 patients who could be evaluated in treated with EBRT alone (35%). No difference in clini-
RTOG 86-10, with 226 patients in the androgen depri- cal local tumor control or OS was detected, however, in
vation arm and 230 patients in the EBRT-only arm. The part because of DES use in relapsed patients receiving
recently reported long-term update of this study (median EBRT-only, and also because of increased intercurrent
6.7-year follow-up) revealed that the 8-year local control mortality in patients initially receiving DES with EBRT.
rate was significantly higher in the patients treated with A phase III trial by the RTOG (study 85-31) examined
neoadjuvant hormonal therapy (NHT) and EBRT (42%) the effect of EBRT (44 to 46 Gy to pelvis with 20 to 25
than in the EBRT-only group (30%, p = 0.016).35 The Gy prostate boost or 60 to 65 Gy to prostatic bed if post-
incidence of distant metastases was also significantly prostatectomy) either with or without adjuvant goserelin
lower in the patients with androgen deprivation (34% (3.6 mg subcutaneously every 28 days) in patients with
versus 45%, p = 0.04). The actuarial and biochemical dis- locally advanced prostate cancer. Goserelin was adminis-
ease-free survival rates were also significantly improved tered to 477 patients during the last week of EBRT or at
in these patients (33% versus 21%, p = 0.004; and 24% relapse to 468 patients initially treated with EBRT-only.
versus 10%, p < 0.0001, respectively). While there was no Statistically significant reduction in rates of local failure
difference in OS of all patients in the two groups, a sur- and distant metastases, and improvement in biologic and
vival benefit was observed in patients with Gleason score clinical response rates were observed in patients receiving
2 to 6 receiving the hormone treatment (70% versus goserelin adjuvantly versus at relapse, both were at 5- and
52%, p = 0.015). 8-year follow-up.38,39 No difference in OS was detected
The treatment delivered in both arms of RTOG 86-10 in the initial versus relapse hormonal therapy groups at
was relatively well tolerated, the highest frequency of 5 years (75% versus 71%) or at 8 years (49% versus
treatment termination being attributed to flutamide toxic- 47%). Subset analysis of patients with Gleason score 8 to
ity (16%). The main criticism of this study is that initial 10 (without prostatectomy) did demonstrate significant
staging was based on digital rectal examination (DRE) and benefit of adjuvant androgen deprivation with respect to
not PSA or biopsy results. Nevertheless, this study set the both OS and cause-specific failure compared with hor-
standard for future trials and clinical application of combi- monal therapy at relapse.39
nation therapy for regionally advanced prostate cancer. A similar phase III study was conducted by the
One such prospective randomized trial from Canada European Organization for Research on Treatment of
investigated 64.8 Gy EBRT alone (n = 41), versus flu- Cancer (EORTC) that investigated EBRT (≤50 Gy to
tamide and LHRH agonist therapy delivered either 3 pelvis with 20 Gy prostate/seminal vesicle boost) with or
months prior to EBRT (n = 43) or 3 months prior to, without hormonal therapy (cyproterone 150 mg orally
during, and 6 months following EBRT (n = 36) for 3 times daily for 4 weeks starting 1 week prior to EBRT
patients with T2a to T4 adenocarcinoma of the and goserelin starting day 1 of EBRT and continuing for
prostate.36 Trans-rectal ultrasound (TRUS)-guided biop- 3 years) in patients with prostate cancer at high risk for
sies performed at 24 months following EBRT showed metastatic disease (high grade T1-2 or any grade T3-4,
65%, 28%, and 5% residual cancer in each group, no nodal disease at/above common iliac chains, M0).
respectively (p = 0.00001). While there is apparent local After median follow-up of 5.5 years, statistically signifi-
benefit of prolonged androgen deprivation versus NHT cant reduction in rates of locoregional failure and distant
with EBRT, no other clinical outcome measures were metastases, and improvement in biologic and clinical
reported to support a survival advantage with the use of response rates were again observed in the group receiving
Chapter 27 Regionally Advanced Adenocarcinoma of the Prostate (T3-TN + M0) 497

adjuvant hormonal therapy as reported in RTOG 86- mined by TRUS and DRE, not by PSA or biopsy. Third,
10.40,41 In contradistinction to the RTOG 85-31 results, the 5-year survival results for the EBRT-only arm is
however, the use of adjuvant hormonal therapy in the lower than expected for this group (compare with RTOG
EORTC study was associated with significantly 85-31 results). Finally, the 5-year follow-up period is
improved disease-specific survival (94% with hormonal considered short for prostate cancer trials.
therapy versus 79% without hormonal therapy, p = The duration of androgen deprivation required for
0.0001) and OS (78% with hormonal therapy versus 62% therapeutic benefit in patients with regionally advanced
without hormonal therapy, p = 0.0002) (Figure 27-1). prostate cancer is not yet established. The effect of
While the EORTC study is of great importance in 2-year maintenance adjuvant goserelin therapy versus
justifying the use of early adjuvant androgen deprivation observation alone following 4 months of goserelin and
in the treatment of regionally advanced prostate cancer, flutamide therapy (2 months prior to and 2 months dur-
this study is not without its criticisms. First, patients were ing EBRT) in patients with T2b-4 disease was investi-
stratified by clinical stage and histologic grade and not by gated in RTOG study 92-02. Treatment response at
PSA or biopsy findings. Second, local control was deter- 4.8-year follow-up reflects the findings of RTOG 85-31,

100 79%
90 (72−86%)
Percentage of patients

80
70 Combined
60 treatment
50 62%
40 (52–72%)
Radiation
30
therapy
20 p = 0.001 (overall log-rank test)
10
0
0 1 2 3 4 5 6 7 8 9 10
Years
No.
No. patients at risk who died
Radiotherapy 208 183 139 96 67 39 23 10 6 1 58
Combined 207 190 144 111 82 55 39 19 7 0 35
treatment
A
85%
100
(78–92%)
90
Combined
Percentage of patients

80
treatment
70
60
50
48% Radiotherapy
40
30 (38–58%)
20 p < 0.001 (overall log-rank test)
10
0
0 1 2 3 4 5 6 7 8 9 10
Years
No. with
disease
No. patients at risk progression
Radiotherapy 208 163 107 59 38 19 11 5 3 1 78
Combined 207 189 138 108 78 51 36 16 5 0 20
treatment
B
Figure 27-1 A, Kaplan-Meier estimate of OS. B, Disease-free survivals in EORTC study.
The number of patients at risk for the event at each time point is the total number of
patients less the number of patients in whom disease progressed or who were lost to
follow-up.40
498 Part V Prostate Gland and Seminal Vesicles

with significantly decreased rates of local progression, and adjuvant hormonal therapy in conjunction with
distant metastases, and biochemical failure in patients EBRT, OS time was increased only in patients or subsets
receiving long-term androgen suppressive therapy com- of patients receiving androgen deprivation early and for
pared with short-term suppression. No OS benefit was prolonged duration. One phase III trial conducted by the
detected (79% long-term suppression versus 77% short- RTOG (94-13) was designed to compare NHT (andro-
term suppression, p > 0.05). Subset analysis did demon- gen deprivation using goserelin or leuprolide with flu-
strate both disease-specific survival (90% versus 78%, p = tamide 2 months before and during EBRT, arms 1 and 2)
0.007) and OS (80% versus 69%, p = 0.02) advantage directly with adjuvant therapy (androgen deprivation
with the longer duration hormonal therapy in patients 4 months after EBRT, arms 3 and 4) to patients with
with Gleason scores 8 to 10.42 The results of this study high-risk localized prostate cancer irradiated either 70.2
support the continued use and study of long-term andro- Gy to the prostate only (arms 2 and 4) or 50.4 Gy to the
gen deprivation in patients with poorly differentiated or whole pelvis with a 19.8 Gy prostate boost (arms 1 and
locally advanced prostate cancer. 3). Roach et al.45 reported initial results in 1323 patients
Further support of long-duration hormonal therapy in after median follow-up of 59.3 months. Significant pro-
patients at high risk for subclinical metastatic prostate can- gression-free survival (PFS) at 4 years was observed in
cer comes from a Swedish prospective randomized trial that patients receiving NHT and whole pelvis irradiation ver-
investigated the effect of orchiectomy in patients receiving sus NHT and prostate-only irradiation (59.6% versus
EBRT (versus EBRT-only) for pelvis-confined T1-4, N0-3, 44.3%, p = 0.001). Four-year PFS in patients receiving
M0 disease.43 This trial, originally designed to accrue 400 adjuvant hormonal therapy with either whole pelvis or
patients, was terminated early after enrolling only 91 prostate-only RT was comparable to that of patients
patients because of a high frequency of disease progression given NHT and prostate-only RT. While OS was similar
in the patients receiving EBRT alone. Among patients with in all 4 treatment arms (88%, 83%, 81%, and 82%,
nodal positive disease, a significant reduction in disease pro- respectively), there is a trend for improved OS in the
gression (61% versus 31%) and overall mortality (61% ver- NHT/whole pelvis RT group. Longer duration follow-
sus 38%) was observed after a median follow-up time of 9.3 up is needed, however, since the median survival in these
years. While OS was improved in node negative patients patients is not likely to be reached for several years.
treated with EBRT alone than in those with lymph node There are several other phase III trials currently
metastases, no difference was seen in either group treated ongoing designed to address the issue of timing and
with EBRT and orchiectomy, suggesting a benefit of early duration of androgen deprivation in patients with locore-
hormonal therapy in locally advanced prostate cancer. gionally advanced prostate cancer. These are discussed in
The issue of early versus delayed hormonal therapy in the section “Future Directions.”
patients with T2-4 or asymptomatic M+ prostate adeno-
carcinoma was addressed by the Medical Research Council
ANDROGEN DEPRIVATION
(MRC) Prostate Cancer Working Party Investigation
AND PROSTATECTOMY
Group.44 In this trial, 934 patients with expected survival
of ≥12 months were randomized at the physicians’ discre- RP is typically reserved for patients with clinically organ-
tion to receive hormonal therapy (orchiectomy or LHRH confined disease (T1-2, N0, M0); pelvic lymph node dis-
agonist with flare protection) either early (within 6 weeks section being recommended for those with Gleason score
of presentation) or deferred until disease progression. 5 to 6 and PSA ≥ 20 ng/ml, Gleason score ≥ 7 and PSA ≥
There was a statistically significant reduction in disease- 15 ng/ml, or clinical T3N0M0.9 Clinical assessment of
specific mortality in both patients with no known metas- extent of disease in prostate adenocarcinoma has been
tases, as well as with any M status (0, X or +) receiving found to underestimate the actual degree of disease
immediate hormonal therapy (54% versus 70% and 62% spread in approximately 50% of cases.46–49 Additional
versus 71%, respectively) at 10 years follow-up. There was therapy for patients with known or suspected extra-
also an increased risk of major complications including glandular disease includes preoperative hormonal ther-
cord compression, extra-skeletal metastases, or ureteral apy or postoperative hormonal therapy with or without
obstruction in the delayed hormone group. The OS of radiation treatment.
patients in both treatment groups was lower than that
observed in other studies (26% at 10 years).
Prostatectomy and Neoadjuvant
Hormonal Therapy
Timing and Duration of Hormonal Therapy
Preoperative androgen deprivation has been investigated
with Radiation
in several prospective randomized trials to test the
Although the studies outlined earlier demonstrate hypothesis that tumors initially considered nonoperable
improved local and distant control with both neoadjuvant (clinical T3, Nx, or greater) could be rendered resectable
Chapter 27 Regionally Advanced Adenocarcinoma of the Prostate (T3-TN + M0) 499

after 3 months of hormonal treatment.50–52 The rate of Secondary analysis of RTOG 85-31 examined the
positive surgical margins in these studies was significantly effect of immediate versus delayed goserelin therapy in
reduced by 40% to 60% in patients receiving the hor- postprostatectomy patients found to have extra-capsular
monal therapy compared with those treated by surgery extension to the resection margin and/or seminal vesi-
alone. This effect on margin status, however, has not cle invasion before initiation of EBRT.55 In contrast
translated into any therapeutic improvement, there being with Messing’s results, no difference was detected in
no difference in biochemical or clinical recurrence rates rates of local progression, distant relapse, or OS in the
in both treatment groups. This lack of therapeutic bene- two groups, with only prolonged freedom from bio-
fit has been attributed, in part, to insufficient follow-up, chemical relapse being noted in patients receiving
underpowered study design, or inadequate duration of immediate hormonal therapy at 5-year follow-up.
hormone therapy. Duration of androgen deprivation, as well as the addi-
To address the issue of optimal length of androgen tion of EBRT, may account for the survival difference
deprivation required before RP, the Canadian Urologic noted in these two trials.
Oncology Group designed a phase III trial comparing
3-month versus 8-month preoperative treatment with TOXICITY
leuprolide and flutamide in patients with T1b-T2 adeno-
carcinoma of the prostate. Gleave et al.53 recently pre- Each of the various treatment modalities for regionally
sented the 3-year follow-up results of this study. advanced adenocarcinoma of the prostate has associated
Prolonged androgen deprivation in these patients was adverse side effects, which need to be taken into account
associated with significant reduction in preoperative PSA when managing patients with this disease. Both acute and
levels (75.1% versus 43.3% with PSA < 0.1 ng/ml, late complications associated with the use of EBRT for
p < 0.0001), positive surgical margins (12% versus 23%, locoregionally advanced prostate cancer have been
p = 0.0106), and incidence of lymph node metastasis described; the former includes increased urinary fre-
(0.4% versus 3.1%, p = 0.038). The longer duration of quency, dysuria, diarrhea, and rectal urgency; and the
hormone therapy was also associated with significantly latter includes urethral stricture, hematuria, urinary
more patients clinically downsized based on DRE or and fecal incontinence, rectal bleeding, and impo-
TRUS evaluation. Prolonged androgen deprivation was tence.56–59 While the incidence of bowel and bladder
associated with more toxicity, 87% of patients treated for complications following EBRT has been reported at or
8 months reporting hot flashes versus 72% of those below 10%, impotence is at least twice this rate by 2
treated for 3 months (p < 0.0001). Longer follow-up is years following treatment, with additional men reporting
required to assess the effect of duration of preoperative loss of potency at longer follow-up intervals. The occur-
hormonal therapy on clinical and biochemical outcomes rence of adverse side effects from radiation for prostate
in these patients. adenocarcinoma is not unexpectedly correlated to
total dose of radiation delivered.60,61 New technologic
advances in the delivery of radiation to the prostate, such
Prostatectomy and Adjuvant Hormonal Therapy as intensity-modulated radiation therapy, are designed to
The role of adjuvant hormonal therapy in patients found reduce toxicity to adjacent normal structures. The
to have positive nodal metastasis after RP with pelvic reports on the effectiveness of this type of targeted ther-
lymphadenectomy for T1-2 prostate cancer was prospec- apy on tumor control and side effect profile are eagerly
tively investigated by Messing et al.54 In this study, anticipated.
patients were randomized to immediate hormonal ther- The side effects associated with the various hormonal
apy (goserelin or orchiectomy) versus observation until therapies for prostate adenocarcinoma have previously
disease progression. After a median follow-up of >7 been delineated in the section “Androgen Deprivation
years, the early use of androgen deprivation was associ- Therapy” and are summarized in Table 27-1. The effect
ated with significant improvement in PFS (77% versus of combination androgen deprivation with EBRT on
18%, p < 0.001), cause-specific survival (92% versus 69%, adverse treatment sequelae has recently become the focus
p < 0.01), as well as OS (85% versus 65%, p = 0.02), com- of several reports.
pared with patients receiving delayed hormonal therapy. Schultheiss et al.60 observed an increased risk of late
The incidence of side effects, most commonly hot gastrointestinal and genitourinary toxicity in patients
flashes, gastrointestinal disturbances, and gynecomastia, treated with NHT as opposed to those receiving EBRT
were reported in up to 20% to 60% of patients receiving alone. A study from the Italian National Institute for
the adjuvant therapy. One criticism of this trial is that Cancer Research also identified adjuvant androgen dep-
hormonal therapy alone (without RP) could have pro- rivation as a significant independent predictor of late rec-
duced similar results but no study would be conducted tal toxicity at 2 years in patients treated with
today to test such a hypothesis. three-dimensional conformal radiation therapy.62 This
500 Part V Prostate Gland and Seminal Vesicles

Table 27-1 Mechanism of Action and Major Side Effects of Hormonal Agents
Agent LHRH LH FSH Testosterone Side Effect(s)

Estrogens — ↓ — — Gynecomastia, thrombosis

LHRH agonist (short-term use) ↑ ↑ ↑ ↑ Flare reaction

LHRH agonist (long-term use) ↑ ↓ — ↓ Hot flashes, loss of libido, osteoporosis,


muscle wasting

LHRH agonist+Antiandrogen ↑ ↑ ↑ Inhibited Hot flashes, no flare, GI side effects,


gynecomastia

Antiandrogen (steroidal) ↑ ↓ — Inhibited Hot flashes, GI side effects, gynecomastia

Antiandrogen (nonsteroidal) — — — Inhibited Gynecomastia

GnRH antagonist Inhibited ↓ ↓ ↓ Histamine release

effect appears to be related to duration of hormonal ther- the earlier sections suggests that even with locoregionally
apy, with treatment in excess of 9 months being associ- advanced disease, men can expect to live on average 8
ated with decreased rectal tolerance to radiation.63 years regardless of treatment modality employed (see
The effect of combined modality therapy on sexual Table 27-2). It is estimated that this number will likely
potency was examined in patients enrolled in RTOG 86- increase with the widespread institution of PSA screen-
10. Preservation of sexual potency was similar for both ing both with earlier initial diagnosis and identification of
patients receiving EBRT or EBRT with hormonal ther- relapse/disease progression.
apy, with 81% return of sexual potency for the latter Roach et al.67 examined >1500 men treated with radia-
compared to 74% for the men receiving EBRT-only.35 tion therapy alone on RTOG phase III trials 75-06, 77-06,
This finding was confirmed by the use of validated self- 85-31, and 86-10; and they identified factors associated with
administered questionnaires given at baseline and serially poorer overall and disease-specific survival. These adverse
after treatment, with change in sexual function scores and prognostic factors include high Gleason score, advanced
overall quality of sex life being similar for both groups.64 clinical stage, and positive nodal status, and of these,
However, additional studies suggest that the use of long Gleason score was found to be the single most important
duration androgen deprivation for periods >6 months predictor of death in these patients. In a more recent analy-
may increase the likelihood of decreased libido secondary sis by this group (specifically addressing the impact of race
to failure of testosterone level normalization following on survival), patients treated with EBRT, both with and
combined radiation and hormonal therapy.65 without hormonal therapy, were included, and Gleason
As patients with advanced adenocarcinoma of the score, clinical T stage, and lymph node status again demon-
prostate can be expected to demonstrate continued strated prognostic value with respect to overall and disease-
improvement in OS, with 5-year survival rates currently specific survivals.69 In addition, low pretreatment PSA levels
being reported at ~70% (Table 27-2), the topic of treat- and addition of hormonal therapy were found to be associ-
ment-related toxicity and effect on quality of life will ated with improved outcomes in these patients.
become ever more important areas of investigation and Currently, the use of PSA testing is commonly
targets for improved treatment design. Based on the evi- employed in the follow-up of patients diagnosed with
dence, concerns of adverse side effects should not neces- adenocarcinoma of the prostate. Biologic control and
sarily deter clinicians from recommending to men with relapse based on PSA values have been reported in recent
locoregionally advanced prostate cancer combined short studies.38,39,41,42 To date, however, no clear correlation
duration (4 to 6 months) NHT and EBRT. between biologic failure and decreased survival has been
established (perhaps due to the long natural history of
this disease), and the significance of this biomarker rela-
OUTCOMES AND PROGNOSIS
tive to patient outcome needs to be further elucidated.
Adenocarcinoma of the prostate is characteristically a
slow growing malignancy with high 10-year survival rates
FUTURE DIRECTIONS
being reported for early stage disease.66 Analysis of the
survival data of patients with prostate cancer enrolled in The randomized trials investigating therapeutic options
the prospective, randomized clinical trials described in for patients with locoregionally advanced adenocarcinoma
Chapter 27 Regionally Advanced Adenocarcinoma of the Prostate (T3-TN + M0) 501

Table 27-2 Survival Data from Trials of Hormonal Therapy for Prostate Cancer
5-year 8-year Median Survival
Study References Arm OS (%) OS (%) p Value (year)

RTOG 86-10 35 RT 68 44 NS <8

NHT+RT 72 53 ~8

RTOG 85-31 38–39 RT+early HT 75 49 NS 8

RT+late HT 71 47 8

EORTC 40–41 RT 62 NA 0.0002 NA

RT+HT 78 NA NA

RTOG 92-02 42 NHT+RT 78 NA NS NA

NHT+RT+HT 79 NA NA

Swedish 43 RT ~70 NA NS ~8

RT+orchiect ~80 NA not met

MRC 44 Early HT n.a. 38* NS

Delayed HT n.a. 29* not reported

RTOG 94-13 46 NHT+WPRT 88 NA NS NA

NHT+PRT 83 NA NA

WPRT+HT 81 NA NA

PRT+HT 82 NA NA

Messing 55 RP? HT 85 85† 0.02 NA

RP? observe 70 65† NA

NA, not available; NS, not significant ( p > 0.05); NHT, neoadjuvant hormonal therapy; WPRT, whole pelvis RT; PRT, prostate-only RT.
*10-year OS.
†7-year OS.

of the prostate conducted over the past two decades have There are several prospective randomized studies cur-
not significantly changed the practice of urologists or rently underway designed to address the timing and
radiation oncologists. The reason for this is due in large duration of hormonal therapy (either alone or in con-
part to flawed protocol design (e.g., non-PSA based strat- junction with other treatment modalities), as well as the
ification schemas) and evaluation of treatment modalities benefit of hormonal therapy in low-risk patients versus
(such as perineal prostatectomy or DES use) already out- intermediate-risk patients with prostate cancer. These
dated by the time the trials were reported. The effects of are briefly discussed in the next paragraph.
the various treatments on local symptom control and on The National Cancer Institute of Canada has a phase
quality of life were also frequently overlooked. The goal III trial investigating hormonal therapy (pharmaceutical
of future clinical trials examining the efficacy and pat- or surgical) with or without EBRT for patients with stage
terns of failure in regionally advanced prostate cancer is III/stage IV disease. A phase III study being conducted
to ensure better study design by taking into account by the EORTC (22961) was designed to address the
patient selection, tumor characteristics, prognostic fac- question of duration of androgen blockade (observation
tors and staging methods and by including quality of life versus continued 2.5-year treatment) following initial 6
and cost-benefit issues in addition to survival, locore- months of hormonal therapy with EBRT for patients with
gional control, and distant, clinical or biochemical failure T1c-2b, N1-2 or T2c-4, N0-2 prostate cancer. The length
as study endpoints. of total androgen suppression is also being investigated
502 Part V Prostate Gland and Seminal Vesicles

by the RTOG in a phase III study (99-10), which is ran- 12. deLaumont Y, Veilleux R, Dufour M, et al:
domizing intermediate risk patients to 8 versus 28 weeks Characteristics of the biphasic action of androgens and
of hormonal therapy prior to concurrent androgen dep- of the potent antiproliferative effects of the new
rivation and EBRT. The RTOG is additionally looking at antiestrogen EM-139 on cell cycle kinetic parameters in
LNCaP human prostatic cancer cells. Cancer Res 1991;
the role of radiation therapy with or without androgen
51:5165–5170.
deprivation both in low-risk (T1b-2b, Nx-0, PSA < 20
13. Manni A, Bartholomew M, Caplan R, et al: Androgen
ng/ml) patients (RTOG 94-08) and in postoperative priming and chemotherapy in advanced prostate cancer:
(pT3N0M0 or pT2N0M0 with positive margins) evaluation of determinants of clinical outcome. J Clin
patients (RTOG 96-01 and RTOG 00-11). The results Oncol 1988; 6:1456–1466.
of these and other prospective randomized trials are 14. Fowler JE Jr, Whitmore WF Jr: The response of
expected to provide important information regarding the metastatic adenocarcinoma of the prostate to exogenous
future care and management of patients with regionally testosterone. J Urol 1981; 126:372–375.
advanced adenocarcinoma of the prostate. 15. Pinault S, Tetu B, Gagnon J, et al: Transrectal ultrasound
evaluation of local prostate cancer in patients treated with
LHRH agonist and in combination with flutamide.
REFERENCES Urology 1992; 39:254–261.
16. Zietman AL, Prince E, Nakfoor BM, et al: Androgen
1. American Joint Commission on Cancer Staging Manual, deprivation and radiation therapy: sequencing studies
5th edition. Philadelphia, Lippincott-Raven Publishers, using the Sinology in vivo tumor system. Int J Radiat
1997. Oncol Biol Phys 1997; 38:1067–1071.
2. Stephenson RA, Smart CR, Mineau GP, et al: The fall in 17. Lim JD, Hasegawa M, Sikes C, et al: Supra-additive
incidence of prostate carcinoma. On the downside of a apoptotic response of R3327-G rat prostate tumors to
prostate specific antigen induced peak in incidence—data androgen ablation and radiation. Int J Radiat Oncol Biol
from the Utah Cancer Registry. Cancer 1996; Phys 1997; 38:1071–1078.
77:1342–1348. 18. Bayoumi AM, Brown AD, Garber AM: Cost effectiveness
3. Catalona WJ, Smith DS, Ratliff TL, et al: Detection of of androgen suppression therapies in advanced prostate
organ-confined prostate cancer is increased through cancer. J Natl Cancer Inst 2000; 92:1731–1739.
prostate-specific antigen-based screening. JAMA 1993; 19. Byar DP: The veterans administration cooperative
270:948. urologic research group’s studies of cancer of the prostate.
4. Partin AW, Kattan MW, Subong ENP, et al: Combination Cancer 1993; 32:1126.
of prostate-specific antigen, clinical stage, and Gleason 20. Shearer RJ, Hendry WF, Sommerville IF, et al: Plasma
score to predict pathological stage of localized prostate testosterone: an accurate monitor of hormone treatment
cancer: a multi-institutional update. JAMA 1997; of prostatic cancer. Br J Urol 1973; 45:668.
277:1445. 21. Geller J, Albert J, Yen SSC: Treatment of advanced cancer
5. D’Amico AV: Combined-modality staging for localized of prostate with megestrol acetate. Urology 1978;
adenocarcinoma of the prostate. Oncology 2001; 12:537–541.
15:1049–1075. 22. Faure N, Lemay A, Larouche B, et al: Preliminary results
6. Duncan W, Warde P, Catton CN, et al: Carcinoma of the on the clinical efficacy and safety of androgen inhibition
prostate: results of radical radiotherapy (1970–1985). Int J by an LHRH agonist alone or combined with an
Radiat Oncol Biol Phys 1993; 26:203–210. antiandrogen in the treatment of prostatic carcinoma.
7. Zagars GK, von Eschenbach AC, Ayala AG: Prognostic Prostate 1983; 4:601–624.
factors in prostate cancer: analysis of 874 patients 23. Soloway MS, Chodak G, Vogelzang NJ, et al: Zoladex
treated with radiation therapy. Cancer 1993; versus orchiectomy in treatment of advanced prostate
72:1709–1725. cancer: a randomized trial. Urology 1991; 37:46.
8. Epstein JI, Partin AW, Sauvageot J, et al: Prediction of 24. The Leuprolide Study Group: Leuprolide versus
progression following radical prostatectomy: a diethylstilbestrol for metastatic prostate cancer. N Engl
multivariate analysis of 721 men with long-term follow- J Med 1984; 311:1281.
up. Am J Surg Pathol 1996; 20:286–292. 25. Waxman J, Man A, Hendry WF, et al: Importance of early
9. Millikan R, Logothetis C: Update of the NCCN tumor exacerbation in patients treated long-acting
guidelines for treatment of prostate cancer. Oncology analogues of gonadotrophin releasing hormone for
1997; 11:180–193. advanced prostatic cancer. Br Med J 1985; 291:1387.
10. Coia LR, Hanks GE: Quality assessment in the U.S.A.: 26. Thompson I M, Zeidman EJ, Rodriguez FR: Sudden death
how the patterns of care study has made a difference. due to disease flare with luteinizing hormone-releasing
Semin Radiat Oncol 1997; 7:146–156. hormone agonist therapy for carcinoma of the prostate.
11. Limonta P, Dondi D, Marelli MM, et al: Growth J Urol 1990; 144:1479.
of the androgen-dependent tumor of the prostate: role 27. Labrie F, Dupont A, Belanger A, et al: Flutamide
of androgens and of weakly expressed growth eliminates the risk of disease flare in prostatic cancer
modulatory factors. J Steroid Biochem Mol Biol 1995; patients treated with a luteinizing hormone-releasing
53:401–405. hormone agonist. J Urol 1987; 138:804.
Chapter 27 Regionally Advanced Adenocarcinoma of the Prostate (T3-TN + M0) 503

28. Schellhammer PF, Sharifi R, Block NL: Clinical benefits 40. Bolla M, Gonzalez D, Warde P, et al: Improved survival in
of bicalutamide compared with flutamide in combined patients with locally advanced prostate cancer treated with
androgen blockade for patients with advanced prostatic radiotherapy and goserelin. N Engl J Med 1997;
carcinoma: final report of a double-blind randomized, 337:295–300.
multicenter trial. Urology 1997; 50:330–336. 41. Bolla M, Collette L, Blank L, et al: Long-term results
29. Crownover RL, Holland J, Chen A, et al: Flutamide- with immediate androgen suppression and external
induced liver toxicity including fatal hepatic necrosis. Int irradiation in patients with locally advanced prostate
J Radiat Oncol Biol Phys 1996; 34:911–915. cancer (an EORTC study): a phase III randomized trial.
30. Iversen P, Tyrrell CJ, Kaisary AV, et al: Casodex Lancet 2002; 360:103–106.
(bicalutamide) 150-mg monotherapy compared with 42. Hanks G, Lu J, Machtay M, et al: RTOG protocol 92-02:
castration in patients with previously untreated a phase III trial of the use of long term androgen
nonmetastatic prostate cancer: results from two suppression following neoadjuvant hormonal
multicenter randomized trials at a median follow-up of cytoreduction and radiotherapy in locally advanced
4 years. Urology 1998; 51:389–396. carcinoma of prostate. ASCO Abstr 1284, 2000.
31. Pilepich MV, Krall JM, Sause W T, et al: Prognostic 43. Granfors T, Modig H, Jan-Erik D, et al: Combined
factors in carcinoma of the prostate-analysis of orchiectomy and external radiotherapy versus
RTOG study 75-06. Int J Radiat Oncol Biol Phys 1987; radiotherapy alone for nonmetastatic prostate cancer with
13: 339. or without pelvic lymph node involvement: a prospective
32. Pilepich MV, Krall JM, John MJ, et al: Hormonal randomized study. J Urol 1998; 159: 2030–2034.
cytoreduction in locally advanced carcinoma of the 44. Medical Research Council Prostate Cancer Working
prostate treated with definitive radiotherapy: preliminary Party Investigators Group: Immediate versus deferred
results of RTOG 83-07. Int J Radiat Oncol Biol Phys treatment for advanced prostate cancer: initial results of
1989; 16:813. the Medical Research Council Trial. Br J Urol 1997;
33. Pilepich MV, John MJ, Krall JM, et al: Phase II 79:235–246.
Radiation Therapy Oncology Group study of hormonal 45. Roach M III, DeSilvio M, Lawton C, et al: Neoadjuvant
cytoreduction with flutamide and Zoladex in locally hormonal therapy (NHT) with whole-pelvis (WP)
advanced carcinoma of the prostate treated with definitive radiotherapy (RT) improves progression-free survival
radiotherapy. Am J Clin Oncol 1990; 13:461. (PFS): RTOG (Radiation Therapy Oncology Group)
34. Pilepich MV, Buzydlowski JW, John MJ, et al: Phase II trial 9413, a phase III randomized trial. ASCO Abstr 711,
of hormonal cytoreduction with megestrol and 2002.
diethylstilbestrol in conjunction with radiotherapy for 46. D’Amico AV, Whittington R, Malkowicz SB, et al:
carcinoma of the prostate: outcome results of RTOG A multivariate analysis of clinical and pathological factors
83-07. Int J Radiat Oncol Biol Phys 1995; 32:175–180. that predict for prostate specific antigen failure after
35. Pilepich MV, Winter K, John MJ, et al: Phase III radical prostatectomy for prostate cancer. J Urol 1995;
Radiation Therapy Oncology Group (RTOG) trial 86-10 154:131.
of androgen deprivation adjuvant to definitive 47. Gerber GS, Thisted RA, Scardino PT, et al: Results of
radiotherapy in locally advanced carcinoma of the radical prostatectomy in men with clinically localized
prostate. Int J Radiat Oncol Biol Phys 2001; prostate cancer. JAMA 1996; 276:44.
50:1243–1252. 48. Kattan MW, Stapleton AM, Wheeler TM, et al:
36. Laverdiere J, Gomez JL, Cusan L, et al: Beneficial effect Evaluation of a nomogram used to predict the pathologic
of combination therapy administered prior and following stage of clinically localized prostate carcinoma. Cancer
external beam radiation therapy in localized prostate 1997; 79:528.
cancer. Int J Radiat Oncol Biol Phys 1997; 37:247–252. 49. Partin AW, Kattan MW, Subong EN, et al: Combination
37. Zagars GK, Johnson DE, von Eschenbach AC, et al: of prostate-specific antigen, clinical stage, and Gleason
Adjuvant estrogen following radiation therapy for stage C score to predict pathological stage of localized prostate
adenocarcinoma of the prostate: long-term results of a cancer: a multi-institutional update. JAMA 1997;
prospective randomized study. Int J Radiat Oncol Biol 277:1445–1451.
Phys 1988; 14:1085–1091. 50. Soloway MS, Sharifi R, Wajsman Z, et al: Randomized
38. Pilepich MV, Caplan R, Byhardt RW, et al: Phase III trial prospective study comparing radical prostatectomy alone
of adjuvant androgen suppression using goserelin in versus radical prostatectomy preceded by androgen
unfavorable-prognosis carcinoma of the prostate treated blockade in clinical stage B2 (T2bNxM0) prostate cancer.
with definitive radiotherapy: report of Radiation Therapy J Urol 1995; 154:424.
Oncology Group protocol 85-31. J Clin Oncol 1997; 51. Goldenberg SL, Klotz LH, Srigley J, et al: Randomized,
15:1013–1021. prospective, controlled study comparing radical
39. Lawton CA, Winter K, Murray K, et al: Updated results prostatectomy alone and neoadjuvant androgen
of the phase III radiation therapy oncology group withdrawal in the treatment of localized prostate cancer.
(RTOG) trial 85-31 evaluating the potential benefit of J Urol 1996; 156:873.
androgen suppression following standard radiation 52. Fair WR, Cookson MS, Stroumbakis N, et al: The
therapy for unfavorable prognosis carcinoma of the indications, rationale, and results of neoadjuvant androgen
prostate. Int J Radiat Oncol Biol Phys 2001; 49:937–946. deprivation in the treatment of prostatic cancer. Memorial
504 Part V Prostate Gland and Seminal Vesicles

Sloan-Kettering Cancer Center results. Urology 1997; 61. Hanks GE, Schultheiss TE, Hanlon AL, et al:
49:46. Optimization of conformal radiation treatment of prostate
53. Gleave ME, Goldenberg SL, Chin JL, et al: Randomized cancer: report of a dose escalation study. Int J Radiat
comparative study of 3 versus 8-month neoadjuvant Oncol Biol Phys 1997; 37:543–550.
hormonal therapy before radical prostatectomy: 62. Sanguineti G, Agostinelli S, Foppiano F, et al: Adjuvant
biochemical and pathological effects. J Urol 2001; androgen deprivation impacts late rectal toxicity after
166:500–507. conformal radiotherapy of prostate carcinoma.
54. Messing EM, Manola J, Sarosdy M, et al: Immediate Br J Cancer 2002; 86:1843–1847.
hormonal treatment compared with observation after 63. Vavassori V, Cozzarini C, Bianchi C, et al: Androgen
radical prostatectomy and pelvic lymphadenectomy in deprivation and prostate gland shrinkage during
men with node-positive prostate cancer. New Engl J Med conformal radiotherapy. Int J Radiat Oncol Biol Phys
1999; 341:1781–1788. 2002; 51:18–19.
55. Corn BW, Winter K, Pilepich MV: Does androgen 64. Chen CT, Valicenti RK, Lu JD, et al: Does hormonal
suppression enhance the efficacy of postoperative therapy influence sexual function in men receiving 3D
irradiation: a secondary analysis of RTOG 85-31. Urology conformal radiation therapy for prostate cancer? Int
1999; 54:495–502. J Radiat Oncol Biol Phys 2001; 50:591–595.
56. Dearnaley DP, Khoo VS, Norman AR, et al: Comparison 65. Padula GD, Zelefsky MJ, Venkatraman ES, et al:
of radiation side-effects of conformal and conventional Normalization of serum testosterone levels in patients
radiotherapy in prostate cancer: a randomized trial. treated with neoadjuvant hormonal therapy and three-
Lancet 1999; 353:267–272. dimensional radiotherapy for prostate cancer. Int J Radiat
57. Schultheiss TE, Hanks GE, Hunt MA, et al: Incidence Oncol Biol Phys 2002; 52:439–443.
of and factors related to late complications in conformal 66. Johansson JE, Adami HO, Andersen SO, et al: High
and conventional radiation and treatment of cancer of the 10-year survival rate in patients with early, untreated
prostate. Int J Radiat Oncol Biol Phys 1995; 32:643–649. prostatic cancer. JAMA 1992; 267:2191–2196.
58. Roach M III, Chinn DM, Holland GP, et al: A pilot 67. Roach M III, Lu J, Pilepich MV, et al: Long-term survival
survey of sexual function and quality of life following 3D after radiotherapy alone: radiation therapy oncology
conformal radiotherapy for clinically localized prostate group prostate cancer trials. J Urol 1999; 161:864–868.
cancer. Int J Radiat Oncol Biol Phys 1996; 35:869–874. 68. Roach M III, Lu J, Pilepich MV, et al: Race and survival
59. Litwin M: Measuring health related quality of life in men of men treated for prostate cancer on radiation therapy
with prostate cancer. J Urol 1994; 152:1882–1887. oncology group phase III randomized trials. J Urol 2003;
60. Schultheiss TE, Lee WR, Hunt MA, et al: Late GI and 169:245–250.
GU complications in the treatment of prostate cancer. Int
J Radiat Oncol Biol Phys 1997; 37:3–11.
C H A P T E R

28Metastatic Adenocarcinoma
of the Prostate
Miah-Hiang Tay, MBBS, MRCP, and William K. Oh, MD

The clinical profile of newly diagnosed prostate cancer has original method used to achieve ADT was bilateral sur-
changed considerably over the past decade. A so-called gical orchiectomy.3,4 Castration by orchiectomy is per-
“stage shift” has occurred, coinciding with the widespread manent and is the treatment modality to which other
use of serum prostate-specific antigen (PSA) testing for forms of ADT have been compared.5 ADT can be
prostate cancer screening. As a result, fewer patients are achieved medically by treatments, such as estrogens and
diagnosed initially with metastatic disease. Between 1986 LHRH agonists.
and 1992, 12% of patients presented with metastatic Seidenfeld et al.6 performed a systematic review of
prostate cancer, but by 1998, only 6% of patients with prostate various forms of ADT, analyzing 24 randomized trials
cancer had metastases at presentation.1 Important advances with over 6600 patients with metastatic disease. No sur-
in the treatment of metastatic prostate cancer have been vival difference was noted between surgical and medical
made since Huggins and Hodges2 first showed that pallia- castration using LHRH agonists. Trials comparing the
tion of metastatic disease could be achieved by androgen estrogen, diethylstilbestrol (DES), with orchiectomy or
deprivation therapy (ADT). Despite progress, however, LHRH agonists also revealed no significant difference in
prostate cancer remains a significant problem in the United survival. However, DES had a higher risk of throm-
States. With an estimated 28,900 deaths in 2003, it is the boembolism, which has limited its use as a primary
second leading cause of cancer death among men. modality for ADT in the United States.
LHRH agonists have become the most commonly
used method of ADT in the United States. This prefer-
HORMONE-SENSITIVE METASTATIC PROSTATE
ence is partly explained by the negative psychologic
CANCER
effects patients associate with surgical castration. In a
There has been little change in the primary concept that survey of 159 men with newly diagnosed prostate cancer
guides the initial treatment of metastatic prostate cancer. given a choice of either bilateral orchiectomy or LHRH
Indeed it was over 60 years ago that prostate cancer was agonist therapy, only 22% chose orchiectomy.7 Despite
first shown to be androgen dependent. Since that time, these findings, orchiectomy does offer several advan-
there has been an increase in the number of therapeutic tages, including lack of the tumor flare response associ-
options, as well as controversies regarding the optimal ated with initiation of LHRH agonists and a significantly
form and timing of ADT. In particular, issues, such as the lower overall cost.8
role of combination androgen blockade (CAB) versus Leuprolide acetate and goserelin acetate are com-
luteinizing hormone releasing hormone (LHRH) agonist monly used LHRH agonists and are available in 1-, 3-,
monotherapy, the duration of treatment and the proper and 4-month depot formulations. Both agents have com-
timing of therapy have been debated recently. parable efficacy, with the primary difference being the
route of delivery. Leuprolide acetate is administered
intramuscularly, while goserelin acetate is deposited sub-
Androgen Deprivation Therapy: Standards of Care
cutaneously as a pellet. Newer systems of delivery for
The standard of care today for a man diagnosed with leuprolide are available, which employ a tiny osmotic
metastatic prostatic cancer is the initiation of ADT. The pump that allows for once yearly dosing. As these drugs

505
506 Part V Prostate Gland and Seminal Vesicles

are LHRH agonists, an initial LH surge is seen with led to contradictory results. For example, an influential
resultant increase in testosterone in the initial 1 to 2 Intergroup study (INT-0036) compared leuprolide
weeks following administration. Though this effect acetate with or without flutamide in 603 men with metasta-
diminishes over a month, there is a risk that such a testos- tic prostate cancer.15 Both progression-free (16.5 months
terone flare may cause an acute growth of cancer, leading versus 13.9 months) and overall survivals (35.6 months ver-
to such complications as spinal cord compression and sus 28.3 months) were significantly longer in patients
bladder outlet obstruction in some patients.9,10 Hence, treated with CAB than those treated with leuprolide
antiandrogen therapy is typically administered for at least alone. In addition, a subgroup analysis found that the
1 month and should be started several days to 2 weeks greatest benefits were seen in patients with good per-
prior to the initiation of LHRH agonist, if possible.11 formance status and minimal disease. Two other studies,
LHRH pure antagonists, such as abarelix, are being EORTC 00853 and the Anandron International Study,
investigated as an alternative to LHRH agonists. Studies also showed that CAB favored an improved median sur-
comparing abarelix and LHRH agonists have suggested vival compared to orchiectomy alone.16,17 On the other
that these agents are comparable in efficacy, though there hand, some clinical trials have failed to demonstrate a
is no associated testosterone flare.12,13 benefit to CAB.18–22 The largest and most important of
Estrogenic drugs were used for many years as the pri- these was also an Intergroup trial (INT-0105). One thou-
mary means of medical castration in men with metastatic sand three hundred and seventy eight patients were ran-
prostate cancer. In the 1960s and 1970s, the VA domized to surgical orchiectomy plus flutamide versus
Cooperative Urological Research Group (VACURG) orchiectomy alone. Although a 10% survival benefit for
performed a series of randomized trials designed to eval- the combination arm was seen, it was not statistically sig-
uate estrogenic therapies for prostate cancer.14 Their nificant. This result was different from INT-0036, which
conclusions were: (1) DES at 5 mg/day leads to excess showed a 7-month absolute survival advantage in favor of
cardiovascular deaths, though there were fewer prostate the CAB compared with leuprolide monotherapy. In
cancer-related deaths; (2) estrogen and orchiectomy were addition, INT-0105 failed to show a survival advantage
equivalent and the two together were no better than for patients with minimal disease. Two reasons could
either alone; (3) DES at 1 mg/day was as effective as 5 explain these discrepant findings. The primary difference
mg/day without excess cardiovascular toxicity. Thus between these trials was the use of surgical castration in
doses of DES between 1 and 3 mg/day became the stan- INT-0105, in which case tumor flares cannot occur. It is
dard medical therapy for metastatic prostate cancer in the possible that the primary value of antiandrogen therapy
United States. However, because of the toxicity of DES, is in suppression of a tumor flare in patients with
other forms of nonsurgical hormonal ablation have been metastatic disease. Furthermore, INT-0105 allowed the
sought over the years. Natural estrogens, such as ethinyl use of salvage antiandrogen therapy in patients who pro-
estradiol, and other synthetic estrogens, such as gressed on leuprolide acetate. Since this was not allowed
polyestradiol phosphate (PEP), have been investigated; for patients on INT-0036, any added benefit to up-front
and indeed these drugs have been extensively evaluated CAB may have been abrogated by the use of delayed
and continue to be used outside of the United States. CAB in INT-0105.
Several meta-analyses have been performed to deter-
mine the overall impact of CAB. The largest meta-
Combined Androgen Blockade
analysis is from the Prostate Cancer Trialists’ Collaborative
Though the testicles produce up to 95% of circulating Group, which analyzed 27 clinical trials with over 8000
androgens, the adrenal glands produce a small but poten- men, the majority with metastatic disease.23 This analysis
tially significant amount. After medical or surgical cas- indicated that the addition of antiandrogen to androgen
tration, the adrenal glands become an important source suppression therapy improved 5-year survival by approx-
of androgen.5 Despite the dramatic response to ADT, imately 2% or 3% depending on whether the analysis
prostate cancer eventually becomes hormone refractory included or excluded cyproterone acetate, respectively.
with a median progression-free survival of 18 to 24 Whether this additional small benefit should alter the
months and a median survival of 24 to 30 months. Based standard of care of single modality LHRH agonists or
on the concept that prostate cancer in a “castrate” envi- orchiectomy as first line treatment for patients with
ronment may be sustained by androgen from the adrenal metastatic prostate cancer is controversial, especially
glands, many randomized trials have been performed to when the additional cost and side effects of antiandrogen
determine whether combined androgen blockade (CAB) therapy are considered.
using either medical or surgical castration together with In summary, though it is a controversial area, no com-
an antiandrogen is superior to castration alone. Many of pelling evidence exists for routine CAB use in patients
these trials were underpowered and used different meth- with hormone sensitive metastatic prostate cancer. Both
ods of castration and/or antiandrogens, which may have LHRH agonists and surgical orchiectomy have equivalent
Chapter 28 Metastatic Adenocarcinoma of the Prostate 507

clinical efficacy and can be considered as primary means ADT is poorly defined. This is especially true in the PSA
of ADT, though an antiandrogen should be used for at era, when evidence of recurrence by PSA generally pre-
least 1 month to block a tumor flare response associated dates metastatic disease by many years.27 Contemporary
with LHRH agonists. Nonsteroidal antiandrogens can be guidelines for when to initiate hormonal therapy incor-
subsequently added upon disease progression after pri- porate factors such as PSA doubling time, patient con-
mary ADT. Estrogen therapy is also a choice for primary cerns over libido, and quality of life, and disease
ADT, but toxicity has generally made estrogen use rare in characteristics, such as Gleason score. Little justification
the United States, though it is more popular in Europe. can be made for withholding ADT in the presence of
metastatic disease, whether symptomatic or not. Further
study is needed to determine the optimal timing of ADT.
Timing of Hormonal Therapy
In the VACURG trials, overall survival in patients with
ALTERNATIVE APPROACHES TO HORMONAL
asymptomatic metastatic disease was not significantly
THERAPY
improved by early use of DES compared to placebo.14
However, conclusions from this study may not justify ADTs, such as LHRH agonists and orchiectomy, are
delaying the use of hormonal therapy in a patient with associated with a growing list of previously underappre-
metastatic prostate cancer in the modern era. Since the ciated toxicities. ADT induces castrate levels of testos-
1970s, newer and safer androgen ablation therapies have terone, causing side effects, such as hot flashes, loss of
been developed that abrogate the significant cardiovascu- libido, and osteoporosis. As a result, patients are seeking
lar risks of DES. Furthermore, there are now three stud- alternative hormonal treatments, including intermittent
ies that support the earlier use of ADT for metastatic therapy and antiandrogen monotherapy. Antiandrogens
prostate cancer, though some of these data remain indi- may have less toxicity than ADT, for instance, with libido
rect. In an EORTC study, Bolla et al.24 studied the role and physical capacity. However, randomized trials sug-
of long term adjuvant hormonal therapy following defin- gest that antiandrogen monotherapy is inferior to stan-
itive radiotherapy for high-risk patients, including those dard ADT in controlling metastatic disease, though the
with lymph node disease. In this randomized placebo- difference is not marked.
controlled study of 415 patients, actuarial 5-year survival
for the radiotherapy plus goserelin arm was significantly
Intermittent Androgen Deprivation
higher than the radiotherapy-alone arm (79% versus
62%, respectively; p = 0.001). In another trial by Medical Prostate cancer progresses from a hormone sensitive to
Research Council (MRC) in the United Kingdom, the hormone refractory state by several possible mechanisms.
authors reported that immediate hormonal therapy with First, there is preferential expansion of androgen-
orchiectomy or an LHRH agonist for men with locally independent clones that are present within the tumor at
advanced or asymptomatic metastatic prostate cancer sig- the time ADT is started. Second, there is an upregulation
nificantly reduced the need for transurethral resection of growth factors as a result of adaptation of cancer cells
for local progression, rates of pathologic fracture, spinal to the androgen-depleted environment, as well as activa-
cord compression, ureteral obstruction, and incidence of tion of alternative pathways in androgen receptor signal-
bone metastasis as compared to men randomized to ing. Finally alternate paracrine and autocrine pathways
deferred therapy.25 In addition, deaths attributable to appear to develop that sustain tumor growth.28 By inter-
prostate cancer occurred more frequently in men whose mittently depriving prostate cancer cells of androgen
treatment was deferred (257 versus 203; p = 0.001). stimulation, the aim is to delay the time to androgen
Unfortunately, this study was criticized for the potential independence and preserve the sensitivity of the cancer
bias created by different follow-up schedules between the cells to further ADT in the future. Another potential
two arms, resulting in a number of patients not receiving advantage of intermittent androgen deprivation is an
hormonal therapy in the deferred arm (54% of whom improvement in quality of life with improvements in
died from prostate cancer). A third study that supports libido, erectile function, energy level, and bone density
earlier ADT use is a study by Messing et al.,26 in which during the period when ADT is suspended.
98 men with pelvic lymph node involvement were ran- Intermittent androgen deprivation is usually achieved
domized to early ADT or observation following radical with LHRH agonists until the serum PSA falls to unde-
prostatectomy. At 7 years of follow-up, 7 of 47 (15%) tectable levels (or at least a nadir <4 ng/ml), at which time
men in the immediate therapy arm had died, compared therapy is held. During the first cycle of treatment, ADT
to 18 of 51 (35%) men in the delayed arm (p = 0.02). typically continues for at least 6 to 9 months after the
In summary, there is growing evidence that earlier nadir has been achieved. Androgen deprivation is
ADT may confer a survival benefit compared with signif- resumed once the PSA climbs to an arbitrary fixed level,
icantly delayed therapy, although the optimal timing of for instance, 50% of the initial pretreatment value, an
508 Part V Prostate Gland and Seminal Vesicles

absolute PSA level >5 to 10 ng/ml or (rarely) clinical evi- after an average of 18 to 24 months. The mechanism by
dence of disease. Current data for intermittent ADT indi- which prostate cancer becomes hormone refractory is
cate that about half of patients have resolution of the side poorly understood. A small number of patients who
effects from treatment during the rest period.29–32 More relapse may in fact have noncastrate levels of testosterone
importantly, the disease tends to remain responsive to and, in this situation, surgical orchiectomy should be
subsequent androgen suppression therapy, although the considered. The majority of patients who progress after
treatment-free intervals shorten with each subsequent primary ADT in fact do have castrate testosterone levels.
cycle. Nonetheless, there are several important unre- Despite disease progression, many investigators advocate
solved questions, such as its potential impact on survival continuing ADT. There is evidence of a persistent popu-
in comparison with continuous ADT. Randomized trials lation of hormone sensitive cells, which may regrow if
comparing these two forms of treatment are currently in testosterone is allowed to return to normal levels. One
progress. Until the results of these trials are known, inter- retrospective analysis showed that continuing ADT was
mittent androgen deprivation remains investigational. associated with a survival benefit of 2 to 6 months com-
pared with discontinuation in the hormone-refractory
setting.37
Peripheral Androgen Blockade
Secondary hormonal maneuvers may benefit some
Antiandrogen monotherapy has been extensively evaluated patients after failing primary androgen ablation. In these
in phases II and III trials as initial hormonal therapy for patients, responses may be elicited through the following
advanced prostate cancer.6,13,33 Flutamide is active when pathways: (1) enhanced blocking of residual serum
used as a single agent but inferior to both DES and androgens to the androgen receptor; (2) reduction
orchiectomy in randomized trials. High-dose bicalutamide in adrenal androgen production; (3) binding of agents
has been compared to CAB in at least two large random- to other nuclear receptors in the prostate cancer cells
ized studies in men with locally advanced or metastatic (Table 28-1).
prostate cancer. Though both studies suggested that high- About 20% to 30% of patients will experience a sig-
dose bicalutamide was equivalent to CAB for nonmetasta- nificant PSA decline after antiandrogen withdrawal, last-
tic disease, one of these trials demonstrated a survival ing from 3 to 5 months, although individual responses
benefit in metastatic disease for CAB. There was also evi- can last for over 2 years. Flutamide paradoxically can
dence that bicalutamide monotherapy was better toler- activate specific mutant androgen receptors cloned from
ated, with improved libido and physical capacity. The use prostate cancers, which may provide a mechanism to
of antiandrogens as single agents is accompanied by an explain the antiandrogen withdrawal syndrome.38 The
increase in gonadotropins due to competition in the hypo- blockade of residual serum androgens may be achieved
thalamus of the antiandrogen with circulating androgens. using antiandrogens. In one of the first large series of 209
Sustained gonadotropin increase results in enhanced men, Labrie et al.39 reported an overall response of
testosterone secretion, which may override the antiandro- 34.5% with addition of flutamide after failing initial hor-
gen effect. The inability to demonstrate a role for antian- monal therapy with orchiectomy, DES, or an LHRH
drogens alone, their cost, and their toxicity spectrum have agonist. The mean duration of response was 24 months.
prevented wide acceptance of antiandrogen monotherapy After antiandrogen withdrawal, another antiandrogen
as the initial therapy for advanced prostate cancer. may be considered, as antiandrogens do not appear to
The 5-alpha-reductase inhibitor finasteride has mini- have complete cross-resistance in their mechanism of
mal activity as monotherapy for prostate cancer. action. The use of a second antiandrogen is predicated on
However, finasteride lowers dihydrotestosterone levels the possibility that despite their functional similarities,
and, when added to the antiandrogens flutamide or bica- antiandrogens interact differently with the androgen
lutamide, demonstrates additional PSA suppression. This receptor. There is evidence, for example, that in the
combination may also spare libido and erectile function, LNCaP cell line, flutamide acts as a partial agonist (and
as testosterone is not depleted. Phase II studies of finas- not an inhibitor), whereas bicalutamide maintains an
teride plus flutamide show significant activity in hor- inhibitory function.40 Bicalutamide at dose of 150 to 200
mone sensitive prostate cancer with 72% to 95% mg have been used in clinical trials in patients who pro-
achieving a complete PSA response.34–36 However, in the gressed after primary hormonal manipulation. Expected
absence of randomized data, combined flutamide and response rates from this second line treatment range
finasteride therapy remains investigational. from 15% to 40%. Unfortunately, the response is typi-
cally short-lived, with progression-free survival of about
4 months.41,42
SECONDARY HORMONAL THERAPY
The adrenal gland is the main source of androgens
Despite a response rate of over 80% to 90% with primary following effective castration. Suppressing adrenal
ADT, nearly all men with metastatic disease progress androgens has been demonstrated with ketoconazole and
Chapter 28 Metastatic Adenocarcinoma of the Prostate 509

Table 28-1 Secondary Hormonal Therapy


>50% PSA Measurable
Therapy Number Decline (%) Response (%)

Antiandrogen withdrawal 153 15–33 13

Second antiandrogen

Bicalutamide 134 23–24 0

Nilutamide 14 50 NR

Adrenal androgen inhibitors

Aminoglutethimide 583 NR 9

Ketoconazole 204 78–80 16

Low-dose steroids 241 18–22 NR

Estrogens

DES 405 21–86 NR

PC-SPES 114 45–81 NR

NR, not reported.

aminoglutethimide. Ketoconazole is effective in sup- Estrogens and their analogs also have some activity in
pressing testicular and adrenal androgen production. In patients with hormone-refractory disease. Using 1 mg
vitro experiments also suggest a possible direct cytotoxic DES in 21 men who failed with ADT, Smith et al.47
effect of ketoconazole on prostate cancer cells. Older demonstrated a response rate of 43% based on more than
studies in androgen-independent prostate cancer using 50% decrease in PSA level. Its utility in prostate cancer
high-dose ketoconazole plus replacement steroids has been unfortunately limited by the risk of throm-
showed measurable responses in approximately 15%. boembolic complications but nevertheless is a potentially
Three recent trials of high-dose ketoconazole demon- useful drug in this setting. DES is no longer marketed in
strate much higher response rates using PSA endpoints. the United States but is available through compounding
Small et al.43 treated 48 patients with 400 mg tid in a pharmacies. PC-SPES was supplement consisting of 8
phase II trial and found PSA declines of 50% or greater different herbs and showed estrogenic activity. Its effi-
in 63%. In another trial of 45 patients treated with high- cacy in both androgen sensitive and androgen insensitive
dose ketoconazole, Millikan et al.44 showed a 40% PSA prostate cancer was demonstrated in several trials.48–50
response rate using a similar dose. In CALGB 9583, con- Despite of its potential, PC-SPES has been taken off the
current antiandrogen withdrawal and high-dose keto- market when it was recently found to contain synthetic
conazole demonstrated a 27% PSA response rate and a agents, such as warfarin and DES.
13% measurable response rate.45 Increased gastric pH
decreases drug absorption, so ketoconazole should be
CHEMOTHERAPY
taken on an empty stomach and, if possible, in the
absence of H2 blockers or antacids. Though toxicity is In the traditional view of chemotherapy in hormone-
generally mild or moderate, including nausea, diarrhea, refractory prostate cancer (HRPC), there is either no or
fatigue, and skin changes, some patients require discon- little impact in the history of the disease. Two reviews
tinuation of the drug because of toxicity. A recent phase from 1985 suggested that the response with chemother-
II study suggests that similar response rates may be apy is poor.51,52 In the first review of 17 randomized clin-
obtained with half the traditional dose (i.e., 200 mg tid), ical trials by Eisenberger et al.,53 total response rate was
with fewer apparent side effects.46 In a review of 13 clin- 4.5%, while a second review of 26 trials performed in the
ical trials of aminoglutethimide plus hydrocortisone, late 1980s showed an overall response rate of only 8.7%.
there was an overall partial response rate of 9%. However, recent studies with new drugs and combina-
Aminoglutethimide toxicity includes fatigue, nausea, skin tions have shown that prostate cancer is in fact
rash, orthostatic hypotension, and ataxia. chemosensitive. At the same time, new endpoints of
510 Part V Prostate Gland and Seminal Vesicles

treatment, such as PSA response and parameters of qual- paclitaxel and docetaxel have moderate activities.57,58
ity of life, as well as better supportive measures and Estramustine, by itself, also has modest activity.59 The
improvement in the management of comorbid condi- activity of this drug is not due to the estrogen moiety as
tions, have allowed a resurgence of interest in the use of first thought to be but rather due to its ability to disrupt
chemotherapy to palliate advance disease.54 mitotic activity through the binding of the microtubule
Two randomized trials reestablished the promise of assembly proteins.60 Although estramustine has been
chemotherapy in HRPC, both analyzing mitoxantrone studied in combination with other chemotherapeutic
and a corticosteroid.55,56 In the first study with 161 symp- agents, it is the synergism with other antimicrotubule
tomatic patients, Tannock et al.55 showed that mitox- agents (paclitaxel and docetaxel) that have generated
antrone plus prednisone significantly improved pain enthusiasm. PSA response rates ranging from 50% to
control compared to prednisone alone (29% versus 12%; 65% and from 39% to 82% for estramustine plus pacli-
p = 0.01). The duration of palliation in the chemotherapy taxel or docetaxel, respectively, have been reported in
arm also was significantly longer (43 weeks versus 18 several clinical studies. In spite of the high activity of the
weeks; p < 0.0001). In a second study by Kantoff et al.56 combination therapy, the optimal dose and schedule in
242 patients with HRPC were randomized to receive HRPC are yet to be defined. In fact, the added value of
either mitoxantrone and hydrocortisone or hydrocorti- estramustine in these combinations is counterbalanced by
sone alone. Although there was no significant difference significant toxicity. Several groups have tested the efficacy
in survival, a trend towards longer time-to-progression of single agent docetaxel chemotherapy with reported
and time-to-treatment-failure in the combination arm PSA response ranging from 41% to 47% and less toxic-
was evident (3.7 months versus 2.3 months; p = 0.25). ity.58,61 The Southwest Oncology Group has completed a
Furthermore, there was a higher percentage of patients phase III study with over 600 patients randomized either
in the combination arm who achieved a >50% maxi- to estramustine and docetaxel or mitoxantrone and pred-
mum reduction in PSA (38% versus 22%; p = 0.008), as nisone. Overall survival is the primary endpoint and
well as a trend for improved pain control. Despite a lack results are pending.
of survival benefit, mitoxantrone was approved for pal- Many other chemotherapeutic drugs have also been
liative use in HRPC patients and has become the stan- tested, including etoposide, vinblastine, and cyclophos-
dard of care to which future agents will be compared phamide. In general, these have modest single agent
(Table 28-2). activity in HRPC. On the other hand, results of triple
Taxanes combinations are among the most active reg- drug combination have also been published. One such
imens tested to date in phase II trials. As single agents, regimen consists of weekly paclitaxel, estramustine, and

Table 28-2 Chemotherapy


>50% PSA Measurable
Regimen Number Decline (%) Response (%)

Paclitaxel 41 33–60 22–33

Paclitaxel/estramustine 62 53–61 38–44

Paclitaxel/estramustine/VP-16 40 45 65

Paclitaxel/estramustine/carboplatin 88 67–100 17–45

Docetaxel 113 41–47 28–33

Docetaxel/estramustine 131 63–82 17–57

Docetaxel/estramustine/carboplatin 38 71 47

Mitoxantrone/steroid 199 38 NR

Vinorelbine 77 17–36 13–66

Vinorelbine/estramustine 25 24–65 NR

Vinorelbine/estramustine/VP-16 25 56 32

NR, not reported.


Chapter 28 Metastatic Adenocarcinoma of the Prostate 511

carboplatin. In 56 patients treated, 50% or greater PSA ent radioisotopes appear similar. However, the main tox-
declines were seen in 67% and median survival was 20 icity of these radiopharmaceutical agents is potentially
months.62 Unfortunately, the trial was complicated by severe and prolonged myelosuppression, which may ren-
thromboembolic event in 25% of the patients. Current der a patient not suitable to participate in any potential
estramustine-based clinical trials now also include low- clinical trials with chemotherapy in future.
dose warfarin to decrease the risk of thromboembolism.
Following hormonal therapy failure, chemotherapy
SUMMARY
may control disease progression, as well as to palliate
symptoms. Although mitoxantrone chemotherapy is the There is no doubt that significant progress has been
first to be approved for treatment of hormone-refractory made in treatment of metastatic prostate cancer. ADT
disease, the most active therapies are probably taxane- remains the mainstay of treatment; LHRH agonists,
based. As chemotherapy continues to make inroads into orchiectomy, and estrogen therapy are all standard meth-
management of prostate cancer, patients should be ods of achieving ADT. Alternative methods of hormonal
encouraged to participate in clinical trials. treatment include intermittent ADT and peripheral
androgen blockade. After progression on initial ADT,
secondary hormonal therapies and chemotherapy have
BONE-DIRECTED THERAPIES
growing promise in both palliating patients and poten-
Skeletal complications are a major cause of morbidity for tially controlling disease, though randomized trials are
men with advanced prostate cancer. Over 80% of men ongoing. Current research is focused on several areas
with metastatic prostate cancer have radiologic evidence with promise of better therapeutics against prostate can-
of bone involvement. Palliative external beam radiother- cer in the future. Promising therapeutic approaches
apy has been used for decades for relief of bone pain, but include antisense oligonucleotides, vaccines, angiogene-
recently, bisphosphonates and radiopharmaceutical sis inhibitors, and signal transduction inhibitors.65–69
agents have been approved for use in HRPC.
There are three potential uses of bisphosphonates in
advanced prostate cancer: (1) to prevent osteopenia that
REFERENCES
commonly accompanies the use of ADT; (2) to prevent
or delay skeletal complications in men with bone metas-
1. SEER Cancer Statistics Review—NCI, Bethesda, MD
tasis; (3) to relieve pain from bony disease as well. In the (available at http://seer.cancer.gov/csr/1975_2000/).
United States, zoledronic acid is a bisphosphonate 2. Huggins C, Hodges C: Studies on prostate cancer, effects
approved for use in HRPC. The landmark randomized, of castration of estrogens and androgen injection on
placebo-controlled trial of zoledronic acid in patients serum phosphatase in metastatic carcinoma of the
with HRPC with bone metastasis was reported by Saad et prostate. Cancer Res 1941; 1:293–297.
al.63 In this trial, there were significantly more skeletal- 3. Zhang XZ, Donovan MP, Williams BT, et al:
related events in men receiving placebo compared to Comparison of subcapsular and total orchiectomy for
zoledronic acid (44.2% versus 33.2%; p = 0.021). treatment of metastatic prostate cancer. Urology 1996;
Skeletal-related events were defined as bone fractures, 47:402–404.
4. Bergman B, Damber JE, Tomic R: Effects of total
spinal cord compression, surgery to bone, radiotherapy
and subcapsular orchidectomy on serum concentrations
to bone, or a change in antineoplastic agents. The time of testosterone and pituitary hormones in patients
to first skeletal event was also significantly longer in the with carcinoma of the prostate. Urol Int 1982;
arm receiving zoledronic acid (363 days versus 321 days; 37:139–144.
p = 0.021). In another study, using high-dose clodronate 5. Robinson MR, Thomas BS: Effect of hormonal therapy
as adjuvant therapy, the time to development of sympto- on plasma testosterone levels in prostatic carcinoma.
matic bone metastasis in 311 men with prostate cancer Br Med J 1971; 4:391–394.
was 23.6 months in those receiving clodronate compared 6. Seidenfeld J, Samson DJ, Hasselblad V, et al: Single-
with 19.3 months in placebo arm (p = 0.08).64 However, therapy androgen suppression in men with advanced
significantly more dose reductions were required in prostate cancer: a systematic review and meta-analysis.
clodronate arm due to adverse events (93 versus 108, Ann Intern Med 2000; 132:566–577.
7. Cassileth BR, Soloway MS, Vogelzang NJ, et al: Patients’
hazard ratio, 0.75).
choice of treatment in stage D prostate cancer. Urology
Strontium-89 (89St) chloride, samarium-153 1989; 33:57–62.
(153Sm), and 32-phosphorus (32-P) are approved radio- 8. Chon JK, Jacobs SC, Naslund MJ: The cost value of
pharmaceutical agents for treatment of symptomatic medical versus surgical hormonal therapy for metastatic
bone metastases. There have been no comparative stud- prostate cancer. J Urol 2000; 164:735–737.
ies between the different radioisotopes although the pain 9. Smith JA Jr., Glode LM, Wettlaufer JN, et al: Clinical
response rates and the patterns of pain relief with differ- effects of gonadotropin-releasing hormone analogue in
512 Part V Prostate Gland and Seminal Vesicles

metastatic carcinoma of prostate. Urology 1985; overview of the randomized trials. Lancet 2000;
25:106–114. 355:1491–1498.
10. Debruyne FM, Fernandez del Moral P, Geboers AD: 24. Bolla M, Collette L, Blank L, et al: Long-term results
LHRH-analogues therapy for metastatic prostate cancer. with immediate androgen suppression and external
Prog Clin Biol Res 1988; 260:27–39. irradiation in patients with locally advanced prostate
11. Labrie F, Dupont A, Belanger A, et al: Flutamide cancer (an EORTC study): a phase III randomized trial.
eliminates the risk of disease flare in prostatic cancer Lancet 2002; 360:103–106.
patients treated with a luteinizing hormone-releasing 25. The Medical Research Council Prostate Cancer Working
hormone agonist. J Urol 1987; 138:804–806. Party Investigators Group: Immediate versus deferred
12. McLeod D, Zinner N, Tomera K, et al: A phase III, treatment for advanced prostatic cancer: initial results of
multicenter, open-label, randomized study of abarelix the Medical Research Council Trial. Br J Urol 1997;
versus leuprolide acetate in men with prostate cancer. 79:235–246.
Urology 2001; 58:756–761. 26. Messing EM, Manola J, Sarosdy M, et al: Immediate
13. Iversen P, Tyrrell CJ, Kaisary AV, et al: Casodex hormonal therapy compared with observation after radical
(bicalutamide) 150-mg monotherapy compared with prostatectomy and pelvic lymphadenectomy in men with
castration in patients with previously untreated node-positive prostate cancer. N Engl J Med 1999;
nonmetastatic prostate cancer: results from two 341:1781–1788.
multicenter randomized trials at a median follow-up of 27. Pound CR, Partin AW, Eisenberger MA, et al:
4 years. Urology 1998; 51:389–396. Natural history of progression after PSA elevation
14. The Veterans Administration Co-operative Urological following radical prostatectomy. JAMA 1999;
Research Group: Treatment and survival of patients with 281:1591–1597.
cancer of the prostate. Surg Gynecol Obstet 1967; 28. Galbraith SM, Duchesne GM: Androgens and prostate
124:1011–1017. cancer: biology, pathology and hormonal therapy. Eur
15. Crawford ED, Eisenberger MA, McLeod DG, et al: J Cancer 1997; 33:545–554.
A controlled trial of leuprolide with and without flutamide 29. Crook JM, Szumacher E, Malone S, et al: Intermittent
in prostatic carcinoma. N Engl J Med 1989; 321:419–424. androgen suppression in the management of prostate
16. Keuppens F, Denis L, Smith P, et al: Zoladex and cancer. Urology 1999; 53:530–534.
flutamide versus bilateral orchiectomy. A randomized 30. Klotz L: Hormone therapy for patients with prostate
phase III EORTC 30853 study. The EORTC GU Group. carcinoma. Cancer 2000; 88:3009–3014.
Cancer 1990; 66:1045–1057. 31. Higano CS, Ellis W, Russell K, et al: Intermittent
17. Bertagna C, De Gery A, Hucher M, et al: Efficacy of the androgen suppression with leuprolide and flutamide
combination of nilutamide plus orchidectomy in patients for prostate cancer: a pilot study. Urology 1996;
with metastatic prostatic cancer. A meta-analysis of seven 48:800–804.
randomized double-blind trials (1056 patients). Br J Urol 32. Gleave M, Bruchovsky N, Goldenberg SL, et al:
1994; 73:396–402. Intermittent androgen suppression for prostate cancer:
18. Tyrrell CJ, Altwein JE, Klippel F, et al: Multicenter rationale and clinical experience. Eur Urol 1998; 34(Suppl
randomized trial comparing Zoladex with Zoladex plus 3):37–41.
flutamide in the treatment of advanced prostate cancer. 33. Kolvenbag GJ, Iversen P, Newling DW: Antiandrogen
Survival update. International Prostate Cancer Study monotherapy: a new form of treatment for patients with
Group. Cancer 1993; 72:3878–3879. prostate cancer. Urology 2001; 58:16–23.
19. Iversen P, Suciu S, Sylvester R, et al: Zoladex and 34. Fleshner NE, Trachtenberg J: Combination finasteride
flutamide versus orchiectomy in the treatment of and flutamide in advanced carcinoma of the prostate:
advanced prostatic cancer. A combined analysis of two effective therapy with minimal side effects. J Urol 1995;
European studies, EORTC 30853 and DAPROCA 86. 154:1642–1645 (Discussion 1645-6).
Cancer 1990; 66:1067–1073. 35. Ornstein DK, Rao GS, Johnson B, et al: Combined
20. Boccardo F, Decensi A, Guarneri D, et al: Zoladex with or finasteride and flutamide therapy in men with advanced
without flutamide in the treatment of locally advanced or prostate cancer. Urology 1996; 48:901–905.
metastatic prostate cancer: interim analysis of an ongoing 36. Brufsky A, Fontaine-Rothe P, Berlane K, et al: Finasteride
PONCAP study. Italian Prostatic Cancer Project and flutamide as potency-sparing androgen-ablative
(PONCAP). Eur Urol 1990; 18(Suppl 3):48–53. therapy for advanced adenocarcinoma of the prostate.
21. Fourcade RO, Cariou G, Coloby P, et al: Total androgen Urology 1997; 49:913–920.
blockade with Zoladex plus flutamide vs. Zoladex alone in 37. Taylor CD, Elson P, Trump DL: Importance of continued
advanced prostatic carcinoma: interim report of a testicular suppression in hormone-refractory prostate
multicenter, double-blind, placebo-controlled study. Eur cancer. J Clin Oncol 1993; 11:2167–2172.
Urol 1990; 18(Suppl 3):45–47. 38. Small EJ, Vogelzang NJ: Second-line hormonal therapy
22. Beland G, Elhilali M, Fradet Y, et al: Total androgen for advanced prostate cancer: a shifting paradigm. J Clin
ablation: Canadian experience. Urol Clin North Am 1991; Oncol 1997; 15:382–388.
18:75–82. 39. Labrie F, Dupont A, Giguere M, et al: Benefits of
23. Prostate Cancer Trialists’ Collaborative Group: Maximum combination therapy with flutamide in patients relapsing
androgen blockade in advanced prostate cancer: an after castration. Br J Urol 1988; 61:341–346.
Chapter 28 Metastatic Adenocarcinoma of the Prostate 513

40. Veldscholte J, Berrevoets CA, Mulder E: Studies on the 56. Kantoff PW, Halabi S, Conaway M, et al: Hydrocortisone
human prostatic cancer cell line LNCaP. J Steroid with or without mitoxantrone in men with hormone-
Biochem Mol Biol 1994; 49:341–346. refractory prostate cancer: results of the cancer and
41. Joyce R, Fenton MA, Rode P, et al: High dose leukemia group B 9182 study. J Clin Oncol 1999;
bicalutamide for androgen independent prostate cancer: 17:2506–2513.
effect of prior hormonal therapy. J Urol 1998; 159:149–53. 57. Trivedi C, Redman B, Flaherty LE, et al: Weekly 1-hour
42. Scher HI, Liebertz C, Kelly WK, et al: Bicalutamide for infusion of paclitaxel. Clinical feasibility and efficacy in
advanced prostate cancer: the natural versus treated patients with hormone-refractory prostate carcinoma.
history of disease. J Clin Oncol 1997; 15:2928–2938. Cancer 2000; 89:431–436.
43. Small EJ, Baron AD, Fippin L, et al: Ketoconazole retains 58. Picus J, Schultz M: Docetaxel (Taxotere) as monotherapy
activity in advanced prostate cancer patients with in the treatment of hormone-refractory prostate cancer:
progression despite flutamide withdrawal. J Urol 1997; preliminary results. Semin Oncol 1999; 26:14–18.
157:1204–1207. 59. Hudes G: Estramustine-based chemotherapy. Semin Urol
44. Millikan R, Baez L, Banerjee T, et al: Randomized phase Oncol 1997; 15:13–19.
2 trial of ketoconazole and ketoconazole/doxorubicin in 60. Stearns ME, Tew KD: Estramustine binds MAP-2 to
androgen independent prostate cancer. 2001; 6:111–115. inhibit microtubule assembly in vitro. J Cell Sci 1988;
45. Oh WK: Secondary hormonal therapies in the 89(Pt 3):331–342.
treatment of prostate cancer. Urology 2002; 60:87–92 61. Berry W, Dakhil S, Gregurich MA, et al: Phase II trial of
(Discussion 93). single-agent weekly docetaxel in hormone-refractory,
46. Harris KA, Weinberg V, Bok RA, et al: Low dose symptomatic, metastatic carcinoma of the prostate. Semin
ketoconazole with replacement doses of hydrocortisone in Oncol 2001; 28:8–15.
patients with progressive androgen independent prostate 62. Kelly WK, Curley T, Slovin S, et al: Paclitaxel,
cancer. J Urol 2002; 168:542–545. estramustine phosphate, and carboplatin in patients
47. Smith DC, Redman BG, Flaherty LE, et al: A phase II with advanced prostate cancer. J Clin Oncol 2001;
trial of oral diethylstilbestrol as a second-line hormonal 19:44–53.
agent in advanced prostate cancer. Urology 1998; 63. Saad F, Gleason DM, Murray R, et al: A randomized,
52:257–260. placebo-controlled trial of zoledronic acid in patients with
48. Oh WK, George DJ, Hackmann K, et al: Activity of the hormone-refractory metastatic prostate carcinoma. J Natl
herbal combination, PC-SPES, in the treatment of Cancer Inst 2002; 94:1458–1468.
patients with androgen-independent prostate cancer. 64. Dearnaley D, Sydes MR, et al. Preliminary evidence that
Urology 2001; 57:122–126. oral clodronate delays symptomatic progression of bone
49. DiPaola RS, Zhang H, Lambert GH, et al: Clinical and metastases from prostate cancer: first results of MRC
biologic activity of an estrogenic herbal combination Pr05 trial (abstract). Proc Am Soc Clin Oncol 2001;
(PC-SPES) in prostate cancer. N Engl J Med 1998; 20:174a.
339:785–791. 65. Signoretti S, Montironi R, Manola J, et al: Her-2-neu
50. Small EJ, Frohlich MW, Bok R, et al: Prospective trial of expression and progression toward androgen
the herbal supplement PC-SPES in patients with independence in human prostate cancer. J Natl Cancer
progressive prostate cancer. J Clin Oncol 2000; Inst. 2000; 92(23): 1918–1925.
18:3595–3603. 66. Simons JW, Mikhak B, Chang JF, et al: Induction of
51. Tannock IF: Is there evidence that chemotherapy is of immunity to prostate cancer antigens: results of a clinical
benefit to patients with carcinoma of the prostate? J Clin trial of vaccination with irradiated autologous prostate
Oncol 1985; 3:1013–1021. tumor cells engineered to secrete granulocyte-macrophage
52. Eisenberger MA, Simon R, O’Dwyer PJ, et al: A reevaluation colony-stimulating factor using ex vivo gene transfer.
of nonhormonal cytotoxic chemotherapy in the treatment of Cancer Res 1999; 59:5160–5168.
prostatic carcinoma. J Clin Oncol 1985; 3:827–841. 67. Vaishampayan U, Fontana J, Du W, et al: An active
53. Oh WK, Kantoff PW: Management of hormone regimen of weekly paclitaxel and estramustine in
refractory prostate cancer: current standards and future metastatic androgen-independent prostate cancer.
prospects. J Urol 1998; 160:1220–1229. Urology 2002; 60:1050–1054.
54. Dawson NA, McLeod DG: The assessment of treatment 68. O’Reilly MS, Boehm T, Shing Y, et al: Endostatin: an
outcomes in metastatic prostate cancer: changing endogenous inhibitor of angiogenesis and tumor growth.
endpoints. Eur J Cancer 1997; 33:560–565. Cell 1997; 88:277–285.
55. Tannock IF, Osoba D, Stockler MR, et al: Chemotherapy 69. Murphy GP, Tjoa B, Simmons SJ, et al: Phase II prostate
with mitoxantrone plus prednisone or prednisone alone cancer vaccine trial: report of a study involving 37 patients
for symptomatic hormone-resistant prostate cancer: a with disease recurrence following primary treatment.
Canadian randomized trial with palliative end points. Prostate 1999; 39:54–59.
J Clin Oncol 1996; 14:1756–1764.
C H A P T E R

29 Anatomic Nerve-Sparing Radical


Retropubic Prostatectomy
Misop Han, MD, and William J. Catalona, MD

The management of clinically localized prostate cancer comorbidity that might make the operation unacceptably
has undergone substantial changes over the past two risky. Actuarial life tables can project the life expectancy
decades. Widespread screening with serum prostate- of U.S. men,8 and with appropriate adjustment for
specific antigen (PSA) testing and digital rectal examina- comorbidities, life expectancy can be estimated for the
tion has allowed much earlier detection of prostate cancer individual patient.
during this era.1,2 In addition, the modification of surgical After confirming the likelihood of a sufficiently long
technique by Walsh has allowed better hemostasis, life expectancy, the next step in patient selection is to
improved visualization during dissection, and preserva- identify those with potentially curable disease. Radical
tion of neurovascular bundles supplying corpora caver- prostatectomy provides the best chance for cure for men
nosa.3 As a result, a skilled surgeon can perform radical whose tumor is confined to the prostate gland. As a
prostatectomy with a high cure rate, while preserving uri- result of widespread screening for prostate cancer and
nary continence and erectile potency in the majority of more restrictive preoperative patient selection, the pro-
patients. Thus, since 1990, radical prostatectomy has portion of men with organ-confined or specimen-
been the most commonly performed treatment for clini- confined disease has increased in recent years.9
cally localized prostate cancer.4 In a landmark study, However, the lack of accuracy of conventional radi-
Holmberg et al.5 recently reported on the first prospec- ographic imaging studies in staging prostate cancer has
tive, randomized trial showing that radical prostatectomy been disappointing. Therefore, nomograms predicting
reduces the rates of metastases and death from prostate the pathologic stage based on preoperative clinical and
cancer. Consequently, the rationale for treating clinically pathologic parameters have been widely used to identify
localized prostate cancer surgically is convincing. patients who are likely to benefit from the surgical resec-
In this chapter, we discuss patient selection, surgical tion and those who are not.10,11 Alternatively, nomo-
technique, outcomes, and complications of anatomic rad- grams predicting postsurgical or postradiation therapy
ical retropubic prostatectomy using the senior author’s recurrence-free survival probabilities also are sometimes
surgical series of more than 3500 anatomic radical useful for patients.12–14 For patients with a low probabil-
prostatectomies as an example. The outcomes are similar ity of resectable disease or a short life expectancy due to
to those of other previously reported, large radical age or comorbidity, an alternative treatment to surgery
prostatectomy series in the PSA era.6,7 It is not only rep- should be recommended.
resentative of large modern prostatectomy series but also For the patient to have realistic expectations concern-
includes all men who underwent surgery in the analysis, ing postoperative potency and continence outcomes, the
even those with known adverse prognostic features. surgeon should provide the patient with relevant infor-
mation on the nerve-sparing aspect of radical prostatec-
tomy during the preoperative consultation. Anatomic
PATIENT SELECTION
nerve-sparing radical retropubic prostatectomy is a safe
An ideal candidate for radical prostatectomy should have choice without compromising cancer control in appropri-
a life expectancy of at least 10 years, a completely ately selected patients. Nerve-sparing radical prostatec-
resectable and biologically significant tumor, and no tomy is inappropriate in men with locally advanced

514
Chapter 29 Anatomic Nerve-Sparing Radical Retropubic Prostatectomy 515

disease, especially if the primary goal of the surgery is and the puboprostatic ligaments; (3) ligation, proximal
cancer control. The feasibility of the nerve-sparing sur- and distal suture ligation, and transection of the dorsal
gery is questionable when a patient has extensive involve- venous complex; (4) dissection of the prostate from the
ment by cancer of prostate biopsies, palpable evidence on neurovascular bundles; (5) vascular control and transec-
digital rectal examination of possible extraprostatic exten- tion of the prostatic pedicles; (6) transection and recon-
sion, a serum PSA level >10 ng/ml, a biopsy Gleason score struction of the bladder neck; (7) dissection of the
>7, poor quality erections preoperatively, a lack of interest seminal vesicles and ampullary portions of the vasa def-
and/or willingness of a partner in restoring potency, or erentia; and (8) performance of the vesicourethral anas-
the presence of other medical conditions that may tomosis. These steps are described in detail here with
adversely affect potency, such as diabetes mellitus, hyper- corresponding illustrations.
tension, psychologic or psychiatric diseases, neurologic
diseases, and medications. Therefore, it is important to
1. Limited Pelvic Lymphadenectomy
review the clinicopathologic features of the tumor and the
patient’s medical history and erectile function status A superficial midline (or transverse) lower abdominal
before embarking on a nerve-sparing operation. incision is made with a scalpel. The linea alba is incised
After discussing the prospects for preservation of and the space of Retzius is entered. Taking care to avoid
potency, information on the treatment of erectile dys- disrupting the lymphatic tissue lateral to the external iliac
function should be imparted. This should include pro- vein and to avoid compression of the vein itself, a Balfour
viding information on phosphodiesterase inhibitors, retractor is placed. A modified pelvic lymphadenectomy
intraurethral and intracorporal vasodilators, vacuum is performed, removing only the lymph nodes medial to
erection devices, venous flow constrictors, and artificial the external iliac vein. Care is taken during the lym-
penile prostheses. The discussion should include the phadenectomy to preserve any accessory arterial
anticipated postoperative erectile rehabilitation program branches to the corpora cavernosa that arise from the dis-
to be used and the timing of the return of erections that tal external iliac or obturator arteries. The obturator
usually begins 3 to 6 months postoperatively and lasts for nerve is identified and preserved. In most incidences, the
up to 36 months. If erectile function is of paramount patient elects to have the prostate gland removed, even if
importance to the patient, he can be reassured that erec- there are pelvic lymph node metastases; otherwise, the
tions can be almost always restored, regardless of excised lymph node packet is sent for frozen-section
whether or not nerve-sparing surgery can be successfully examination. If the frozen section examination reveals
performed. metastatic cancer, it is unlikely that the patient will be
Finally, the surgeon should discuss possible need for cured by radical prostatectomy, and the operation is ter-
and potential side effects of adjuvant radiation therapy or minated. Lymphadenectomy is optional in patients who
hormonal therapy if the final pathology report reveals are at low risk for pelvic lymph node metastases by virtue
adverse prognostic features. At the end of the preopera- of a low Gleason grade, low PSA, and low biopsy tumor
tive counseling session, if nerve-sparing radical retropu- volume.
bic prostatectomy is appropriate, the patient and his After completing the lymphadenectomy, the adipose
spouse or partner should sign an informed consent form and areolar tissues are swept gently from the anterior
authorizing a surgeon to perform the procedure. surface of the prostate and the endopelvic fascia to expose
the puboprostatic ligaments. Care is taken to avoid injury
to the perforating branches of Santorini’s plexus that
SURGICAL TECHNIQUE
pierce the endopelvic fascia between the puboprostatic
Before the operation, a first-generation cephalosporin ligaments and pass cephalad on the anterior surface of
antibiotic is given intravenously. After a general endotra- the prostate gland and bladder.
cheal or regional anesthesia is administered, thigh-high
elastic hose are placed on the patient. Sequential com-
2. Incision of the Endopelvic Fascia and the
pression devices are used only in patients with increased
Puboprostatic Ligaments
risk for thromboembolic complications. The patient is
positioned with his legs on spreader bars, and the operat- The endopelvic fascia is incised in the groove between
ing table is dorsiflexed with the break just above the the levator ani muscles and the lateral border of the
patient’s anterosuperior iliac spine (Figure 29-1). The prostate (Figure 29-2). Inside the endopelvic fascia, the
abdomen and genitalia are appropriately prepped and lateral surface of the prostate is covered by a smooth,
draped. glistening membrane overlying the lateral portion of
There are eight key steps in performing anatomic Santorini’s plexus. Strands of the levator ani muscles are
nerve-sparing radical prostatectomy: (1) a limited pelvic gently dissected off the prostate to the level of the uro-
lymphadenectomy; (2) incision of the endopelvic fascia genital diaphragm. Often, venous tributaries pass from
516 Part V Prostate Gland and Seminal Vesicles

Figure 29-1 Positioning of the patient. A,Legs are separated on spreader bars. B, The
operating table is flexed with the break just above the patient’s anterosuperior iliac spine.

the levator ani muscles to the prostate just lateral to the between the anterior surface of the urethra and the dor-
puboprostatic ligaments. These vessels are either cauter- sal venous complex is developed with a pinching motion
ized, secured with hemostatic clips, or ligated laterally, of the left index finger and thumb. The plane between
and then clamped medially with a delicate snub-nose the urethra and the dorsal venous complex is then devel-
right-angle clamp. After the vein is transected sharply, its oped gently, first with a large right-angle clamp. This
medial portion is ligated. When the endopelvic fascia has facilitates tight ligation of the dorsal venous complex.
been opened from the base to the apex of the prostate, the After the dorsal venous complex has been ligated, it is
superficial branch of Santorini’s plexus is gently retracted also suture ligated in a slightly more caudal site with a 2-0
medially, and the puboprostatic ligaments are placed on chromic catgut suture on a CT-1 needle (Figure 29-4). A
stretch and divided close to the pubic symphysis (Figure suture ligature is also placed in the anterior surface of the
29-3). Care is taken not to divide the puboprostatic liga- prostate to reduce the back-bleeding from Santorini’s
ments too medially or too far under the pubic symphysis plexus (Figure 29-5).
to avoid injuring the dorsal venous complex. The right-angle clamp is then passed behind the dor-
sal venous complex and the jaws of the clamp are spread.
The dorsal venous complex is transected with electro-
3. Suture Ligation and Transection of the Dorsal
cautery or a scalpel (Figure 29-6). Back-bleeding from
Venous Complex
the dorsal venous complex is controlled with figure-of-
After the puboprostatic ligaments have been divided, the eight 3-0 sutures. It is important to obtain good hemo-
lateral surfaces of the urethra are palpated. The groove stasis so that the apical dissection of the prostate may be
Chapter 29 Anatomic Nerve-Sparing Radical Retropubic Prostatectomy 517

Figure 29-2 The endopelvic fascia is incised in the groove between the levator ani muscles
and the lateral border of the prostate.

Figure 29-3 The puboprostatic ligaments are placed on stretch and incised.
518 Part V Prostate Gland and Seminal Vesicles

Figure 29-4 The dorsal venous complex is suture ligated with a 2-0 chromic catgut suture
on a CT-1 needle.

Figure 29-5 To reduce back-bleeding from Santorini’s plexus, the cephalad aspect of the
dorsal venous complex is suture ligated.
Chapter 29 Anatomic Nerve-Sparing Radical Retropubic Prostatectomy 519

Figure 29-6 The dorsal venous complex is transected with a right-angle clamp jaws spread
behind the complex.

performed in a relatively bloodless field. If the dorsal used to elevate the lateral pelvic fascia from the underly-
venous complex ligature slips off, the complex is over- ing veins on the surface of the prostate. Small perforating
sewn using a 3-0 chromic catgut suture on a 5/8-circle vessels are secured with hemoclips, ties, or ligatures to
needle. The goal in oversewing the complex is to pass the ensure adequate hemostasis. The posterolateral groove
suture just through the lateral borders of the complex between the prostate and the neurovascular bundles is
itself in its anterior, middle, and posterior aspects, developed using sharp and blunt dissection, allowing the
respectively. Wide, imprecisely placed sutures may dam- prostate to assume a more anterior position in the pelvis.
age the neurovascular bundles. The lateral aspect of the prostate is then dissected
The anterior surface of the urethra is palpated from the neurovascular bundles, allowing the bundles to
between the neurovascular bundles. The circumurethral retract laterally. In a case of extensive fibrosis, the dissec-
sphincter muscle and the anterior wall of the urethra are tion is performed sharply. The dissection is carried
incised with a scalpel just distal to the apex of the prostate cephalad until the portion of Denonvilliers’ fascia cover-
without dissecting around the lateral or posterior sur- ing the ampullary portions of the vasa deferentia and the
faces of the urethra (Figures 29-7 and 29-8). The incision seminal vesicles is exposed (Figure 29-10). Denonvilliers’
should not be carried too far lateral, where it may injure fascia is incised with the cautery. The Metzenbaum scis-
the neurovascular bundles. The urethral catheter is sors are then used to develop the proper plane of dissec-
exposed and carefully hooked with a delicate right-angle tion for the prostatic vascular pedicles. If there is
clamp. Gentle traction on the clamp in a cephalad direc- continued bleeding from the periurethral tissues and api-
tion exposes the posterior urethral wall. The catheter is cal pedicles of the prostate, hemostatic sutures should be
divided and placed on cephalad traction, the posterior placed at this juncture to avoid continued blood loss dur-
urethral wall is sharply transected. Fibromuscular bands ing the remainder of the procedure.
tethering the apex of the prostate to the pelvic floor are
incised using sharp dissection (Figure 29-9). The rec-
5. Vascular Control and Transection of Prostatic
tourethralis muscle is incised, exposing the prerectal fat.
Pedicles
The prostatic pedicles are divided by inserting the right-
4. Separation of the Prostate from the
angle clamp medial to them, with the tip of the clamp
Neurovascular Bundles
directed almost parallel to the lateral surface of the
The lateral pelvic fascia is incised from the apex of the prostate. The prostatic pedicle is ligated or hemoclipped
prostate to the base. A delicate right-angle clamp may be laterally, taking care to place the tie or clip medial to the
520 Part V Prostate Gland and Seminal Vesicles

Figure 29-7 The circumurethral external sphincter muscle fibers are incised to expose the
urethra.

Figure 29-8 The anterior wall of the urethra is incised with a scalpel without dissecting
around the lateral or posterior surfaces of the urethra.
Chapter 29 Anatomic Nerve-Sparing Radical Retropubic Prostatectomy 521

Figure 29-9 The apical pedicles of the prostate may require suture ligation. Fibromuscular
bands tethering the apex of the prostate to the pelvic floor are incised using sharp
dissection. The prostate gland is dissected from neurovascular bundles.

Figure 29-10 The dissection is carried cephalad until the portion of Denonvilliers’ fascia
covering the ampullary portions of the vasa deferentia and the seminal vesicles is exposed.
Denonvilliers’ fascia is incised with the cautery. Denonvilliers’ fascia is incised to expose
vascular pedicles at prostate base.
522 Part V Prostate Gland and Seminal Vesicles

neurovascular bundle (Figure 29-11). The pedicle is


7. Dissection of Seminal Vesicles and Ampullary
divided close to the prostate. This dissection is per-
Portions of the Vasa Deferentia
formed on both sides to a point just cephalad to the sem-
inal vesicles. Care is taken when dissecting near the The seminal vesicles are dissected first along their lateral
seminal vesicles to avoid injuring the neurovascular bun- edges, carrying the plane of dissection medially. Many
dles that are situated just lateral to the seminal vesicles. small perforating arteries enter the lateral and terminal
The seminal vesicles are freed from the bladder base portions of the seminal vesicles. These are secured with
using sharp and blunt dissection, and a large right-angle small hemoclips. The ampullae are freed, using sharp and
clamp is used to further develop this plane. Two hemo- blunt dissection, and then are clipped and transected.
static sutures of 3-0 chromic catgut are placed in the lat- After the seminal vesicles have been dissected to their tips
eral bladder pedicles cephalad to the seminal vesicles, one and the hemoclips placed, the surgical specimen is
just lateral to the prostate and another just medial to the removed. At this point, the pelvis is carefully inspected
neurovascular bundles. The lateral bladder neck fibers for hemostasis. Small bleeders on the neurovascular bun-
are then partially incised with the cautery but are not dles may require 4-0 absorbable suture ligatures. It is
incised through their entire thickness. important not to use the cautery for hemostasis on the
neurovascular bundles, to avoid cautery injury to the cav-
ernosal nerves. Suture ligatures of 3-0 or 4-0 absorbable
6. Transection and Reconstruction of the Bladder
material are placed in the “pockets” of the seminal vesi-
Neck
cle pedicles on the medial aspects of the neurovascular
The anterior bladder neck is transected with electro- bundles to ensure good hemostasis in this difficult-to-
cautery in the natural groove between the bladder and visualize region.
the prostate. The bladder neck opening is enlarged with
scissors, and the catheter is pulled through and used as a
8. Vesico-Urethral Anastomosis
tractor on the prostate (Figure 29-12). The posterior
bladder neck is incised with the cautery. The muscular Reconstruction of the bladder neck begins by placing a
attachments between the bladder and prostate are continuous running everting suture of 3-0 chromic
divided using electrocautery and/or hemostatic clips for catgut that encompass bladder mucosa and underlying
hemostasis. muscle for a distance of nearly the entire anastomotic

Figure 29-11 Prostate base pedicle is ligated or hemoclipped laterally, taking care to place
the tie medial to the neurovascular bundle.
Chapter 29 Anatomic Nerve-Sparing Radical Retropubic Prostatectomy 523

Figure 29-12 The anterior bladder neck is transected in the natural groove between the
bladder and the prostate. The bladder neck opening is enlarged with scissors. The ureteral
orifices are identified.

circumference (Figure 29-13). The bladder neck is then the bladder neck is guided gently toward the cut end of
reconstructed in a tennis racket fashion, with the handle the urethra. The anastomotic sutures are tied carefully
of the racket directed posteriorly. The bladder neck clo- under direct vision. The bladder is then irrigated free of
sure is accomplished in two layers with a continuous 3-0 clots, a single suction drain is placed in the pelvis and
chromic catgut suture on the first layer and a continuous brought out the lower end of the wound. The incision is
2-0 chromic catgut suture on the second layer. Care closed with #1 loop Maxon running sutures on the fascia,
should be taken to avoid compromising the ureteral ori- 2-0 chromic catgut suture on the subcutaneous tissue,
fices. The bladder neck is closed to a size of approxi- and a 4-0 polyglycolic acid subcuticular suture on the
mately 22Fr to 24Fr. skin. The skin incision is covered with Steri-Strips.
An 18Fr catheter with a 30-ml balloon is passed
through the urethra. While an assistant exerts pressure
Postoperative Care
on the perineum with a sponge forceps to better expose
the cut end of the urethra (Figure 29-14), double-armed Patients are ambulated with assistance once the night of
2-0 chromic catgut sutures are used for the vesi- surgery, 5 times on the first postoperative day and 7 times
courethral anastomosis (Figure 29-15). A 5/8-circle nee- on the second postoperative day. A clear liquid diet is
dle is used to place the sutures in the urethra from inside given on the night of surgery, advancing to a regular diet
to outside, avoiding placing the suture into the neurovas- as tolerated on the following days. A suction drain and
cular bundles. The tip of the catheter is grasped and dressing are removed on the second postoperative day.
brought out of the wound to expose the posterior lip of Intravenous antibiotics are discontinued after the suction
the cut end of the urethra. The posterior sutures are sim- drain is removed. For analgesia, Ketorolac (30 to 60 mg)
ilarly placed. The anterior sutures are placed at the 10 is given intravenously every 6 hours for the first 48 hours.
o’clock and 2 o’clock positions and the posterior sutures It may be supplemented sparingly with morphine, as
are placed at the 5 o’clock and 7 o’clock positions. The needed. Antibiotic ointment is applied to the urethral
other ends of the sutures containing an SH 3/8-circle meatus around the catheter 4 to 6 times a day until the
needle are placed in the corresponding positions of the catheter removal. Most patients are discharged from the
bladder neck from inside to outside. These sutures hospital on the second or third postoperative day.
encompass mucosa and muscle and exit at the edge of the The catheter may be removed on either the seventh,
mucosa. The catheter tip is placed in the bladder, and 10th, or 14th postoperative day, depending on the perceived
524 Part V Prostate Gland and Seminal Vesicles

Figure 29-13 A continuous running mucosa-everting suture of 3-0 chromic catgut is placed
for a distance of nearly the entire anastomotic circumference.

Figure 29-14 Perineal pressure is applied with a sponge forceps to better expose the cut
end of the urethra.
Chapter 29 Anatomic Nerve-Sparing Radical Retropubic Prostatectomy 525

Figure 29-15 Double-armed 2-0 chromic catgut sutures are used for the vesicourethral
anastomosis.

amount of tension on the vesicourethral anastomosis. A An analysis of the senior author’s series, including
cystogram is not performed before removing the catheter more than 3170 men who underwent anatomic radical
unless an anastomotic leak is suspected. The catheter retropubic prostatectomy between 1983 and 2002,
should not be removed before 7 days, as 10% to 15% of including those with adverse prognostic features, has
men may experience urinary retention from edema and been presented.18,19 Cancer progression was defined as
require recatheterization.15,16 Oral fluoroquinolone is detectable serum PSA (>0.2 ng/ml), local recurrence, or
given 1 day before and 1 week following the catheter distant metastases. With a median follow-up of 4.5 years
removal. Daily Kegel exercises are performed in four sets (mean 5.3, range 0 to 18), cancer progression occurred in
of 10, before the surgery and following the catheter 19% of the men following radical prostatectomy.
removal until continence returns. A protective pad or Actuarial 10-year cancer progression-free survival proba-
diaper is used until a complete urinary control is bility was 67%.
achieved. The first postoperative serum PSA level is Cancer progression following radical prostatectomy
measured 2 weeks or more after the operation. was strongly associated with many clinical and pathologic
parameters, including Gleason grade, clinical, and patho-
logic tumor stage, era of treatment, and patient age. For
CANCER CONTROL OUTCOME
example, preoperative serum PSA level was inversely
The most important objective of radical prostatectomy is associated with both the rate of organ-confined disease
cancer control. To cure prostate cancer with radical and the 10-year progression-free survival rate. Patient
prostatectomy, the patient must have a resectable tumor selection and the duration and frequency of follow-up
and the surgery must completely encompass the tumor.9 monitoring are critical in determining outcomes as well.
A rising serum PSA level is usually the earliest evidence Therefore, factors other than treatment effectiveness can
of recurrence or progression following surgery.17 Follow- influence treatment outcomes. Accordingly, caution is
up data are still not sufficiently mature to effectively eval- indicated in comparing the results of contemporary rad-
uate cancer-specific survival trends. For example, in large ical prostatectomy series using different patient selection
contemporary radical retropubic prostatectomy series, criteria and follow-up protocols.
including the current series, actuarial 10-year cancer-
specific survival ranged between 96% and 98%.6,7,18
URINARY CONTINENCE OUTCOME
Therefore, biochemical recurrence (detectable serum
PSA)-free survival has been used frequently as a surro- The overall urinary continence outcome following
gate in evaluating the treatment efficacy in radical nerve-sparing radical retropubic prostatectomy was
retropubic prostatectomy series. excellent in the current series. More than 93% of men
526 Part V Prostate Gland and Seminal Vesicles

achieved complete urinary continence, defined as requir- experience. The most common complications of
ing no protection for daily activities.19 The return of uri- anatomic nerve-sparing radical retropubic prostatectomy
nary continence was strongly associated with the age of included anastomotic stricture/bladder neck contracture,
the patient. For example, more than 95% of men thromboembolic complications (deep vein thrombosis
younger than age 50 were continent following surgery. In and pulmonary embolism), and postoperative inguinal
contrast, 85% of men above age 70 were continent post- hernia occurrence. In the current series, the rate of anas-
operatively. There was no significant difference in conti- tomotic stricture/bladder neck contracture decreased
nence rate according to the era. Only 4 men (0.2%) from 8% in the 1980s to <1% in the 1990s. Similarly, a
eventually required an artificial urinary sphincter place- drastic decrease in thromboembolic events was observed
ment for stress urinary incontinence. with the rate decreasing from 3% to 1% during the past
20 years. Other less frequent complications (<1%) asso-
ciated with radical prostatectomy included infection,
POTENCY OUTCOME
lymphocele formation, neurologic deficit, and cardiovas-
There are several possible goals of the nerve-sparing cular events.
aspect of radical retropubic prostatectomy. Patients with Anastomotic stricture can be initially managed with a
intact libido and erectile potency want to maintain cur- gentle, serial dilation. Alternatively, a careful internal
rent quality of erections or erections sufficient for pene- urethrotomy can be performed. For a long and persistent
tration with the help of oral medication, such as stricture, a transurethral resection of the scar tissue above
phosphodiesterase inhibitors. Others with poor quality the external sphincter may be necessary. After resection,
erections preoperatively might accept erections that at triamcinolone can be injected via cystourethroscopic
least offer some rigidity to provide sensory satisfaction approach to prevent inflammatory response and subse-
for both sexual partners. The erectile potency in the cur- quent, recurrent scar formation.
rent series was defined as an ability to maintain erections Inadvertent injury to the obturator nerve can occur
strong enough for penetration with or without the help during the pelvic lymphadenectomy. One management
of oral phosphodiesterase inhibitor. option is to treat it conservatively with physical therapy
The return of erectile potency following radical since some patients usually do not exhibit significant
retropubic prostatectomy was strongly associated with thigh adductor deficit following the injury.21 Otherwise,
the age of the patient, preoperative potency status, nerve- a primary nerve repair can be attempted. When a ten-
sparing status (bilateral sparing versus partial sparing), sion-free primary nerve repair is not feasible, a nerve
and the era of surgery (1980s versus 1990s).19 More than grafting can be performed utilizing either the sural nerve
75% of men younger than age 60 regained potency fol- or the lateral antebrachial cutaneous nerve.22
lowing bilateral nerve-sparing radical retropubic prosta- An injury to the ureter can occur inadvertently during
tectomy. For men below age 50, more than 95% the transection of bladder neck or the dissection of the
recovered potency following surgery, in the modern era. lateral prostate pedicles. When recognized, a simple
Between 62% and 72% of men in their 60s became mobilization of the ureter and ureteroneocystostomy
potent following bilateral nerve-sparing surgery. Finally, should be performed. The reimplanted ureter can be
there was a significant improvement in recovery of cannulated using a 5Fr or 8Fr pediatric-feeding tube to
potency in men treated in the 1990s compared to those prevent an edema at the reimplantation site.
treated in the 1980s, even after correcting for the age and Usually, a rectal injury can be repaired primarily using
nerve-sparing status. a multiple layer closure.23 However, a diverting
colostomy should be strongly considered in men with a
large rectal defect or a history of pelvic radiation therapy.
COMPLICATIONS
A diverting colostomy should also be considered in those
The hospital cancer registry survey performed by the on a long-term preoperative steroid therapy.
American College of Surgeons reported a perioperative A vesicorectal fistula can develop following anatomic
(within 30 days of surgery) mortality rate of 0.4% fol- radical retropubic prostatectomy. This uncommon,
lowing radical prostatectomy.20 There was no intraoper- iatrogenic fistula can be repaired using a transrectal,
ative or immediate postoperative mortality in the current transsphincteric (York-Mason) approach with excellent
series. With a careful selection of patients and perform- results.24–26
ance of necessary cardiovascular evaluation, perioperative
mortality can be largely avoided.
SUMMARY
The overall complication rate of radical prostatectomy
was 8% in the current series. Initially, the complications Anatomic nerve-sparing radical retropubic prostatec-
occurred more commonly in older men, but the overall tomy provides excellent cancer control with an accept-
complication rate gradually decreased with the surgeon’s able rate of complications in appropriately selected
Chapter 29 Anatomic Nerve-Sparing Radical Retropubic Prostatectomy 527

patients. Many clinical and pathologic parameters are 12. Kattan MW, Eastham JA, Stapleton AM, Wheeler TM,
associated with cancer control and return of urinary con- Scardino PT: A preoperative nomogram for disease
tinence and potency following surgery. Over the past recurrence following radical prostatectomy for prostate
2 decades, widespread screening for prostate cancer and cancer. J Natl Cancer Inst 1998; 90:766–771.
13. Han M, Partin AW, Zahurak M, et al: Biochemical
better patient selection have resulted in a favorable shift
(prostate specific antigen) recurrence probability following
of these parameters and improved surgical outcomes.
radical prostatectomy for clinically localized prostate
The treatment outcomes following radical prostatectomy cancer. J Urol 2003; 169:517–523.
will most likely continue to improve as active screening 14. Kattan MW, Zelefsky MJ, Kupelian PA, et al:
for prostate cancer is expanded in the future. Pretreatment nomogram for predicting the outcome of
three-dimensional conformal radiotherapy in prostate
cancer. J Clin Oncol 2000; 18:3352–3359.
REFERENCES 15. Dalton DP, Schaeffer AJ, Garnett JE, Grayhack JT:
Radiographic assessment of the vesicourethral anastomosis
1. Catalona WJ, Smith DS, Ratliff TL, Basler JW: Detection directing early decatheterization following nerve-sparing
of organ-confined prostate cancer is increased through radical retropubic prostatectomy. J Urol 1989; 141:79–81.
prostate-specific antigen-based screening. JAMA 1993; 16. Lepor H, Nieder AM, Fraiman MC: Early removal of
270:948–954. urinary catheter after radical retropubic prostatectomy is
2. Catalona WJ, Smith DS, Ratliff TL, et al: Measurement both feasible and desirable. Urology 2001; 58:425–429.
of prostate-specific antigen in serum as a screening test 17. Pound CR, Christens-Barry OW, Gurganus RT, Partin
for prostate cancer. N Engl J Med 1991; 324:1156–1161. AW, Walsh PC:: Digital rectal examination and imaging
3. Walsh PC, Donker PJ: Impotence following radical studies are unnecessary in men with undetectable prostate
prostatectomy: insight into etiology and prevention. specific antigen following radical prostatectomy. J Urol
J Urol 1982; 128:492–497. 1999; 162:1337–1340.
4. Stanford JL, Stephenson RA, Coyle LM, et al: Prostate 18. Roehl KA, Antenor JA, Catalona W: Cancer recurrence
Cancer Trends 1973–1995, SEER Program. National and survival rates after anatomic radical retropubic
Cancer Institute, NIH, Bethesda, 1999. prostatectomy in 3032 consecutive patients. J Urol 2002;
5. Holmberg L, Bill-Axelson A, Helgesen F, et al: A 167 (Suppl 4):1364.
randomized trial comparing radical prostatectomy with 19. Catalona W, Roehl KA, Antenor JA: Potency, continence,
watchful waiting in early prostate cancer. N Engl J Med complications, and survival analysis in 3032 consecutive
2002; 347:781–789. radical retropubic prostatectomies. J Urol 2002; 167
6. Han M, Partin AW, Pound CR, Epstein JI, Walsh PC: (Suppl 4):625.
Long-term biochemical disease-free and cancer-specific 20. Mettlin CJ, Murphy GP, Sylvester J, et al: Results of
survival following anatomic radical retropubic hospital cancer registry surveys by the American College
prostatectomy. The 15-year Johns Hopkins experience. of Surgeons: outcomes of prostate cancer treatment by
Urol Clin North Am 2001; 28:555–565. radical prostatectomy. Cancer 1997; 80:1875–1881.
7. Hull GW, Rabbani F, Abbas F, et al: Cancer control with 21. Kirdi N, Yakut E, Meric A, Ayhan A: Physical therapy in a
radical prostatectomy alone in 1000 consecutive patients. patient with bilateral obturator nerve paralysis after surgery.
J Urol 2002; 167:528–534. A case report. Clin Exp Obstet Gynecol 2000; 27:59–60.
8. Arias E: United States life tables, 2000. Natl Vital Stat 22. Millesi H: Techniques for nerve grafting. Hand Clin
Rep 2002; 51:1–38. 2000; 16:73–91, viii.
9. Han M, Partin AW, Piantadosi S, Epstein JI, Walsh PC: 23. Lepor H, Nieder AM, Ferrandino MN: Intraoperative
Era specific biochemical recurrence-free survival and postoperative complications of radical retropubic
following radical prostatectomy for clinically localized prostatectomy in a consecutive series of 1000 cases. J Urol
prostate cancer. J Urol 2001; 166:416–419. 2001; 166:1729–1733.
10. Partin AW, Kattan MW, Subong EN, et al: Combination 24. Bukowski TP, Chakrabarty A, Powell IJ, et al: Acquired
of prostate-specific antigen, clinical stage, and Gleason rectourethral fistula: methods of repair. J Urol 1995;
score to predict pathological stage of localized prostate 153:730–733.
cancer. A multi-institutional update. JAMA 1997; 25. Prasad ML, Nelson R, Hambrick E, Abcarian H: York
277:1445–1451 [see comments, published erratum appears Mason procedure for repair of postoperative
in JAMA 9 July, 1997; 278(2):118]. rectoprostatic urethral fistula. Dis Colon Rectum 1983;
11. Partin AW, Mangold LA, Lamm DM, et al: 26:716–720.
Contemporary update of prostate cancer staging 26. Wood TW, Middleton RG: Single-stage transrectal
nomograms (Partin Tables) for the new millennium. transsphincteric (modified York-Mason) repair of
Urology 2001; 58:843–848. rectourinary fistulas. Urology 1990; 35:27–30.
C H A P T E R

30Radical Perineal Prostatectomy


Philipp Dahm, MD, Johannes Vieweg, MD,
and David F. Paulson, MD

Modern radical perineal prostatectomy (RPP) traces its


PATIENT SELECTION
origins back to 1904, when Dr. Hugh Hampton Young
performed the first prostatectomy to treat prostate can- The main indication for RPP is the primary treatment of
cer using the perineal approach. Today, RPP remains an localized adenocarcinoma of the prostate, and many con-
attractive modality to treat localized prostate cancer, hav- temporary patients of the prostate-specific antigen (PSA)
ing retained its distinct advantage over alternative surgi- era with newly diagnosed prostate cancer are excellent RPP
cal approaches: It provides the most direct access to the candidates. Modern staging algorithms, based on clinical
prostate gland. This characteristic is reflected in the low stage, serum PSA level, and Gleason score, allow an accu-
perioperative morbidity of this approach, which has led rate prediction of local extent and the likelihood of regional
some to perform RPP as outpatient surgery.1 Meanwhile, lymph node spread, thereby obviating the need for a staging
its long-term efficacy in treating clinically organ-confined node dissection in most patients. A bilateral pelvic lymph
disease has been established in large patients series with node dissection (BPLND) is recommended in patients with
extended follow-up of over 20 years.2–5 Compared to poorly differentiated disease Gleason scores of 8 to 10. In
radical retropubic prostatectomy (RRP), RPP allows pro- addition, patients with a PSA ≥ 20 ng/ml should undergo a
static dissection in a relatively avascular surgical field, BPLND. According to patients’ and surgeons’ preferences,
providing good exposure for reconstruction of the ure- BPLND may be performed through an open minilaparo-
throvesical anastomosis and allowing dependent postop- tomy or by laparoscopic technique. Either approach may be
erative drainage of the prostatic fossa. Modifications of performed as a separate procedure, yet more commonly
the classic RPP allow sparing of the neurovascular bundles BPLND is completed immediately before RPP under the
to preserve potency with equal efficacy as in RRP.6,7 If same anesthesia and frozen sections of the sampled nodes
capsular invasion is suspected, wide field dissection of are analyzed during the time of repositioning.
the prostate permits the removal of the prostate with the The perineal approach has few contraindications, the
same margin of extracapsular tissue as RRP.8,9 While the most important being the inability of the patients to
inability to perform a simultaneous staging pelvic assume an exaggerated lithotomy position due to condi-
node dissection was previously considered a major disad- tions, such as spinal stenosis, hip ankylosis, or a history of
vantage of the perineal approach, the recent trend vertebral fractures. If vertebral mobility is in question,
towards detection of prostate cancer at earlier stages the position may be readily simulated at the time of the
combined with the availability of accurate staging nomo- preoperative office visit. While obese patients may bene-
grams have made staging lymph node dissections unnec- fit from a perineal approach by avoiding the dissection of
essary in most patients.10–12 These features make modern deep layers of abdominal adipose tissue, a subset of mas-
RPP an attractive treatment choice for patients with sively obese patients with a “large barrel abdomen” may
organ-confined prostate cancer that should be offered to not be suitable candidates because of the high ventilatory
patients as part of an individualized, rational approach to pressures (in excess of 40 mm Hg) required during gen-
the surgical management of localized disease. Since the eral anesthesia. These patients should be counseled about
resurgence of the perineal approach will likely continue, the possibility that RPP cannot be performed for the
it appears prudent for contemporary urologists to famil- above reasons and alternative treatment forms should be
iarize themselves with this procedure. discussed beforehand.

528
Chapter 30 Radical Perineal Prostatectomy 529

While a large prostate (greater than 80 or 100 g) has blunt dissection. The dorsal venous complex is not
been cited as a relative contraindication for the perineal encountered and the urethra may be divided with preci-
approach, it is the relative size of the prostate to the dis- sion in a relatively avascular field. A Young retractor or
tance between the ischial tuberosities that determines the straight Lowsley prostatic retractor aids in further dis-
width of the surgical field. A digital rectal examination section of the posterior and the lateral aspects of the
(DRE) in the office prior to surgery provides a good prostate. It is important to adequately dissect free the
estimate of the spatial proportions and the feasibility anterior bladder neck so that a tension-free vesi-
of RPP in very large prostates but it requires some clini- courethral anastomosis can later be achieved. The
cal experience for making this judgment. Transrectal prostatovesical junction is identified and the prostate is
ultrasound examinations have not been helpful in our sharply cut away from the bladder. After sharp entry into
experience. the bladder between 10 o’clock and 2 o’clock, the Young
retractor is withdrawn and a Foley catheter is passed
through between the prostate and the bladder neck to
SURGICAL TECHNIQUE
serve as a retractor device. Intravenous indigo-carmine
Patient Preparation and Positioning
may be administered to aid in visualization of the ureteral
RPP is routinely performed after same day admission. orifices as the prostate is dissected away from the circular
Due to the proximity of the anus and rectum to the inci- detrusor fibers. Finally, the posterolateral pedicles, vas
sion line and plane of dissection, particular emphasis is deferens, and seminal vesicles are isolated, clipped, and
placed on a thorough mechanical and antibiotic bowel transected. Complete excision of the seminal vesicles is
preparation at home. The patient is placed in an exag- readily achieved 13 in most patients; however, is felt to
gerated lithotomy position taking care to pad all extrem- rarely benefit the patient from an oncologic perspective.
ities well and avoid overextension. The perineum is
correctly positioned by bringing the sacrum downwards
Bladder Neck Reconstruction and Urethrovesical
on the table until the buttocks reach approximately
Anastomosis
15 cm beyond the edge. A bolster is placed below the
sacrum to further elevate the pelvis. Correct positioning Once hemostasis is established, the bladder neck is
places the perineum parallel to the floor to provide opti- reconstructed. The bladder mucosa is averted to mini-
mal exposure. Following a routine preparation of a wide mize the risk of stricture formation, and the bladder neck
surgical field, the patient is draped in such a way that a is approximated in a racket-handle fashion. Two of the
DRE is possible at the beginning of the case to reorient racket-handle sutures and two additional sutures tied to
the surgeon to the individual anatomy and confirm cor- the bladder neck at 10 o’clock and 2 o’clock position
rect placement of the prostate retractor. The anus is then serve as Vest sutures to align the urethrovesical anasto-
covered by an impermeable drape and excluded from the mosis and are later brought out through the perineal
sterile surgical field for the remainder of the case. body and tied. Four anastomotic sutures are routinely
used that are placed and tied under direct vision over an
18 French urinary catheter to establish a watertight anas-
Access to the Prostate
tomosis. The operating field is copiously irrigated and a
Skin incision is made approximately 1.5 cm above the DRE is performed to rule out rectal injury. A soft drain
anal verge and extended posterolaterally on either side, is placed and the fibers of the rectourethralis, levator ani,
medial to the ischial tuberosities. The central tendon is and central tendon are approximated thereby obliterating
identified and divided. An anterior retractor is then any dead space and restoring the anatomy, followed by
placed and the rectourethralis muscle identified as a closure of the skin.
fibromuscular band in the midline, which is partially
swept to the side and partially divided. At this point, the
Postoperative Care
prostate is separated from the posterior surface of the
rectum in the midline by gentle, blunt dissection. A moist From the operating room the patient is taken to the
gauze is placed over the rectum to protect it from inad- recovery room, where he is typically monitored for
vertent injury and displacement is maintained by a approximately 2 hours to ensure appropriate recovery
retractor blade. from general anesthesia, adequate urinary drainage, and
good pain control. Since the average blood-loss in RPP is
low and averages at <500 cc, postoperative transfusions
Dissection of the Prostate
are a rare event. Oral analgesics are started in the recov-
Classic perineal prostatectomy proceeds with a transverse ery room. Patients will routinely tolerate a clear liquid
incision of Denonvilliers’ fascia followed by exposure of diet the evening of surgery and advance to their regular
the apex of the prostate using a combination of sharp and preoperative diet on the morning of postoperative day 1.
530 Part V Prostate Gland and Seminal Vesicles

The drain is left in place until postoperative day 2 or the carried so close to the ureteral orifices, either for onco-
time of the patient’s first bowel movement. At that time logic reasons or inadvertently. In this case, the ureteral
the patient is instructed to either take an antiseptic sitz orifices can be stented using open-ended ureteral
bath or alternatively to clean himself using a hand-held catheters followed by bladder neck closure from 6 o’
shower. Criteria for patient discharge are the ability to clock to 12 o’clock of the detrusor musculature only, not
tolerate a regular diet, ambulate without assistance, as incorporating the bladder mucosa. The stents are
well as good pain control with oral analgesics. These cri- brought out separately, secured and typically left in place
teria are normally met towards the end of postoperative for 5 to 7 days. Alternatively, the ureteral orifice may be
day 1 and patients are typically discharged home by the reimplanted through the perineal incision. The limited
morning of postoperative day 2. exposure, however, makes this maneuver technically
challenging. Should primary ureteral reimplantation not
be feasible, patients may alternatively be managed by
MODIFICATIONS OF CLASSIC RPP
placing a percutaneous nephrostomy tube on postopera-
Nerve-Sparing Radical Perineal Prostatectomy
tive day 1. This delay will allow for some degree of dilata-
Preservation of the neurovascular bundles is as feasible in tion of the collecting system to occur which facilitates
RPP as in RRP, yet requires an intimate understanding of tube placement. Ureteral reimplantation may then be
the course of the neurovascular bundles as viewed from performed in a delayed fashion using an abdominal
the perineum to prevent unintended injury. The proce- approach, ideally at 6 to 12 weeks postoperatively.
dure differs in that initially a vertical instead of a trans-
verse incision of the posterior layer of Denonvilliers’
Rectal Injury
fascia is made. Reflection of this layer over the apex of
the prostate gland and laterally, allows establishing a Rectal injury is a well-recognized risk of radical prostate
plane between the prostate and the neurovascular bun- surgery. It is a potentially concerning, but rare complica-
dles. Posterolateral dissection of the prostate is kept close tion of the perineal approach, with an incidence of 1% to
to the prostatic substance and small clips rather than 3% in the hands of experienced surgeons.9 The risk of
electrocautery are used to achieve hemostasis. Finally, rectal injury in RPP relates to the intimate relationship of
care is taken not to inadvertently injure the neurovascu- the plane of initial dissection to the rectum and most
lar bundles, which course in close proximity, when dis- commonly occurs either at the time of dissection of the
secting out the tips of the seminal vesicles. rectourethralis muscle or during placement of the poste-
rior weighted retractor. With strict adherence to the cor-
rect surgical technique and careful placement of the
Wide-Field Dissection Radical Perineal
retractors, however, injury appears avoidable in most
Prostatectomy
cases. If rectal injury does occur, prompt recognition and
Patients at high risk for extracapsular extension may be appropriate management may minimize subsequent mor-
considered for a wide-field dissection of the prostate, bidity to the patient. This should consist in copious irri-
which encompasses the lateral pelvic fascia and sacrifices gation of the surgical field followed by primary closure of
both neurovascular bundles. The prostate is exposed as the defect in several layers using an absorbable monofil-
described. However, after the rectum is displaced poste- ament. A diverting colostomy is rarely necessary, unless
riorly, a surgical plane is developed between the posterior gross fecal soiling is present, which is not encountered
layers of Denonvilliers’ fascia, the anterior rectal surface if the patient has been adequately prepared using both
posteriorly and the levator musculature anteriorly. The mechanical and antibiotic bowel preparations. Post-
prostate remains surrounded by the periprostatic lateral operative management includes the use of broad-spectrum
pelvic fascia. All fibrovascular pedicles are divided as far antibiotics and maintaining patients on a clear liquid diet
away from the prostate as possible. The seminal vesicles for 3 days. Placement of a rectal tube is unnecessary. The
are dissected as described but clipped and dissected at outlined management usually results in uncomplicated
such a level to include the neurovascular bundle. healing of the rectal injury with no adverse consequences
Additionally, wider margins of the bladder neck may be to the patient.
taken to achieve negative margins followed by recon-
struction as described.
Lower Extremity Neuropraxia
Lower extremity neuropraxia is a unique problem associ-
COMPLICATIONS OF RPP
ated with the exaggerated lithotomy position. While it
Ureteral Injury
is relatively common in the immediate postoperative
Problems in reconstruction of the bladder neck may setting occurring in up to 25% of patients, symptoms are
occur if the posterior margin of the incision has been usually transient and consist of sensory deficits below the
Chapter 30 Radical Perineal Prostatectomy 531

knee only.14 These symptoms invariably resolve, in most prostate cancer. The Duke Radical Perineal Prostatectomy
cases prior to the patients discharge from the hospital on Database represents one of the largest series of radical
postoperative day 2. The etiology of these injuries is prostatectomy patients from a single institution and is
mostly attributable to a stretch injury to the sciatic or based solely on RPP patients. A large cohort of patients
common peroneal nerve, less often due to direct com- who underwent RPP at our institution have been fol-
pression injury to the sensory nerves of ankle of foot. lowed for clinical, biochemical, and radiographic evi-
The recognition of the mechanisms of injury, often dences of recurrent disease for an extended period of
resulting from a lack of awareness by inexperienced oper- time of up to 20 years, offering the unique opportunity
ating room personnel has resulted in a significant reduc- to study the long-term outcome of patients that chose
tion of this problem at our institution. In addition, a this approach as treatment for clinically localized prostate
recent study from our institution suggests that the cancer. Outcome is best described as time to cancer-asso-
likelihood of postoperative lower extremity neuropraxia ciated death, which is used for any death associated with
correlates with surgery time; patients with a surgery recurrent disease irrespective of treatment as docu-
time of greater than 2 hours were twice as likely to mented by a rising PSA level of ≥0.5. In addition, patho-
report symptoms than patients who’s procedure was logic staging using the categories “organ-confined”
completed in less time. Based on these findings it appears (OC), “specimen-confined” (SC), and “margin positive”
critical to minimize the amount of time patients spend in (MP) provides a robust classification system that has
the exaggerated lithotomy position. If neuropraxia does withstood the many changes of the TNM system over
occur, management is expectant and conservative. the last 2 decades, yet carries important prognostic
Further diagnostic work-up is usually not indicated implications.
unless neurologic symptoms are progressive, suggesting a The overall 10- and 15-year cancer-associated survival
different etiology, such as spinal stenosis or a herniated rates for all RPP candidates in this series (n = 2006) are
disk. 84.5 ± 1.4% and 71.7 ± 2.3%, respectively. Figure 30-1
illustrates the strong prognostic impact of local extent;
15-year cancer-associated survival rates for patients
DISEASE CONTROL
with organ-confined, specimen and margin positive dis-
RPP has been demonstrated to achieve long-term disease ease are 86.6 ± 2.5, 70.7 ± 6.5, and 49.8 ± 4.4, respec-
control in patients with clinically organ-confined tively. In addition, the Gleason score of the prostatectomy

10

.9
OC (n = 1179)
.8
Probability of cancer-associated survival

.7

SC (n = 399)
.6 p = 0.001

.5

.4
MP (n = 428)
.3

.2

.1

0.0
0 2 4 6 8 10 12 14 16 18 20 Follow-up (years)
2006 1222 884 249 106 35 Patients at risk

Figure 30-1 Kaplan-Meier analysis of cancer-associated survival of patients following RPP


(n = 2006) grouped by tumor extent, applying the categories organ-confined (OC),
specimen-confined (SC), and margin-positive (MP).
532 Part V Prostate Gland and Seminal Vesicles

specimen, as a marker for the biologic aggressiveness depending on the type of definition used.5,18 Urinary
of the tumor, has consistently demonstrated an impact continence is typically achieved early in the postoperative
on long-term survival15 as illustrated in Figure 30-2. course; Weldon et al.18 reported a return of continence
Fifteen-year cancer-associated survival rates for (defined as “no routine pad use”) in 23% of patients at 1
patients with Gleason score 5 to 6, 7, and 8 to 10 are month, 56% by 3 months, and 90% by 6 months. It has
81.1 ± 3.4%, 67.3 ± 4.6%, and 49.2 ± 6.1%, respectively. furthermore been suggested that patients may seek to
When combining both prognostic variables one finds minimize adverse outcomes when discussing them with
that patients with the most unfavorable combination their surgeon, and physician and patient assessment of uri-
of margin positive, high Gleason score disease of 8 to nary symptoms following radical prostatectomy may differ
10 (n = 146) experience extended survival periods with significantly.19,20 Thus, more recent investigations have
10- and 15-year cancer-associated survival rates of utilized validated instruments that are self-administered
43.1 ± 6.8% and 31.3 ± 8.2%, respectively. These find- to more comprehensively characterize urinary function
ings emphasize the need for long follow-up periods and minimize potential bias.21,22 In a prospective study at
when studying prostate cancer and suggest that many our institution of 78 RPP candidates that underwent
current series of alternative curative therapies lack RPP in a standardized technique and applying a defini-
follow-up of sufficient duration to permit comparison tion of urinary incontinence of greater than “occasional
with the results of radical surgery. While the use of dif- dribbling” we found a proportion of patients self-
ferent staging systems and endpoints of outcome cloud a reporting urinary incontinence of 35.9%, 15.4%, 5.1%,
direct comparison between RPP and RRP, the disease and 3.7% after 3, 6, 9, and 12 months, respectively.
control achieved by RPP and RRP may be considered Meanwhile, 5.6% of patients had some degree of preex-
equivalent.16,17 isting urinary incontinence when questioned prior to sur-
gery and applying the same definition. These results may
be regarded as preliminary, based on a small cohort of
URINARY CONTINENCE
patients. Nevertheless, they compare favorably with the
Urinary continence is one of the most significant out- reported self-assessed urinary incontinence rates
come parameters of radical surgery for prostate cancer reported in a series of RRP patients reported by Wei et al.22
and perineal prostatectomy achieves excellent urinary and document the early and consistent return of urinary
continence rates, ranging between 92% and 96%, continence following RPP in a majority of patients.

10
GS 2−4 (n = 85)
.9

.8 GS 5−6 (n = 888)
Probability of cancer-associated survival

.7

.6 p = 0.001
GS 7 (n = 704)
.5

.4
GS 8−10 (n = 292)
.3

.2

.1

0.0
0 2 4 6 8 10 12 14 16 18 20 Follow-up (years)
1969 1222 884 249 106 35 Patients at risk

Figure 30-2 Kaplan-Meier analysis of cancer-associated survival of patients following RPP


(n = 1969) grouped by Gleason score (GS), applying the GS categories 2 to 4, 5 to 6, 7,
and 8 to 10.
Chapter 30 Radical Perineal Prostatectomy 533

unexpected high prevalence of occasional fecal inconti-


POTENCY
nence symptoms in 11.5% of patients (9/78) prior to sur-
The nerve-sparing modification of RPP may be regarded gery. Of these 9 patients, 3 suffered from involuntary
as equally effective as the retropubic approach6,7,23 and is stool leakage once a week or less frequently, yet 6 patients
offered to select patients with favorable preoperative experienced this symptom at least once daily. Meanwhile,
staging characteristics. These include patients with clini- only 2 patients (2.6%) identified fecal incontinence as
cally organ-confined disease, a serum PSA ≤ 10 ng/ml greater than a “very small problem.” All bowel-related
and a Gleason score ≤ 6. It has been our personal prac- symptoms, including involuntary stool leakage, were
tice to offer the bilateral nerve-sparing approach only to transiently increased 1 month following RPP yet
those patients with a single positive biopsy core. returned to baseline levels within 3 to 6 months postop-
Therefore, most patients undergo a unilateral nerve- eratively. Compared to their individual baseline, 15.4%,
sparing RPP with deliberate sacrifice of the neurovascu- 7.7%, 5.1%, and 3.9% of patients reported worsened
lar bundle on the side where biopsies are positive, symptoms of fecal incontinence after 3, 6, 9, and 12
suggesting the location of the bulk of the tumor. When months, respectively (Figure 30-3). When analyzing the
reviewing the potency rates following RPP at our institu- rate of fecal incontinence in patients who denied preop-
tion, Frazier et al.6 found 77% of patients to be potent 1- erative problems (n = 69) the incidence after 12 months
year after nerve-sparing RPP. Weldon et al.18 was only 2.9%, which is considerably lower than the
additionally analyzed the time course to recovery of range of 15% to 18% previously reported by Bishoff et
potency and found that potency returned in 50% of al.24 Potential explanations for this discrepancy include
patients after 1 year and in 70% after 2 years. An addi- differences in the patient populations, as well as a recall
tional, important observation was the strong inverse bias in the previous study, resulting in the inadvertent
correlation of the rate of successful potency sparing and inclusion of patients with some degree of preexisting fecal
increasing age. Potency returned in all patients <50 years incontinence that was not accounted for retrospectively.
of age but only 29% of older patients. It appears impor- Finally, the patients in the study reported by Bishoff et
tant to emphasize these two aspects pertaining to the al.,24 underwent surgery at least 1 year prior to the time
recovery of potency—the extended time interval neces- they were questioned. While our results give us no indi-
sary and the decreased potential for functional recovery cation that fecal incontinence initially improves and then
with increasing age—with the patient prior to surgery to subsequently deteriorates, information about the long-
generate realistic expectations with regard to the level of term outcome of the patients remains pending. Longer
postoperative sexual functioning that may be anticipated. follow-up of our cohort of patients may allow us to char-
acterize the incidence of fecal incontinence further.
FECAL INCONTINENCE
QUALITY OF LIFE FOLLOWING RPP
A recent study suggested that RPP is associated with a
high risk of postoperative fecal incontinence.24 These Quality of life is being increasingly recognized as an
findings were based on a retrospective study of two important complementary endpoint, when considering
groups of patients—from the authors’ institution and treatment options for prostate cancer. A small number of
from a nationwide database. Patients in either group well-designed studies addressing this issue have been
denied any preexisting preoperative fecal incontinence, reported that provide longitudinal rather than cross-sec-
yet reported fecal incontinence rates of 18% and 15%, tional observations and include a measurement of quality
respectively, at least 1 year following RPP. A particular of life before initiation of therapy, allowing each patient
concerning observation of this study was that a majority to serve as his own control. Litwin et al.25 investigated
of patients did not seek medical assistance for the prob- the recovery of general and prostate-specific quality of
lem of involuntary stool leakage despite detrimental life in 247 men following RRP. The authors found that
effects to their quality of life. The suggestion that fecal 21% of patients reached their individual baseline level of
incontinence may be common after RPP, yet under- HRQOL in the urinary domain within 3 months follow-
appreciated because it is not routinely addressed during ing surgery, and 56% by 12 months. In the bowel
follow-up, prompted us to design a study to evaluate the domain, over two-thirds of patients recovered their base-
incidence of bowel-related symptoms and complaints, line scores by 3 months, and >90% recovered by 12
and fecal incontinence in particular, following RPP. months. Meanwhile, sexual function and bother took
Using a validated patient self-assessment tool we per- longer to return, and was recovered by 33% and 51% of
formed a longitudinal assessment of bowel function and patients within 1 year, respectively.
bowel-related bother of 78 consecutive patients undergo- To our knowledge, no dedicated investigation of the
ing RPP, while accounting for preoperatively determined recovery of HRQOL in RPP patients has been pub-
baseline characteristics for comparison. We found an lished, yet preliminary data from a cohort of 78 patients
534 Part V Prostate Gland and Seminal Vesicles

10

.9

.8

.7
Probability of stool leakage
.6

.5

.4

.3

.2

.1

0.0
0 1 2 3 4 5 6 7 8 9 10 11 12
Time after RPP (months)

Figure 30-3 Kaplan-Meier analysis of the probability of patients (n = 78) to experience


worsening symptoms of involuntary stool leakage compared to their individual preoperative
status, based on responses to a validated patient self-assessment instrument, the EPIC,
completed by patients prior to surgery, at 4 weeks, 3, 6, 9, and 12 months following RPP.

who underwent RPP at our institution with a minimum number of patients, these results compare favorably with
follow-up of 6 months have recently become available. the available RRP data, particularly with respect to the
The patients have been studied longitudinally prior to urinary domain.
surgery, after 1 month and subsequently at 3-month
intervals following RPP using a validated patient self-
SUMMARY
assessment tool, the Expanded Prostate Cancer Index
Composite (EPIC).21 The mean age was 60.3 years Modern RPP constitutes an excellent treatment modality
(range 43 to 78) and the majority (78.2%) of patients for select patients with clinically organ-confined prostate
were treated using a non nerve-sparing approach. Of cancer. Features of this approach include documented
these patients, 47.1% and 81.7% recovered their individ- long-term disease control, favorable functional results,
ual urinary and bowel-related HRQOL within 3 months low morbidity, and fast patient recovery. These charac-
following RPP (Table 30-1). While based on a small teristics make RPP an attractive treatment choice that

Table 30-1 Longitudinal Assessment of the Recovery of Health-related Quality of Life (HRQOL) Scores
in the Domains Urinary, Bowel, and Sexual within 12 Months Following RPP
1 Month (n = 78) 3 Months (n = 71) 6 Months (n = 78) 9 Months (n = 66) 12 Months (n = 64)

Urinary (%) 19.5 47.1 71.8 78.8 82.8

Bowel (%) 35.1 81.7 92.3 90.9 90.6

Sexual (%) 19.4 20.6 20.3 24.6 23.3

Patients were assessed prior to surgery, after 4 weeks, and subsequently at 3 months intervals using the EPIC, a validated patient self-assessment
tool. HRQOL scores of each patient were calculated on a scale of 0 to 100 for every domain. Patients were considered to have recovered their
individual level of HRQOL if they reached a score within ±10 points of their baseline value.
Chapter 30 Radical Perineal Prostatectomy 535

should be offered to patients as part of an individualized, 13. Vordos D, Delmas V, Hermieu JF, et al: Can a precise
rational approach to the surgical management of local- vesiculectomy be performed during radical prostatectomy?
ized disease. Prog Urol 2001; 11:1259.
14. Price DT, Vieweg J, Roland F, et al: Transient lower
extremity neurapraxia associated with radical perineal
REFERENCES prostatectomy: a complication of the exaggerated
lithotomy position. J Urol 1998; 160:1376.
15. Iselin CE, Box JW, Vollmer RT, et al: Surgical control of
1. Ruiz-Deya G, Davis R, Srivastav SK, Wise, AM, Thomas
clinically localized prostate carcinoma is equivalent in
R: Outpatient radical prostatectomy: impact of standard
African-American and white males. Cancer 1998; 83:2353.
perineal approach on patient outcome. J Urol 2001;
16. Sullivan LD, Weir MJ, Kinahan JF, Taylor DL: A
166:581.
comparison of the relative merits of radical perineal and
2. Belt E, Schroeder FH: Total perineal prostatectomy for
radical retropubic prostatectomy. BJU Int 2000; 85:95.
carcinoma of the prostate. J Urol 1972; 107:91.
17. Lance RS, Freidrichs PA, Kane C, et al: A comparison of
3. Iselin CE, Robertson JE, Paulson DF: Radical perineal
radical retropubic with perineal prostatectomy for
prostatectomy: oncological outcome during a 20-year
localized prostate cancer within the Uniformed Services
period. J Urol 1999; 161:163.
Urology Research Group. BJU Int 2001; 87:61.
4. Paulson DF, Robertson JE, Daubert LM, Walther PJ:
18. Weldon VE, Tavel FR, Neuwirth H: Continence, potency
Radical prostatectomy in stage A prostatic
and morbidity after radical perineal prostatectomy. J Urol
adenocarcinoma. J Urol 1988; 140:535.
1997; 158:1470.
5. Gibbons RP: Total prostatectomy for clinically localized
19. Gray M, Petroni GR, Theodorescu D: Urinary function
prostate cancer: long-term surgical results and current
after radical prostatectomy: a comparison of the
morbidity. NCI Monographs 1988; 123.
retropubic and perineal approaches. Urology 1999;
6. Frazier HA, Robertson JE, Paulson DF: Radical
53:881.
prostatectomy: the pros and cons of the perineal versus
20. Wei JT, Montie JE: Comparison of patients and
retropubic approach. J Urol 1992; 147:888.
physicians rating of urinary incontinence following radical
7. Weldon VE, Tavel FR: Potency-sparing radical perineal
prostatectomy. Semin Urol Oncol 2000; 18:76.
prostatectomy: anatomy, surgical technique and initial
21. Wei JT, Dunn RL, Litwin MS, Sandler HM, Sanda MG:
results. J Urol 1988; 140:559.
Development and validation of the expanded prostate
8. Korman HJ, Leu PB, Huang RR, Goldstein NS:
cancer index composite (EPIC) for comprehensive
A centralized comparison of radical perineal and
assessment of health-related quality of life in men with
retropubic prostatectomy specimens: is there a difference
prostate cancer. Urology 2000; 56:899.
according to the surgical approach? J Urol 2002; 168:991.
22. Wei JT, Dunn RL, Marcovich R, Montie JE, Sanda MG:
9. Thrasher JB, Paulson DF: Reappraisal of radical perineal
Prospective assessment of patient reported urinary
prostatectomy. Eur Urol 1992; 22:1.
continence after radical prostatectomy. J Urol 2000;
10. Gingrich JR, Paulson DF: The impact of PSA on prostate
164:744.
cancer management. Can we abandon routine staging
23. Weldon VE: Technique of modern radical perineal
pelvic lymphadenectomy? Surg Oncol Clin N Am 1995;
prostatectomy. Urology 2002; 60:689.
4:335.
24. Bishoff JT, Motley G, Optenberg SA, et al: Incidence of
11. Partin AW, Mangold LA, Lamm DM, et al:
fecal and urinary incontinence following radical perineal
Contemporary update of prostate cancer staging
and retropubic prostatectomy in a national population.
nomograms (Partin tables) for the new millennium.
J Urol 1998; 160:454.
Urology 2001; 58: 843.
25. Litwin MS, Melmed GY, Nakazon T: Life after radical
12. Parra RO, Isorna S, Garcia PM, Cummings JM, Boullier JA:
prostatectomy: a longitudinal study. J Urol 2001; 166:587.
Radical perineal prostatectomy without pelvic
lymphadenectomy: selection criteria and early results.
J Urol 1996; 155:612.
C H A P T E R

31Laparoscopic Radical Prostatectomy


Guy Vallancien, MD, and Xavier Cathelineau, MD

HISTORY
motivated teams, composed of instrument nurses, scrub
Laparoscopic surgery, which was developed in gynecol- nurses, anesthetists, and the hospital administration. It
ogy during the 1940s, was used in gastrointestinal would be impossible to perform laparoscopic surgery
surgery from 1986 onward, especially following develop- without such an environment.
ment of the technique by Philippe Mouret in Lyon and
François Dubois in Paris. It was not, however, immedi-
INDICATIONS AND CONTRAINDICATIONS
ately extended to the field of urology. The first laparo-
Indications
scopic nephrectomy was performed in 1990 by Ralph
Clayman in St. Louis, but the urologic community The indications of laparoscopic radical prostatectomy are
remained reticent for a long time about the value of these exactly the same as the open radical prostatectomy. The
time-consuming techniques, which had only limited indi- development of the laparoscopic procedure has not
cations. From 1995 onward, the development of intra- changed the indications and the selection of the patient
corporeal suture, allowing repair of anomalies of the candidates to a surgical treatment.
ureteropelvic junction and nephrectomies for cancer, led The best indication is probably the young patient,
to a renewed interest in laparoscopy. whose PSA is <15 ng/ml, with less than 50% of positive
In 1992, the first attempt to perform laparoscopic biopsies and Gleason score <8.
prostatectomy in two cases was published by Schuessler Laparoscopic radical prostatectomy is feasible for
et al.1 in an abstract presented to the American Urology some selected T3N0M0, with condition not preserving
Association Congress. In 1997, the same team published the neurovascular bundles and informing the patient of
9 cases of laparoscopic radical prostatectomy and reached possible complementary treatment.
the conclusion that this technique did not provide any Salvage laparoscopic radical prostatectomy is also fea-
advantages over open surgery due to the duration and sible after radiation therapy or brachytherapy, but the
difficulty of the operation, especially when performing patient has to be informed of the risk of temporary
the vesicourethral anastomosis. colostomy.
In the same year, Raboy et al.2 published a case of
extraperitoneal radical prostatectomy. In December
Contraindications
1997, Richard Gaston (Bordeaux, France), in a personal
Anesthetic Contraindications
communication, indicated that he had performed a
transperitoneal radical prostatectomy in less than 6 There are no specific anesthetic contraindications for
hours. Six weeks later, Guillonneau and Vallancien3,4 laparoscopic radical prostatectomy. Contraindications
started to perform their first radical prostatectomies. Five for this procedure are the same as those for all laparo-
months later, Abbou,5 in Créteil, also started to perform scopic procedures.
this new surgical technique. The main absolute contraindication for laparoscopic
Laparoscopy requires appropriate instruments, an approach is high-intracranial pressure whatever its origin
intellectual investment (long training) and a physical (primary or secondary to intracranial process).
investment (this surgery is more tiresome than open sur- The relative anesthetic contraindications are: severe
gery). Most importantly, surgeons must be assisted by emphysema, severe cardiac injury, chronic respiratory

536
Chapter 31 Laparoscopic Radical Prostatectomy 537

disease, and glaucoma. Those contraindications have An exaggerated Trendelenburg position is essential, at
to be underlined specifically for extraperitoneal least for the initial posterior phase of the operation.
approach. Effectively, there is an increased partial The upper limbs are positioned alongside the body to
pressure of carbon dioxide (pCO2) during all the avoid the risk of stretch injuries to the brachial plexus.
extraperitoneal procedure, which requires increased An adhesive elastic bandage is placed in cross fashion
minute ventilation, in order to maintain a p CO 2 over the thorax, which ensures better comfort for the
between 30 and 35 mm Hg. patient than shoulder rests.
The surgeon stands to the left of the patient and the
assistant stands to the right. The camera is attached to a
Anatomic Contraindications
voice-controlled robotic arm.
There are no anatomic contraindications for laparo-
scopic radical prostatectomy.
Material
Obviously, some cases are more difficult, and especially
at the beginning of the experience, the surgeon has to take There is no specific instrument for laparoscopic radical
care of those difficulties and probably select the best cases. prostatectomy. As for all the others laparoscopic proce-
dures adapted material is primordial. All the instruments
had to be checked before the procedure, especially bipo-
Difficult Cases
lar forceps and scissors, in order to detect a leak, which
Different factors can increase the difficulty of the proce- could induce a burning of intraabdominal organ.
dure, especially:
Main Steps of the Operation
● Previous hormonotherapy
● Previous TURP or prostatectomy Insufflation is performed with a Veress needle in the
● Prostatitis umbilicus or left hypochondrium.
The 10-mm trocar for the scope is placed in the
All of these factors can modify the prostatic and umbilicus. Four other trocars are used for the surgeon
periprostatic tissues and make the surgery really difficult. and the assistant, arranged according to the surgeon’s
Moreover, very big prostate (more than 100 g) usual practice; either in a triangular pattern (the operator
increase the difficulty of the dissection. According to his uses a left pararectal trocar and a right pararectal trocar,
experience, the surgeon has probably to avoid those cases and the assistant uses a trocar in the right iliac fossa and
at the beginning of his learning curve. an interumbilico-pubic trocar) or in a parallel pattern
In any case, conversion is not a shame but a sign of (the operator uses a trocar in the left iliac fossa and a left
wisdom. pararectal trocar, and the assistant uses two symmetrical
trocars on the right side).
Pelvic lymph node dissection with frozen section
OPERATIVE TECHNIQUE
examination is performed according to the preoperative
Transperitoneal Approach: the Montsouris
findings.
Technique6
The operation comprises seven successive steps:
Medical Preparation
Prophylactic antibiotics are not given to the patient; they 1. Posterior phase. The posterior peritoneum is
do not decrease the risk of infection but can induce selec- incised over the vasa deferentes, which are
tion of bacteria, especially enterococci. sectioned, and the seminal vesicles are completely
Thromboembolic complications are prevented by dissected. The median part of Denonvilliers’ fascia
injection of low molecular weight heparin on the day is incised.
before the operation, which is continued for at least 7 2. Anterior phase. The anterior parietal peritoneum is
days postoperatively, associated with varicose veins stock- incised from one umbilical artery to the other
ing during hospitalization. providing access to the retropubic space after section
No gastrointestinal or skin preparation is required. of the urachus.
The pelvic fascia is incised as far as the
puboprostatic ligaments, which are sectioned. The
Installation of the Patient
prostatic apex is completely dissected. Santorini’s
The operation is performed under general anesthesia. venous plexus and the preprostatic venous drainage
The patient is placed in the dorsal supine position, are then ligated with 2/0 Vicryl (this stitch can also
with the lower limbs in abduction allowing intraoperative be placed after the dissection of prostatic vascular
access to the rectum. pedicles, just before cutting the uretra).
538 Part V Prostate Gland and Seminal Vesicles

3. Vesicoprostatic dissection is performed by preserving, as the vasa deferentes. The anastomosis is then performed
far as possible, the fibers of the bladder neck. with interrupted sutures.
Dissection of the posterior surface of the bladder Overall, this technical modification is designed to
neck provides access to the plane of the seminal reproduce the various steps of retropubic prostatectomy.
vesicles and vasa deferentes.
4. Dissection of prostatic pedicles and neurovascular bundles.
Number of Trocars
The prostatic vascular pedicles are selectively
coagulated with bipolar forceps. Neurovascular Most teams6,9,10 performing laparoscopic radical prosta-
bundles are preserved depending on anatomical and tectomy use 5 trocars, which may be arranged in differ-
oncological conditions. ent ways according to the surgeon’s usual practice.
5. Section of the urethra. The urethra is sectioned away Rassweiler et al.7,8 add a sixth 5-mm trocar at Mac
from the prostatic apex. The rectourethralis muscle is Burney’s point, which is designed to improve exposure,
also sectioned and the prostate is then completely especially during seminal vesicle dissection.
released (a bougie in the rectum can be useful to
identify the rectum in the case of difficult posterior
Use of an Arm for the Scope
dissection).
6. Vesicourethral anastomosis. It is performed by The use of an arm for the scope provides valuable assis-
interrupted 3/0 Vicryl sutures. An 18 F Foley tance when performing a laparoscopic procedure, such as
catheter is inserted. The absence of anastomotic leak radical prostatectomy, as it ensures a fixed image and
is verified intraoperatively. frees one of the assistant’s hands, allowing more active
7. Extraction of the operative specimen. The prostate is participation in the operative procedure.
placed in a laparoscopic bag and removed by enlarging Many teams6–10 use a voice-controlled robotic arm
the umbilical incision. A Redon drain is inserted in (AESOP), but other types of arms are also available, espe-
the retropubic space or the pouch of Douglas. The cially less expensive, manually guided, compressed air arms.
5-mm trocar orifices are closed in one layer and the
10-mm trocar orifices are closed in two layers.
Modalities of Dissection of Neurovascular
Bundles
Postoperative Management
Preservation of the neurovascular bundles precludes the
The bladder catheter is left in place for 3 to 7 days use of mechanical staplers for section of the superior pro-
depending on the quality of the suture (cystography is static pedicles. Control and section of these pedicles and
not performed). preservation of neurovascular bundles require precise
Analgesia is limited to paracetamol (8 g by intravenous dissection, ideally performed by selective bipolar coagu-
injection) during the first 24 hours, followed, on D1, by lation6,9,11 or even the use of clips.10 The choice between
oral paracetamol/dextropropoxyphene pro re nata (p.r.n.) these two approaches is a matter of personal preference,
Major analgesics are not administered routinely but are as neither technique has been shown to be superior.
prescribed p.r.n.
The IV infusion is stopped on D1.
Technical Modalities of Vesicourethral
Oral fluids are started on D1 and a normal diet can
Anastomosis
generally be resumed on D2.
The vesicourethral anastomosis can be performed with
interrupted sutures or a running suture. Whatever the
Variants of the Transperitoneal Approach
choice, the rules are the same and especially the necessity
Transperitoneal Approach With Initial Anterior
of using both hands with forehand and backhand.
Approach
The running suture has the advantage of being less
Rassweiler et al.7,8 reported their experience with a mod- time-consuming, but it requires constant, good quality
ified version of the Montsouris technique. They perform traction to ensure a perfectly leak proof suture. In every
an initial anterior approach providing access to the case, the operator’s experience and that of his team
retropubic space. The pelvic fascia is incised, the pubo- appear to be essential for both the rapidity and the qual-
prostatic ligaments are sectioned, and Santorini’s venous ity of the procedure.
plexus is ligated. The urethra is then sectioned, and when
the neurovascular bundles are preserved, they are dis-
Extraperitoneal Approach
sected in an ascending direction.
The vesicoprostatic dissection is then performed, In 1997, Raboy et al.2 described, for the first time, their
prior to dissection of the seminal vesicles and section of experience of extraperitoneal laparoscopic radical
Chapter 31 Laparoscopic Radical Prostatectomy 539

prostatectomy. In 2001, Bollens et al.12 reported their ● Dissection


experience of 46 cases performed according to this ● Extraction of surgical specimen and removal of
technique. trocars
The scope is introduced via a 10-mm trocar placed at ● Early postoperative phase
the inferior margin of the umbilicus. Four trocars are ● Late postoperative phase
used: two 10-mm trocars placed in the left iliac fossa and
right iliac fossa; a 5-mm trocar is introduced about 5 cm
Complications of Patient Positioning
above the pubic symphysis; and the last 5-mm trocar is
placed in the right flank. The main complications at this step are:
The first phase of the procedure consists of creation of
a prevesical working space. Bladder neck dissection is ● Compartment syndrome
performed prior to seminal vesicle dissection. The pelvic ● Peripheral neuropathy
fascia is then incised and Santorini’s venous plexus is lig- ● Scapular pain
ated. The neurovascular bundles are then dissected
before coagulation and section of the superior prostatic The risk is related to protections used for the posi-
pedicles. Denonvilliers’ fascia is then incised, releasing tioning and also, obviously, to the length time of the pro-
the posterior surface of the prostate. The apex is then cedure. So, the surgeon has always to check by himself
dissected and the urethra is sectioned. The anastomosis is the protections of the patients and to take care of the
performed by interrupted sutures. length time of the operation (conversion is sometimes
Overall, the advantages of the extraperitoneal the best solution).
approach, according to its supporters, would be a reduc- Those complications are exceptional (<0.1%), and
tion of the risk of damage to intraperitoneal organs and a their risk decrease significantly with experience of the
similar surgical approach to that of open retropubic surgeon and his team.
prostatectomy. Its disadvantages would be a limited
working space and the difficulty of the anastomosis due
Complications of Insufflation and Trocar
to the more limited mobilization of the bladder.
Placement
We reviewed 200 consecutive procedures performed
in Montsouris Institute by two surgeons; 100 transperi- The main risk is vascular or bowel injuries with the first
toneal procedures were compared to the first 100 trocar. There is no rule to use or not open laparoscopic
extraperitoneal cases. This study showed equivalent procedure and the choice depends on the usefulness and
results in term of operative, postoperative, and patholog- the experience of each team.
ical data. Small vessels injury can also occur during the place-
ment of other trocars, especially epigastric artery (0.3%).
PERIOPERATIVE COMPLICATIONS
Intraoperative Complications
Improvement of the surgical technique has allowed a
Blood Loss
reduction of the morbidity of radical prostatectomy.
Comparative studies of the complications according to Median intraoperative bleeding reported by experienced
the surgical technique are often difficult and must always teams performing retropubic prostatectomy varies
take into account not only the operator’s level of experi- between 1000 and 1500 ml.14,19 Dillioglugil19 reported a
ence with the technique but also the patient characteris- transfusion rate of 29% for all the series and 15% for the
tics and finally the modalities of evaluation of last 135 patients among a total of 472. For Catalona,14
complications, especially functional complications. using the technique of hemodilution, most of the patients
The morbidity is related not only to the technique received autologous transfusion during or after surgery
itself but also to the patient’s comorbidities, particularly and 9% need also heterologous postoperative transfu-
the ASA3 score13 and blood loss. sion. In the same series, the rate of hematoma was 0.05%.
The mortality rate is currently close to 0%, regardless The median blood loss during laparoscopic prostatec-
of the technique used (retropubic, perineal or laparo- tomy varies from 370 ml20 to 1230 ml11 for patient series
scopic)14–17 (Tables 31-1 and 31-2). with a comparable experience.11,20 The transfusion rates
Considering the different phase of the laparoscopic for these two series were 7% and 30%, respectively, at
procedure, the complications can be divided according the beginning of their experience.
each step: Guillonneau et al.17 reported, for the last 100
patients, a mean estimated blood loss of 290 ml and a
● Patient positioning transfusion rate of 3.5%. The rate of hematoma in this
● Insufflation and placement of trocars series was 1%.
540 Part V Prostate Gland and Seminal Vesicles

Table 31-1 Complications of Radical Prostatectomy


Catalona *Guillonneau *Rassweiler
Study Lepor et al.39 et al.14 (14) et al.17 et al.11

Number of patients 150 1000 1870 567 180

Rectal injury 6/0.6 3/2 5/0.5 1/0.05 8/1.4 1.1

Ileocolic injury nc nc nc nc 4/0.7 nc

Ileus nc nc 4/0.4 nc 6/1.1 2.8

Bladder injury nc nc nc nc 9/1.6 nc

Ureteral injury nc 7/4.7 1/0.1 1/0.05 3/0.5 0

Deep venous thrombosis 13/1.3 4/2.7 2/0.2 39/2 2/0.4 0

Pulmonary embolism 7/0.7 nc 4/0.4 nc 0 0

Anastomotic leakage nc 15/10 2/0.2 nc 46/8.1 2.2

Lymphocele 1/0.1 3/2 1/0.1 11/0.6 1/0.2 0

Hydronephrosis nc nc nc nc nc 2.8

Acute renal failure Nc 2/1.3 nc nc 1/0.2 nc

Pelvic hematomas 1/0.1 nc nc nc 5/0.9 8.9

Wound /0.9 nc 9/0.9 15/0.8 7/1.2 0.6

Nerve injury nc nc 1 5/0.3 3/0.5 0

Myocardiac infarction 7/0.7 0 5/0.5 2/0.1 0 nc

Cerebral vascular accident nc nc nc 0 0 nc

Death nc 1/0.7 1/0.1 0 0 nc

Anastomotic stricture nc nc 10/1 71/3.8 nc 2.8

Total complications 35/3.5 143/8 95/16.7 nc

*Laparoscopic approach.

Table 31-2 Evolution of the Complication Rate with Experience in Laparoscopy (Montsouris Experience)
Patients 1–200 Patients 200–400 Patients 400–567

Lymphadenectomy (%) 25 19 14

Mean operative time (minutes ) 234 190 180

Mean estimated blood loss (ml) 372 398 350

Transfusions (%) 7.5 5 3.8

Conversions (%) 3.5 0 0

Number rectal injuries 3 1 4

Reinterventions (%) 6 7 9
Chapter 31 Laparoscopic Radical Prostatectomy 541

Digestive Injuries Urinary Incontinence


Digestive injuris are rare and directly related to the oper- The quality of continence after radical prostatectomy is
ator’s experience and to the patient’s history (especially difficult to assess (Table 31-3), as reflected by the marked
previous radiation therapy). variability of incontinence rates reported in the litera-
The rate of rectal injuries is similar regardless of the ture. This variability is related to three main factors: def-
technique (retropubic or laparoscopic), ranging from inition of incontinence, modalities of evaluation, and
0.5% to 2%.11,19–21 follow-up.
In laparoscopic procedure, using a rectal bougie can The definition of incontinence varies considerably
help to identify the rectal wall but cannot ensure risk-free from one study to another; total absence of protection
surgery. or use of a maximum of one protection. Geary et al.23
reported that 80.1% of patients did not require any
protection, while Eastham et al., 32 considering
Ureteric Injuries
patients who required a maximum of one protection to
Ureteric injuries are also rare, occurring in 0.05% to be continent, reported that 91% of patients were
0.3% of cases, regardless of the technique. continent.
No consensus has therefore been reached concerning
the definition of incontinence. In Table 31-2, continence
Complications of Specimen Extraction
is defined as complete absence of either occasional or
and Trocar Removal
permanent protection.
The complications that can occur are: bleeding from ori- The modalities of evaluation also vary from one author
fice of 10 mm trocar and ileum injury (with the endobag to another: clinical interview by the surgeon, clinical
or during the opening for extraction of the specimen). interview by another doctor, self-administered question-
These complications are exceptional. naire. The method of data collection is essential to obtain
perfectly objective information. Development of a vali-
dated questionnaire, based on a standardized definition,
Early Postoperative Complications
would facilitate comparison of the various results
Thromboembolic Complications
reported in the literature.27,30
Thromboembolic complications constitute the main cause The follow-up also frequently differs from one
of postoperative mortality in open procedure.1,22 series to another. Although about one-half of patients
Dillioglugil19 reported 2% of thromboembolic accidents, are “dry” between 1 and 3 months, and most are dry
while Rassweiler11 and Guillonneau17 reported <0.5% of at 1 year, some patients can still recover for up to 2
thromboembolic accidents. years. A follow-up of at least 12 months is therefore
Prophylactic anticoagulation and early mobilization essential. 23,28,32,33
(especially after laparoscopic procedure) decrease the fre- Furthermore, the main predisposing factor for post-
quency of these complications. operative incontinence appears to be age >70
years.14,16,24,32,34 The respective roles of preoperative
continence, stage of the disease, and development of
Anastomotic Leaks
anastomotic stenosis are also discussed.14,15,23,29,32
Anastomotic leaks are often missed when minimal and Finally, some authors consider that certain technical
correctly drained, and their incidence is therefore often modifications appear to facilitate preservation of conti-
underestimated. nence: quality of apical dissection,15 neurovascular bun-
Length time of bladder catheter has to be directly dle preservation,25 preservation of the bladder neck, and
related to the quality of the anastomosis. preservation of puboprostatic ligaments. In any case,
experience of the surgeon is an essential factor to
improve the recovery of continence.
Lymphoceles
Lymphoceles are now less frequent, whatever the way
laparoscopic or open, due to better selection of patients Impotence
requiring pelvic lymph node dissection.
As for continence, objective evaluation of sexual potency
encounters a number of difficulties: absence of a consen-
Late Postoperative Complications sual definition of sexual potency, various methods of eval-
uation, and variable follow-up (Table 31-4).
Urinary incontinence and impotence are the two most The definition of sexual potency varies according to the
frequent and most disabling functional sequelae. Stenosis criteria adopted: erection with or without sexual inter-
of the vesicourethral anastomosis is observed more rarely. course and erection allowing sexual intercourse.
542 Part V Prostate Gland and Seminal Vesicles

Table 31-3 Continency


Number Follow-Up
Study of Points (months) Evaluation Mean Age =1 Pad/Day (%)

Catalona et al.14 1325 50 Physician 63(38-79) 8

Geary et al.23 458 >18 Physician 64.1±0.3 19.90

Leandri et al.24 620 12 Physician 68 (46–84) 5

Steiner25 593 12 Physician ? (34–76) 5.50

*Turk et al.9 125 9 Physician 60 (37–72) 8

*Bollens et al.12 50 6 Physician 63 (47–71) 15

Igel et al.22 692 Physician 21

*Rassweiler et al.7 100 6 Questionnaire 68 (55–74) 22

Fontaine et al.26 116 51.6 Patient quest 65.2 (48–76) 19.80

McCammon et al.27 203 40.3 Patient quest 63.7 (43–73) 23.70

Jonler et al.28 86 22.5 Patient quest 64 (49–75) 47

Bates et al.29 83 22 Patient quest 65 (49–73) 24

Walsh et al.15 64 18 Patient quest 57 (36–67) 7

Wei and Montie30 145 12 Patient quest 62.3 (40–80) 13

Talcott et al.31 94 12 Patient quest 61.5 39

*Guillonneau et al.17 567 12 Patient quest 63 (49–77) 21

*Hoznek et al.10 29 12 Patient quest 64.8 (47–77) 13.80

*Laparoscopic approach.

Anastomotic Stenosis
The methods of evaluation of sexual potency are also
very heterogeneous, as shown in Table 31-4: clinical Stenosis of the vesicourethral anastomosis occurs in
interview by the surgeon, clinical interview by another 0.5% to 4% of cases.14
doctor, self-administered questionnaire. The possible use
of a treatment for erectile dysfunction, not systematically
ONCOLOGIC RESULTS
reported, also makes it difficult to compare various series.
Follow-up also constitutes an important parameter in The aim of laparoscopic procedure that is to increase the
this evaluation. While a large number of series have quality of the dissection and to reduce the morbidity is
demonstrated the possibility of late recovery, most stud- only acceptable if the oncologic efficacy is demon-
ies are limited to a relatively short follow-up. The assess- strated. For patients with organ-confined disease, at
ment of recovery of sexual function requires a follow-up 3 years, Catalona et al.14 had a recurrence-free survival of
of at least 18 months.15,36 93%. Similar results are observed by the Johns Hopkins
Other elements must also be taken into account: qual- group where their 5-year recurrence-free was 97% for
ity of erections before surgery, patient’s age, and type of organ-confined cancer. In retropubic approach, the rate
surgery (preservation of one or two neurovascular bun- of positive margins range between 15% and 40%
dles). Moreover, the surgeon’s experience is essential for depending on the experience of the team and the preser-
preserving the neurovascular bundles. Finally, the selec- vation or not of the neurovascular bundles. In their expe-
tion of patients eligible for preservation of neurovascu- rience of laparoscopy, after 3 years, Guillonneau et al.37
lar bundles is an important factor, rarely mentioned in observed that 92% of the pT2a and pT2b had a PSA
the literature.14 < 0.1 ng/ml. Among the patients with a PSA < 10 ng/ml
Chapter 31 Laparoscopic Radical Prostatectomy 543

Table 31-4 Potency


Number Mean Follow-up Impotence
Study of Points Age Months Evaluation (%) Definition

Leandri et al.24 620 68 12 Physician 29 Intercourse

Igel et al.22 692 Physician 97.5 Erection

*Turk et al.9 44 60 ? Physician 41 Intercourse with sildenafil

*Bilatéral 5 nc

*Unilatéral 39 nc

*Rassweiler et al.11 100 68 ? Physician Intercourse

*Unilatéral 10 6/10 Intercourse with sildenafil


or PGE1

Geary et al.23 459 64.1 18 Questionnaire 44.4 Intercourse

Bilatéral 69 68.1

Unilatéral 203 86.7

Catalona et al.14 858 63 18 Questionnaire 33.5 Intercourse

Bilatéral 798 32

Unilatéral 60 53

Fowler et al.35 739 ? 24 Patient quest + 79 Erection


questionnaire

Walsh et al.15 64 57 18 Patient quest 14 Intercourse 1/3 with


sildenafil

Stanford et al.34 1291 62.9 18 Patient quest + 59 Intercourse


questionnaire

Bilatéral 44

Unilatéral 46.6

No preservation 34.4

Talcott et al.31 94 61.5 12 Patient quest

Bilatéral 19 4/19 Incomplete erection

*Bollens et al.12 50 63 6 Patient quest

*Bilateral 6 1/6 Intercourse with sildenafil


or PGE1

*Hoznek et al. 25 64.8 12 Patient quest 11/25 Erection (intercourse?)

*Laparoscopic approach.
544 Part V Prostate Gland and Seminal Vesicles

and a Gleason score < 7, 95.5% had a PSA < 0.2 ng/ml. Teaching could be ensured by virtual reconstitution of
The overall rate of positive margin was 17%. No port prostatectomies from operative videos. The trainee sur-
seeding was observed. geon would be able to practice on the operating console
in the same position as when performing remote-
controlled laparoscopic surgery.
REMOTE-CONTROLLED ASSISTED RADICAL
Finally, a senior surgeon would be able to control two
PROSTATECTOMY
or three operating rooms, each equipped with a remote-
Laparoscopy is probably a transitional technique controlled system and console operated by a junior surgeon.
between open surgery and remote-controlled surgery. As The senior surgeon would be in a separate room, super-
it is no longer necessary to open the abdomen to operate vising the external and endoscopic screens of each room
and almost all of the necessary instruments can be intro- under his direction. His console could be connected in a
duced via trocars, it is only logical to develop remote- fraction of a second to the other operating consoles in the
controlled surgery for the following reasons; the event of difficulties. Such “industrialization of surgery”
laparoscopy position is not very ergonomic. The surgeon will probably considerably change our everyday practice.
stands to the side of the patient and has to operate by Performing surgery at distant sites would appear to be
crossing his hands over the midline, especially for more difficult; a surgeon would still need to be present
sutures. This position can cause scapular pain. The vesi- on site to insert the trocars and to treat any complica-
courethral suture at the bottom of the lesser pelvis is also tions. The transmitted image could be used to help
a difficult procedure, requiring an experienced operator another surgeon seeking advice, but there is no future for
to ensure a good quality anastomosis. remote-controlled surgery of the entire procedure.
Many papers have been written on experimental In conclusion, remote-controlled surgery does not
remote-controlled surgery in animals, but very few have currently provide any significant benefit for the patient.
yet been published in man. A first series of 10 cases of Bleeding is higher than with laparoscopic surgery, but the
remote-controlled assisted radical prostatectomy has been development of a hemostasis system, such as good qual-
published.38 The operating time was 9 hours. It should be ity bipolar forceps, should reduce bleeding. Suture is eas-
noted that this team started to perform remote-controlled ier, but a surgeon experienced in laparoscopy can achieve
assisted radical prostatectomy without any previous expe- similar results. For the operator, the operation is less tir-
rience of laparoscopic surgery. Abbou et al.39 published 1 ing and the various procedures are facilitated. Virtual
case and, more recently, Guglielmo et al.40 and Pasticier teaching systems must be developed to allow young oper-
et al.41 also published their cases. In the United States, ators to learn the basic techniques of remote-controlled
Tewari et al.42 in Detroit, after 1 year of training in pelvic surgery.
laparoscopy, have acquired a certain experience with
remote-controlled laparoscopic surgery (70 cases).
The Institut Montsouris experience, based on 30 cases, REFERENCES
together with the cases operated in Detroit, illustrates
the following advantages of remote-controlled surgery: 1. Schuessler WW, Schulam PG, Clayman RV, Kavoussi LR:
good ergonomy for the surgeon, whose forearms are Laparoscopic radical prostatectomy: initial short-term
supported to allow precise manipulation of the joysticks. experience. Urology 1997; 50(6):854–857.
2. Raboy A, Ferzli G, Albert P: Initial experience with
A possible reduction of the amplitude of displacement of
extraperitoneal endoscopic radical retropubic
the remote-controlled robotic arms and especially 6 prostatectomy. Urology 1997; 50(6):849–853.
degrees of freedom, allowing rotation of the remote- 3. Guillonneau B, Cathelineau X, Barret E, Rozet F,
controlled needle holders in all directions, makes sutur- Vallancien G: Laparoscopic radical prostatectomy:
ing much easier. The 3D vision provided by a dual scope technical and early oncological assessment of 40
is extremely precise, but it is not specific to remote- operations. Eur Urol 1999; 36:14–20.
controlled surgery. 4. Guillonneau B, Vallancien G: Laparoscopic radical
The disadvantages of remote-controlled surgery, at the prostatectomy: the Montsouris experience. J Urol 2000;
present time, are the absence of practical bipolar coagula- 163:418.
tion to ensure reliable hemostasis and intraoperative 5. Abbou CC, Salomon L, Hoznek A, et al: Laparoscopic
bleeding is slightly greater than with that of laparoscopy. radical prostatectomy: preliminary results. Urology 2000;
55(5):630–634.
As good quality bipolar coagulation is essential to ensure
6. Guillonneau B, Vallancien G: Laparoscopic radical
an optimal laparoscopic procedure, engineers are cur- prostatectomy: the Montsouris technique. J. Urol 2000;
rently working to rapidly develop this instrument. 163:1643.
The future of surgery is clearly remote-controlled sur- 7. Rassweiler J, Sentker L, Seemann O, et al: Heilbronner
gery, as it is less tiring for the surgeon and ensures more laparoscopic radical prostatectomy. Technique and results
precise sutures. after 100 cases. Eur Urol 2001; 40:54–64.
Chapter 31 Laparoscopic Radical Prostatectomy 545

8. Rassweiler J, Seeman O, Hatzinger M, Frede T: 24. Leandri P, Rossignol G, Gautier JR, Ramon J: Radical
Laparoscopic anatomical radical prostatectomy: retropubic prostatectomy: morbidity and quality of life.
experience after 600 cases. J Urol 2003; 169(4):249. Experience with 620 consecutive cases. J Urol 1992;
9. Turk I, Deger S, Winkelmann B, Schonberger B, Loening 147(3 Pt 2):883–887.
SA: Laparoscopic radical prostatectomy. Technical aspects 25. Steiner MS: Continence-preserving anatomic
and experience with 125 cases. Eur Urol 2001; radical retropubic prostatectomy. Urology 2000;
40(1):46–52 [Discussion 53]. 55:427–435.
10. Hoznek A, Salomon L, Olsson LE, et al: Laparoscopic 26. Fontaine E, Izadifar V, Barthelemy Y, Desgrippes A,
radical prostatectomy. The Creteil experience. Eur Urol Beurton D: Urinary continence following radical
2001; 40(1):38–45. prostatectomy assessed by a self-administered
11. Rassweiler J, Sentker L, Seemann O, Hatzinger M, questionnaire. Eur Urol 2000; 37(2):223–227.
Rumpelt HJ: Laparoscopic radical prostatectomy with the 27. McCammon KA, Kolm P, Main B, Schellhammer PF:
Heilbronner technique: an analysis of the first 180 cases. Comparative quality-of-life analysis after radical
J Urol 2001; 166(6):2101–2108. prostatectomy or external beam radiation for localized
12. Bollens R, Vanden BM, Roumeguere T, et al: prostate cancer. Urology 1999; 54(3):509–516.
Extraperitoneal laparoscopic radical prostatectomy. 28. Jonler M, Madsen FA, Rhodes PR, et al: A prospective
Results after 50 cases. Eur Urol 2001; 40(1):65–69. study of quantification of urinary incontinence and quality
13. Koch MO, Smith JA Jr: Clinical outcomes associated with of life in patients undergoing radical retropubic
the implementation of a cost-efficient program for radical prostatectomy. Urology 1996; 48(3):433–440.
retropubic prostatectomy. Br J Urol 1995; 76(1):28–33. 29. Bates TS, Wright MP, Gillatt DA: Prevalence and impact
14. Catalona WJ, Carvalhal GF, Mager DE, Smith DS: of incontinence and impotence following total
Potency, continence and complication rates in 1870 prostatectomy assessed anonymously by the ICS-male
consecutive radical retropubic prostatectomies. J Urol questionnaire. Eur Urol 1998; 33(2):165–169.
1999; 162(2):433–438. 30. Wei JT, Montie JE: Comparison of patients’ and
15. Walsh PC, Marschke P, Ricker D, Burnett AL: physicians’ rating of urinary incontinence following radical
Patient-reported urinary continence and sexual function prostatectomy. Semin Urol Oncol 2000; 18(1):76–80.
after anatomic radical prostatectomy. Urology 2000; 31. Talcott JA, Rieker P, Propert KJ, et al: Patient-reported
55:58–61. impotence and incontinence after nerve-sparing radical
16. Zincke H, Bergstralh EJ, Blute ML, et al: Radical prostatectomy. J Natl Cancer Inst 1997;
prostatectomy for clinically localized prostate cancer: 89(15):1117–1123.
long-term results of 1143 patients from a single 32. Eastham JA, Kattan MW, Rogers E, et al: Risk factors for
institution. J Clin Oncol 1994; 12(11):2254–2263. urinary incontinence after radical prostatectomy. J Urol
17. Guillonneau B, Rozet F, Cathelineau X, et al: 1996; 156(5):1707–1713 (review).
Perioperative complications of laparoscopic radical 33. Donnellan SM, Duncan HJ, MacGregor RJ, Russell JM:
prostatectomy: the Montsouris 3-year experience. J Urol Prospective assessment of incontinence after radical
2002; 167(1):51–56. retropubic prostatectomy: objective and subjective
18. Lepor H, Nieder A, Ferrandino M: Intraoperative and analysis. Urology 1997; 49(2):225–230.
postoperative complications of radical retropubic 34. Stanford JL, Feng Z, Hamilton AS, et al: Urinary and
prostatectomy in a consecutive series of 1000 cases. J Urol sexual function after radical prostatectomy for clinically
2001; 166:1729–1733. localized prostate cancer: the prostate cancer outcomes
19. Dillioglugil O, Leibman BD, Leibman NS, et al: Risk study. JAMA 2000; 283(3):354–360.
factors for complications and morbidity after radical 35. Fowler FJ Jr, Barry MJ, Lu-Yao G, et al: Patient-reported
retropubic prostatectomy. J Urol 1997; 157(5):1760–1767 complications and follow-up treatment after radical
(review). prostatectomy. The National Medicare experience:
20. Guillonneau B, Rozet F, Barret E, Cathelineau X, 1988–1990 (updated June 1993). Urology 1993;
Vallancien G: Laparoscopic radical prostatectomy: 42(6):622–629.
assessment after 240 procedures. Urol Clin North Am 36. Kim ED, Nath R, Slawin KM, et al: Bilateral
2001; 28(1):189–202. nerve grafting during radical retropubic
21. Guillonneau B, Gupta R, El Fettouh H, et al: prostatectomy: extended follow-up. Urology 2001;
Laparoscopic management of rectal injury during 58(6):983–987.
laparoscopic radical prostatectomy. J Urol 2003; 37. Guillonneau B, Gerard C, El Fettouh H, et al: Mid-term
169(5):1694–1696. oncological follow-up of laparoscopic radical
22. Igel TC, Barrett DM, Segura JW, Benson RC Jr, Rife prostatectomy: mono-institutional experience based on
CC: Perioperative and postoperative complications from 800 consecutive patients. In 97th Annual Meeting, AUA,
bilateral pelvic lymphadenectomy and radical retropubic Orlando, May 2002.
prostatectomy. J Urol 1987; 137(6):1189–1191. 38. Binder J, Kramer W: Robotically assisted laparoscopic
23. Geary ES, Dendinger TE, Freiha FS, Stamey TA: radical prostatectomy. BJU Int 2001; 87:408–410.
Incontinence and vesical neck strictures following radical 39. Abbou CC, Hoznek A, Salomon L, et al: Laparoscopic
retropubic prostatectomy. Urology 1995; 45(6):1000–1006 radical prostatectomy with a remote controlled robot.
(review). J Urol 2001; 165:1964–1966.
546 Part V Prostate Gland and Seminal Vesicles

40. Guglielmo B, Nakada SY, Rassweiler JJ: Future 42. Tewari A, Desari R, Hemal A, et al: A prospective
developments and perspectives in laparoscopy. Eur Urol comparison of robot-assisted anatomic prostatectomy and
2001: 40:84–91. conventional radical retropubic prostatectomy (RRP): the
41. Pasticier G, Rietbergen JBW, Guillonneau B, et al: Vattikuti Urology Institute Experience. J Urol 2003;
Robotically assisted laparoscopic radical prostatectomy: 169(4):78.
feasibility study in men. Eur Urol 2001; 40:70–74.
C H A P T E R

32Complications of Surgical Treatment


for Localized Prostate Cancer
Glen W. Barrisford, MD,
and Michael P. O’Leary, MD, MPH

In developed countries prostate cancer represents the ing preservation of the cavernous nerves, improved
most commonly detected malignancy and the second hemostasis, superior exposure, and a very defined apical
leading cause of cancer death in men over 50. In the year dissection. These improvements, coupled with improve-
2000, over 180,000 American men were newly diag- ments in anesthesia and surgical care, have lead to a
nosed.1 Despite the 30% lifetime risk of developing steady decline in morbidity and mortality over the last
prostate malignancy, the protracted course of this disease 20 years.11
only results in a comparatively low 3% risk of death.2 Despite the great success of surgical treatment for
The number of prostate cancer deaths has been esti- localized prostate disease, the complications remain sig-
mated to rise in accordance with an aging worldwide nificant. Persistent imperfections of surgical therapy have
population. However, the emphasis on screening and encouraged clinicians to seek other, less morbid nonsur-
early detection, through digital rectal examination gical treatment modalities. Alternatives to surgical ther-
(DRE) and prostate-specific antigen (PSA) testing, has apy have included watchful waiting,12,13 transperineal
resulted in a downward trend in the United States. prostate brachytherapy,14,15 external and conformal beam
Accordingly, 77% of newly detected cancers will be lim- radiation therapy, cryotherapy, and laser ablation.
ited to the prostate (T1 to T2).3 However, these alternative modalities offer an equally
The objective of treatment for localized prostate complex combination of risks and benefits. Ultimate
malignancy is cure. Historically, prostatectomy has been choice of treatment is therefore a comprehensive deci-
treatment of choice. Originally described in 1894 by sion based on a number of factors, including patient pref-
Fuller,4 prostatectomy was performed via the suprapubic erence and physician bias.
approach. However, this method was a blind 15-minute This chapter addresses several central topics. First, the
procedure that required digital enucleation of the gland. discussion will focus on the surgical complications.
In 1900, Freyer5,6 completed a series of 1600 cases that Second, the methods designed to reduce risks will be con-
carried a 5% mortality. This mortality was considered sidered. Third, the treatment options for the presented
diminutive in the absence of antimicrobials and blood complications will be evaluated. To conclude, there will
product replacement. In 1947, Millin7 introduced the be consideration of quality-of-life evaluation and treat-
retropubic prostatectomy, which was summarily ment satisfaction in the setting of common complications.
embraced as the new method of choice. Several modifi-
cations and new techniques followed, but radical change
SURGICAL APPROACHES
did not again take place until 1979. At this time, a more
complete description of the surgical anatomy resulted in The anatomic prostatectomy (radical retropubic prosta-
an innovative and meticulous surgical approach to the tectomy) described by Walsh remains the mainstay of
dissection of the prostate and with preservation of the current surgical technique. Many variations and modifi-
neurovascular bundles.8,9 The anatomic prostatectomy cations are utilized, but the critical elements are near uni-
described by Walsh10 offered several advantages, includ- versal. The dissection can be completed in a retrograde

547
548 Part V Prostate Gland and Seminal Vesicles

or anterograde fashion with judicious preservation of the Several general guidelines are followed when prepar-
circular fibers of the bladder neck and neurovascular ing for operative therapy. Radical prostatectomy should
bundles.16 In comparison to the 5% mortality described be delayed 4 to 8 weeks following transrectal ultrasound
by Freyer, contemporary series utilizing the anatomic guided biopsy10,21 and 3 to 4 months following
retropubic prostatectomy carry a morbidity and mortal- transurethral resection of the prostate.21 The purpose of
ity in the range of 0% to 1.7%.10 this delay is to afford resolution of the postprocedure
Perineal prostatectomy remains as an alternative to inflammatory response, resulting in a more precise intra-
radical retropubic prostatectomy. The main advantages operative dissection.
include an avascular field, improved exposure for vesi- At our institution, bowel preparation and autologous
courethral anastomosis, and dependent postoperative blood donation are offered but are not routinely under-
drainage.10 However, there are several disadvantages. taken. Transfusion rates approximate 20%. Perioperative
First, there exists a requirement for two incisions to com- pain control is established with intravenous narcotic
plete the pelvic lymph node dissection. Second, this boluses and 48 hours of standing dose ketorolac.
method presents a more difficult anatomic approach for Transition to oral narcotics routinely occurs by postoper-
the task of preserving the neurovascular bundles. ative day 1. The expected length of hospital stay is 2 days.
Additionally, the perineal approach is contraindicated in The average cost of hospitalization (in the absence of
individuals who have undergone prior open prostate sur- physician reimbursement) is approximately $25,500.
gery, in those who are obese patients, and in patients with
musculoskeletal conditions that would prohibit exagger-
OPERATIVE COMPLICATIONS
ated lithotomy positioning.
Laparoscopic prostatectomy has recently gained wide- Hemorrhage predominates as the most common intraop-
spread recognition following its initial description in erative complication associated with prostatectomy.
1999.17–19 However, there exists ongoing debate regard- Venous hemorrhage may occur during pelvic lym-
ing the benefits. For the individual surgeon, it is a techni- phadenectomy and originates from branches of the
cally demanding procedure with a relatively steep hypogastric vein. Additionally, venous backbleeding from
learning curve. Initial cases often require extended opera- the Doral vein complex can occur during several stages of
tive time and result in increased urinary leakage and rectal the procedure, including incision of the endopelvic fas-
injuries.20 These differences eventually become statisti- cia, division of the puboprostatic ligaments, and during
cally insignificant once the preliminary obstacles have exposure of the prostatic apex. Expected blood loss is
been overcome. In experienced hands, the laparoscopic typically 1 liter,22 and is routinely greater during nerve-
approach offers a reduction in lymphoceles, wound infec- sparing approaches. Persistent hemorrhage characteristi-
tion, pulmonary embolism, pneumonia, and anastomotic cally resolves once the dorsal vein complex is divided and
stricture.20 Laparoscopic prostatectomy offers potential ligated.
new alternatives for reducing surgical morbidity and Rectal injury is a rare complication of radical prostate
improving the results of radical prostate surgery. surgery (0% to 5.3%),22–26 typically occurring during the
apical dissection while developing a plane between the
rectum and Denonvilliers’ fascia. In the event of injury,
PATIENT SELECTION
the rectum should be repaired in 2 layers with an inter-
Preoperative evaluation and appropriate patient selection posed pedicle of omentum. This method has been asso-
is the critical first step in complication reduction. At our ciated with a low incidence of rectourethral fistula and
institution, the preoperative evaluation begins in the uro- abscess formation.23,24 Rectal injuries that are not identi-
logic clinic with a complete history, physical examina- fied intraoperatively carry the highest probability of fis-
tion, and appropriate screening. Criteria for selection of tula formation. Intestinal diversion can usually be
the surgical candidate includes: biopsy proven histologic avoided, with the exception of patients who have received
presence of prostate malignancy; clinically established previous pelvic irradiation. In these cases, diversion
localized disease (T1 to T2); a life expectancy of >10 years; should be the standard of care.
absence of surgical contraindications (comorbid condi- Ureteral injury is another rare complication that
tions); and informed consent of the patient. Under the occurs on the order of 1%.25 This injury classically occurs
guidance of the anesthesia team, the assessment contin- during extended pelvic lymph node dissection at the iliac
ues in the preadmission testing center. The American bifurcation or while dissecting the posterior aspect of the
Society of Anesthesiology (ASA) scoring system is utilized bladder neck. Injuries are more commonly associated with
to estimate the risks associated with various existing comor- advanced stages of disease and are repaired with uretero-
bidities. Additional evaluation is tailored to the individual neocystostomy if identified intraoperatively.
patient. In the absence of further preoperative assess- Nerve injury is another rare complication associated
ment, the patient returns on the morning of surgery. with pelvic lymph node dissection. These injuries often
Chapter 32 Complications of Surgical Treatment for Localized Prostate Cancer 549

occur from sharp dissection or as a result of retractor unintentionally removed, a single attempt at replacement
positioning. The two most commonly injured nerves are can be initiated with a smaller caliber coude tip catheter.
the obturator and the femoral. Obturator nerve injury If the catheter does not easily pass on the first attempt,
may result in a thigh abduction deficit, whereas femoral placement should be undertaken under direct vision with
nerve injury may lead to distressing weakness of the cystoscopy. If catheter removal occurs after postoperative
quadriceps, combined with anteromedial thigh paresthe- day 5, the patient can be safely monitored without
sia. Intraoperative identification of these injuries indi- catheter reinsertion.21
cates primary repair with fine absorbable sutures. Injury Anastomotic leakage is another rare early complica-
that is not identified intraoperatively is managed conser- tion of radical prostate surgery. Typically the vesi-
vatively with physical therapy. The majority of patients courethral anastomosis consists of 4 to 6 precisely placed
will eventually fully recover from these injuries. sutures. Leakage can occur when the anastomotic
integrity is incomplete or compromised. The observation
of high serous output from the surgical drains suggests
POSTOPERATIVE COMPLICATIONS
urinary leakage. To verify suspected urinary leakage, a
Early Complications
creatinine level can be measured in the drain output.
In the immediate postoperative, period the most com- Drain output creatinine that mirrors urinary creatinine is
mon and potentially devastating complications include diagnostic of an anastomotic leak. The majority of small
thromboembolism, pulmonary embolism, and myocar- anastomotic leaks resolve spontaneously or with
dial infarction. Subcutaneous heparin, sequential com- extended Foley catheter drainage. In the setting of a large
pression devices, and early ambulation have all been leak, urinoma formation can occur and may require CT-
successfully utilized postoperatively to minimize risks. guided drainage.
Recent series have demonstrated a 0.8% to 2.7% throm-
boembolic complication rate.21 Prophylactic low dose
Late Complications
heparin has not been associated with increased risks of
Bladder Neck Contracture
postoperative bleeding. Additionally, intraoperative
Trendelenburg positioning has been thought to enhance Bladder neck contracture is a late complication of prosta-
passive venous drainage of the lower extremities. tectomy. It is known to occur weeks to months postoper-
Myocardial infarction remains an uncommon, though atively and typically presents with urinary incontinence
grave, complication, requiring special efforts to minimize and/or a weak urinary stream. Generally, bladder neck
risks. Specific preoperative evaluation and perioperative contracture is thought to be an uncommon complication.
management recommendations are addressed in high- However, it has been reported in 3% to 12% of cases.10
risk patients during the preoperative assessment. The It is suspected to be a consequence of poor vesicourethral
coordination of the surgical, anesthesia, and cardiology anastomotic quality. The construction of a dependable
teams is crucial in order to implement optimum cardio- anastomosis requires adequate vascularity and an imper-
vascular medical management. Likewise early recogni- meable seal. Meticulous surgical technique, hemostasis,
tion of symptoms and intervention are essential in eversion of the bladder neck, superior mucosal apposi-
appropriately managing this complication. tion, and the use of 4 to 6 fine absorbable sutures pro-
Wound infection/dehiscence, seroma, lymphocele motes a proper anastomosis. Intraoperative bleeding can
formation, and delayed hemorrhage are all uncommon lead to pelvic hematoma and the formation of an anasto-
early postoperative complications. Sterile technique and motic disruption. Poor surgical technique can lead to uri-
perioperative gram-positive prophylactic antibiotic cov- nary extravasation and subsequent periurethral scarring.
erage is utilized to reduce the risk of wound infection. This scarring can lead to urinary stricture and inconti-
Appropriate fascial closure, hemostasis, and lymphostasis nence.
reduce risks for dehiscence, hemorrhage, and lympho- Bladder neck contracture is a difficult diagnosis to
cele, respectively. Hematoma or lymphocele formation establish when overflow incontinence is the only pre-
may require computed tomography (CT)-guided senting complaint. Evaluation includes measurement of
drainage in order to prevent the development of a pelvic postvoid residual urine volumes and identification using
abscess. urethroscopy. Once the diagnosis is established it can be
Anastomotic disruption commonly occurs in the early treated with dilation. Typically, 1 to 2 dilation procedures
postoperative period when the Foley catheter is uninten- will resolve the contracture. Rarely dilation can result in
tionally dislodged. Intraoperative testing of the balloon a perforation into the rectum and result in a rectourethral
establishes catheter integrity. Further methods to prevent fistula. This can become a very challenging problem.
dislodgment include secure catheter anchorage via intra- Failure of dilation can be resolved with incisions made in
operative cystotomy and attachment of the catheter to an the bladder neck at the 12 o’clock, 3 o’clock, and
abdominal wall button. In the event that the catheter is 9 o’clock positions.
550 Part V Prostate Gland and Seminal Vesicles

nence rates are observed with advancing age. This has


Urinary Incontinence
been attributed to rhabdosphincter atrophy32 and neural
Urinary incontinence has been considered by many to be degeneration.33,34 Preoperative urinary incontinence can
the most distressing and disabling complication of persist postoperatively. More advanced stages of disease
prostate surgery. Prior to the description of the anatomic are likely to be associated with greater dissection and lead
prostatectomy, total urinary incontinence was observed to increased rates of incontinence. Surgical technique
in 10% of men.27 Fortunately, complete urinary inconti- and surgeon’s experience have also been determined to be
nence is now an uncommon occurrence. However, lesser significant factors in predicting postoperative continence
degrees of incontinence exist and represent one of the rates.35
most important quality-of-life measures in radical prosta- Evaluation of postoperative incontinence begins with a
tectomy patients. complete history and physical examination. Type and
Historically assessing the complication rate has been a severity of incontinence, precipitating factors, number of
challenge. In extensive academic series, incontinence daily episodes, and degree of protection (pads, penile
rates have been reported in 0.3% to 12.5% of clamp, etc.) required should be established. A history con-
patients.28–30 However, some degree of urinary leakage sistent with stress incontinence has been associated with
was reported in 30% of men according to the National the existence of sphincter dysfunction.36 Initial blood tests
Medicare Experience.31 The wide range of reported may incorporate blood urea nitrogen, creatinine, and PSA
incontinence could be attributed to varied definitions (to detect cancer recurrence). Measurement of postvoid
applied by both physicians and patients. As a conse- residual urine volume and noninvasive uroflow are
quence, studies reported in the literature have been diffi- included in the initial evaluation. However, urodynamic
cult to interpret. Several questionnaires have been testing remains the most valuable method available to
developed in order to standardize the definition of incon- accurately diagnose the cause of urinary incontinence.
tinence, in turn allowing for greater comparison among Cystourethroscopy can also be utilized to evaluate the
studies. mucosal surfaces, vesicourethral anastomosis, and bladder
Normal urinary continence is achieved when a stable wall, but it is particularly useful for direct visualization if
compliant detrusor is coupled with a competent bladder surgical intervention is considered.
outlet (sphincter mechanism). The sphincter mechanism The problem of postoperative urinary incontinence
is considered as two functionally distinct units, the proxi- can be dealt with effectively at many levels. Therapy
mal and distal urethral sphincters. The proximal sphinc- should be tailored to maximize patient’s quality of life
ter mechanism consists of the bladder neck, prostate, and and minimize additional risks and complications. Many
prostatic urethra to the level of the verumontanum. The patients will meet the criteria for urinary incontinence
distal urethral sphincter is comprised of mucosal infold- but do not consider it to negatively impact quality of life.
ing, longitudinal smooth muscle, striated muscle, and the It is vital to identify patient bother and expectations of
intrinsic periurethral rhabdosphincter distal to the veru- treatment. Once the diagnosis has been established, the
montanum. Passive continence is maintained by the stri- problem can be addressed in a stepwise fashion.
ated urethral sphincter. It consists of slow twitch fibers Fluid restriction, behavioral modification, anticholin-
that are capable of maintaining tone over prolonged peri- ergic/tricyclic antidepressant medication and pelvic floor
ods of time. This is in comparison to, periurethral leva- exercises have been utilized to address postoperative uri-
tor ani muscle fibers, which are fast twitch and are nary incontinence attributed to bladder dysfunction. For
associated with rapid forceful muscle contraction. This patients with significant urinary complaints who fail
mechanism is designed to maintain continence during these methods, augmentation cystoplasty,37 or neuro-
events that raise intra-abdominal pressure. Failure of modulation (not yet studied in postprostatectomy
the sphincter mechanism leads to urinary incontinence. patients) can be considered.
The proximal sphincter mechanism is removed during Sphincter dysfunction can be addressed similarly with
radical prostatectomy and the distal sphincter mechanism biofeedback and pelvic floor exercises. In cases of sphinc-
is then solely responsible for the maintenance of urinary ter dysfunction, these methods have met with mixed
continence. results with respect to restoration of continence.
A number of risk factors have been thought to be asso- However, they have been effective in reducing the
ciated with increased risk of urinary incontinence follow- postoperative interval required to regain continence.38
ing radical prostatectomy. Patient age at surgery, Alpha agonists (ephedrine, phenylpropanolamine) and
preoperative continence status, previous transurethral imipramine have been used in females with sphincteric
resection of the prostate, anastomotic stricture, stage of (stress) urinary incontinence but have not been evaluated
disease, surgical technique, and experience of the sur- in prostatectomy patients. These agents are unlikely to
geon have been evaluated to determine significance with offer substantial results and will likely be of limited use.39
respect to postoperative continence. Greater inconti- Bulking agents injected beneath the urethral mucosa
Chapter 32 Complications of Surgical Treatment for Localized Prostate Cancer 551

(glutaraldehyde cross-linked bovine collagen, silicone nerve sparing (unilateral or bilateral) were substantial
macroparticles) have offered short-term efficacy. The factors affecting potency.53,54 Younger age, less advanced
difficult identification of anatomic landmarks and scar- disease and preservation of both neurovascular bundles
ring has resulted in low rates of total dryness. Sling pro- were all found to be associated with greater postoperative
cedures have been used in limited cases as prophylaxis erectile function.
during radical prostate surgery. A rectus muscle fascial Postoperatively patients often may experience a wide
sling has effectively resulted in earlier and more complete variety of emotions related to a change in body image,
return of urinary continence.40 The most efficacious pro- the possibility of cancer recurrence, incontinence, erec-
cedure available to address urinary incontinence is the tile dysfunction, and a change in relationship dynamics.
artificial urinary sphincter. It has been widely used for During the months following surgery erectile dysfunc-
treating urinary incontinence of various etiologies.41–44 tion may be partially attributed to psychogenic factors.
Of those receiving an artificial urinary sphincter 77% However, preoperative and postoperative libido meas-
have been satisfied and have reported a significant reduc- urements by O’Leary et al.55 brief sexual function inven-
tion in the number of pads used for protection.45 In cases tory was not found to be significantly different from age
refractory to all forms of therapy urinary diversion has matched controls. Despite the numerous reasons to have
been employed to relieve intolerable incontinence. psychogenic erectile dysfunction, the majority of patients
A better understanding of the pelvic floor anatomy has have been found to have an organic origin as determined
resulted in improved continence rates. Despite the vari- by nocturnal penile tumescence or Rigiscan studies.56
ous modalities used to treat urinary incontinence it The origin of the erectile dysfunction is likely multifac-
remains a difficult problem. A number of intraoperative torial but largely a result of neurovascular damage and
techniques have been utilized to preserve urinary conti- preexisting disease.
nence. Bladder neck preservation,46 intussusception,27 Treatment for postprostatectomy erectile function has
and tubularization (preservation of functional urethral been addressed at several levels of invasiveness. Patient
length)47,48 have provided efficacy in reducing postoper- desire to regain potency should dictate the vigor of the
ative time to continence. Unfortunately, these techniques treatment regimen. An initial wait and see approach has
have not offered an improvement in overall continence. been advocated to allow spontaneous recovery of erectile
A longer functional urethral length has been associated function. Resolution of surgical inflammation, tissue
with greater continence.31,35 Consequently, preoperative healing, and resolution of neuropraxia can take 1 to 2
evaluation of the functional urethral length via endorec- years. After that time it is reasonable to assume that
tal magnetic resonance imaging may offer predictive greater improvement is unlikely.
information regarding postoperative continence rates.49 Vacuum erection devices offer a mildly effective non-
Such information could influence patient choice of ther- invasive therapy. However, the ring used to maintain
apy. Sparing of the puboprostatic46 ligament and mainte- tumescence may serve to prolong natural revitalization.57
nance of the urethral mucosal vascularity have not been Intraurethral prostaglandin therapy (MUSE) became
shown to affect,30 postoperative continence rates.50 widely popular in 1997. Initial reports boasted a 57% to
70% success, with a disproportionate 20% satisfaction
rate.58 However, intraurethral therapy offered a less inva-
Erectile Dysfunction
sive option for men than the more invasive intracaver-
When Young described radical perineal prostatectomy in nosal injection therapy (Caverject/PGE1/trimix). Since
1903, the procedure had been associated with a 60% 1983, intracavernosal therapy has been highly effective,
mortality.51 Following Young’s anatomic description once men overcome the fear of self-injection. It has been
mortality was reduced to 16.6% (incontinence 13%).52 associated with pain (14%), penile fibrosis (2% to 15%),
At that time preservation of potency had not been cumbersome mixtures, and relative expense.59 Sildenafil,
deemed a priority. Consequently, over 60% of patients a selective phosphodiesterase-5 inhibitor, gained wide
were left with erectile dysfunction. Despite the reduction popularity in 1998 as the first oral therapy for erectile
of mortality over time, emphasis had not shifted toward dysfunction. Initial trials reported a 43%60 response rate,
preservation of potency until the nerve-sparing approach while subsequent trials report satisfaction rates from
was described by Walsh and Donker.9 Subsequent to the 15% to 80%.61–63 Despite the mixed response rates it
development of the anatomic prostatectomy, preserva- became the best selling medication of all time and soon
tion of potency soon became a reasonable objective. became the first line of therapy for erectile dysfunction.
Potency has been defined as the ability to establish and Penile prosthesis has remained the most invasive and last
maintain an erection that is suitable for intercourse. In a line of therapy. In the event of treatment failure at all lev-
large series, 503 patients were deemed preoperatively els a prosthetic device can be placed with great efficacy.
potent. Postoperatively 68% were found to have retained Since 1973, these devices have successfully restored
erectile function. Age, stage of disease, and extent of potency. Infrequent infections can occur and may require
552 Part V Prostate Gland and Seminal Vesicles

explanation of the device. Fortunately, those cases are the plexus during radical retropubic surgery. J Urol 1979;
rare exception. 121:198–200.
Erectile dysfunction has historically been a low profile 9. Walsh PC, Donker PJ: Impotence following radical
problem of great magnitude. Over the last 2 decades prostatectomy: insight into etiology and prevention.
J Urol 1982; 128:492–497.
research has focused on improving the quality of life in
10. Walsh PC: Campbell’s Urology, 6th edition, pp
postprostatectomy patients. Treatment efficacy continues
2865–2886. Philadelphia, WB Saunders, 1992.
to rise while delivery methods become less invasive. The 11. Thompson IM, Middleton RG, Optenberg SA, et al:
future for therapy in erectile dysfunction will likely rely Have complication rates decreased after treatment for
on more selective and mechanistically varied oral agents, localized prostate cancer? J Urol 1999; 162:107.
and neurogenic and vascular growth factors aimed at the 12. Holmberg L, et al: A randomized trial comparing radical
restoration/preservation of function. prostatectomy with watchful waiting in early prostate
cancer. N Engl J Med 2002; 347(11):781–789.
13. Steineck G, Helgesen F, Adolfsan J, et al: Quality of life
Quality of Life after radical prostatectomy or watchful waiting. N Engl J
Quality of life has become a central theme to consider Med 2002; 347(11):790–796.
14. Albert M, et al: Late genitourinary and gastrointestinal
when counseling patients with localized prostate disease.
toxicity following magnetic resonance image guided
Over the last decade several nonsurgical therapies have
prostate brachytherapy with or without neoadjuvant
arisen that may offer less objectionable side effects while external beam radiation therapy. In press.
providing a comparable rate of survival. Patients are less 15. Shah SA, Cima R, Benoit E, et al: Rectal-urethral fistulas
likely to seek a therapy that will provide a lower quality after prostate brachytherapy. Diseases of Colon and
of life. In order to evaluate patient satisfaction with radi- Rectum. In press.
cal prostatectomy a number of questionnaires have been 16. Pontes J: Radical prostatectomy. In Surgery of Genitourinary
created, validated, and administered.64,65 These patient Pelvic Tumors, p 73. New York, Wiley-Liss, 1993.
surveys report that the primary concern remains survival, 17. Guillonneau B, Vallancien G: Laparoscopic radical
and than a strong concern for urinary incontinence and prostatectomy: initial experience and preliminary
erectile dysfunction. Difficulty with continence and assessment after 65 operations. Prostate 1999; 39(1):71–75.
18. Guillonneau B, Vallancien G: Laparoscopic radical
potency are most associated with a decrease in a patient’s
prostatectomy: the Mountsouris experience. J Urol 2000;
perception of his own well-being.21 However, as surgical
163:418.
techniques have improved in treating prostate cancer and 19. Dahl DM, et al: Laparoscopic radical prostatectomy:
as our ability to manage complications has also improved, initial 70 cases at a US university medical center. Urology
more men have and will continue to be effectively treated 2002; 60(5):859–863.
for this very common cancer. 20. Rassweiler J, et al: Laproscopic versus open radical
prostatectomy: a comparative study at a single institution.
J Urol 2003; 169:1689–1693.
21. Shekarriz B, Upadhyay J, Wood DP: Intraoperative,
REFERENCES perioperative, and long term complications of radical
prostatectomy. Urol Clin N Am 2001; 28(3):639–653.
1. Greenlee RT, Murray T, Bolden S, Wingo PA: Cancer 22. Rainwater LM, Segura JW: Technical consideration in
statistics, 2000. CA Cancer J Clin 2000; 50:7–33. radical retropubic prostatectomy: blood loss after ligation
2. Kirby RS, Brawer MK, Denis LJ: Prostate Cancer, of dorsal venous complex. J Urol 1990; 143:1163.
3rd edition, p 7. Oxford, UK, Health Press Limited, 23. Borland RN, Walsh PC: The management of rectal injury
2001. during radical retropubic prostatectomy. J Urol 1992;
3. Sylvester J, Blasko JC, Grimm P, Radge H: Interstitial 147:905.
implantation techniques in prostate cancer. J Surg Oncol 24. Harpster LE, Rommel FM, Sieber PR, et al: The
1997; 66:65–75. incidence and management of rectal injury associated with
4. Fuller E: The question of priority in the adoption of the radical prostatectomy in a community based urology
method of total enucleation, suprapubically, of the practice. J Urol 1995; 154:1435.
hypertrophied prostate. Ann Surg 1905; 41:520. 25. Hautman RE, Sauter TW, Wenderoth UK: Radical
5. Freyer PJ: A new method of performing prostatectomy. retropubic prostatectomy: morbidity and urinary
Lancet 1900; 1:774. continence in 418 consecutive cases. Urology 1994; 43:47.
6. Freyer PJ: One thousand cases of total enucleation of the 26. Guilloneau B, et al: Laparoscopic [correction of
prostate for radical cure of enlargement of that organ. Br laparoscopic] management of rectal injury during
Med J 1912; 2:868. laparoscopic [correction of laparoscopic] radical
7. Millin T: Retropubic urinary surgery. London, prostatectomy. J Urol 2003; 169(5):1694–1696.
Livingstone, 1947. 27. Walsh PC, Marschke PL: Intussusception of the
8. Reiner WG, Walsh PC: An anatomical approach to the reconstructed bladder neck leads to earlier continence
surgical management of the dorsal vein and Santorini’s after radical prostatectomy. Urology 2002; 59(6):934–938.
Chapter 32 Complications of Surgical Treatment for Localized Prostate Cancer 553

28. Schroder FH, van den Ouden D: Incontinence after 45. Gousse AE, et al: Artificial urinary sphincter for post-
radical prostatectomy-a study of 188 patients. J Urol radical prostatectomy urinary incontinence: long-term
1992; 147(Suppl):465A (abstract 1009). subjective results. J Urol 2001; 166:1755–1758.
29. Steiner MS, Morton RA, Walsh PC: Impact of anatomical 46. Deliveliotis C, et al: Radical prostatectomy: bladder neck
radical prostatectomy on urinary incontinence. J Urol preservation and puboprostatic ligament sparing-effects
1991; 145:512. on continence and positive margins. Urology 2002;
30. Rudy DC, Woodside JR, Crawford ED: Urodynamic 60(5):855–858.
evaluation of incontinence in patients undergoing 47. Peyromaure M, Ravery V, Boccon-Gibod L: The
modified Campbell radical retropubic prostatectomy: a management of stress urinary incontinence after radical
prospective study. J Urol 1984; 132:708. prostatectomy. Br J Urol 2002; 90:155–161.
31. Fowler FJ, et al: Patient-reported complications and 48. Connolly JA, Presti JC Jr, Carroll PR: Anterior bladder
follow-up treatment after radical prostatectomy. The neck tube reconstruction at radical prostatectomy
National Medicare Experience: 1988–1990. Urology preserves functional urethral length-a comparitive
1993; 42:622. urodynamic study. Br J Urol 1995; 75:766–770.
32. Burnett AL, Mostwin JL: In situ anatomical study of the 49. Coakley FV, et al: Urinary continence after radical
male urethral sphincter complex: relevance to continence retropubic prostatectomy: relationship with membranous
preservation following major pelvic surgery. J Urol 1998; urethral length on preoperative endorectal magnetic
160:1301. resonance imaging. J Urol 2002; 168:1032–1035.
33. Hollabaugh RS, Dmochowski RR, Kneib TG, et al: 50. John H, Suter S, Hauri D: Effect of radical prostatectomy
Preservation of putative continence nerves during radical on urethral blood flow. Urology 2002; 59(4):566–569.
retropubic prostatectomy leads to more rapid return of 51. McCullough AR: Prevention and management of erectile
urinary continence. Urology 1998; 51:960. dysfunction following radical prostatectomy. Urol Clin
34. Narayan P, Konety B, Aslam K, et al: Neuroanatomy of N Am 2001; 28(3):613–627.
the external urethral sphincter: implications for urinary 52. Young HH, Davis DM: Neoplasms of the urogenital tract.
continence preservation during radical prostate surgery. In Young’s Practice of Urology, pp 653–654. Philadelphia,
J Urol 1995; 153:337. WB Saunders, 1926.
35. Eastham JA, Kattan MW, Rogers E, et al: Risk factors for 53. Quinlan DM, Epstein JI, Carter BS, et al: Sexual function
urinary incontinence after radical prostatectomy. J Urol following radical prostatectomy: influence of preservation
1996; 156:1707. of neurovascular bundles. J Urol 1991; 145:998.
36. Chao R, Mayo ME: Incontinence after radical 54. Rabbani F, et al: Factors predicting recovery of erections
prostatectomy: detrusor or sphincteric causes. J Urol after radical prostatectomy. J Urol 2000; 164:1929–1934.
1995; 154:16. 55. O’Leary MP, Fowler FL, Lendrking WR, et al: A brief
37. Flood HD, Malhorta SJ, O’Connell HE, et al: Long term male sexual function inventory for urology. Urology 1995;
results and complications using augmentation cystoplasty 46:697–706.
in reconstructive urology. Neurourol Urodynam 1995; 56. Sohn MH, Seeger U, Sikora R, et al: Criteria for
14:297. examiner-independent nocturnal penile tumescence and
38. Van Kampen M, De Weerdt W, Van Poppel H, et al: rigidity monitoring (NPTR): correlations to invasive
Effect of pelvic-floor reeducation on duration and degree diagnostic diagnostic methods. Int J Impot Res 1995;
of incontinence after radical prostatectomy: a randomized 5:59–68.
controlled trial. Lancet 2000; 355:98. 57. Montorsi F, Guazzoni G, Strambi L, et al: Recovery of
39. Carlson KV, Nitti VW: Prevention and management of spontaneous erectile function after nerve sparing radical
incontinence following radical prostatectomy. Urol Clin retropubic prostatectomy with and without early
N Am 2001; 28(3):595–612. intracavernous injections of alprostadil. J Urol 1997;
40. Jorion JL: Rectus fascial sling suspension of the 158:1408–1410.
vesicourethral anastomosis after radical prostatectomy. 58. Costabile RA, Spevak M, Fishman IJ, et al: Efficacy and
J Urol 1997; 157:926. safety of transurethral alprostadil in patients with erectile
41. Elliott DS, Barrett DM: Mayo clinic long-term analysis dysfunction following radical prostatectomy. J Urol 1998;
of the functional durability of the AMS 800 artificial 160:1325–1328.
urinary sphincter: a review of 323 cases. J Urol 1998; 59. Evans C: Complications of intracavernosal therapy for
159:1206. impotence. In Carson C, Kirby R, Goldstein I (eds):
42. Gundian JC, Barrett DM, Parulkar BG: Mayo clinic Textbook of Erectile Dysfunction, pp 365–370. Oxford,
experience with the AS800 artificial urinary sphincter for Isis Medical Media, 1999.
urinary incontinence after transurethral resection of 60. Pfizer data on file. New York, Pfizer, 1998.
prostate or open prostatectomy. Urology 1993; 41:318. 61. Marks LS, Duda C, Dorey FJ, et al: Treatment of erectile
43. Perez LM, Webster GD: Successful outcome of artificial dysfunction with sildenafil. Urology 1999; 53:19–24.
urinary sphincters in men with post-prostatectomy urinary 62. Zippe CD, Jhaveri FM, Klein EA, et al: Role of Viagra
incontinence despite adverse implantation features. J Urol after radical prostatectomy. Urology 2000; 55:241–245.
1992; 148:1166. 63. Zagaja GP, et al: Sildenafil in the treatment of erectile
44. Scott FB: The artificial urinary sphincter. Experience in dysfunction after radical prostatectomy. Urology 2000;
adults. Urol Clin N Am 1989; 16:105. 56(4):631–634.
554 Part V Prostate Gland and Seminal Vesicles

64. Sebesta M, et al.: Questionnaire-based outcomes of 65. Penson DF, Litwin MS, Aaronson NK: Health related
urinary incontinence and satisfaction rates after radical quality of life in men with prostate cancer. J Urol 2003;
prostatectomy in a national study population. Urology 169:1653–1661.
2002; 60(6):1055–1058.
C H A P T E R

33 Seminal Vesicles: Diagnosis, Staging,


Surgery, and Management
Nelson N. Stone, MD, Richard G. Stock, MD,
and Pamela Unger, MD

Malignant tumors of the seminal vesicles are uncommon. Primary malignancies should undergo extensive radio-
The most frequent etiology results from secondary exten- logic evaluation to help ascertain the feasibility of surgi-
sion from prostate cancer. The diagnosis of prostate can- cal or radiotherapeutic intervention.
cer within the vesicles requires pathologic assessment of Transrectal ultrasound-guided biopsy of the vesicles is
seminal vesicle tissue. Unlike with prostate cancer, where easy to perform and for the most part is done the same
an elevated prostate-specific antigen (PSA) or an indurated way as prostate biopsy.8–16 The only differences is that
nodule motivates the urologist to perform a biopsy, there the spring-loaded needle should be positioned just poste-
are few indications for sampling seminal vesicle tissue. rior to the wall of the seminal vesicle so the core will
Generally, there are no presenting signs or symptoms include both posterior and anterior walls and not punc-
that would indicate tumor of the seminal vesicles. ture the base of the bladder (Figure 33-1). The cores of
However, pain, a sensation of prostate congestion, irrita- tissue are similar to prostate cores and, when normal,
tive bladder symptoms, hematospermia or hematuria should not be difficult for the pathologist to interpret.
may suggest to the clinician that a digital rectal and The decision to perform a seminal vesicle biopsy when
prostate exam is indicated.1–6 A palpable mass, involving evaluating patients with a diagnosis of prostate cancer is
the base of the prostate in association with an elevated controversial. Several studies have determined the accu-
PSA, usually indicates the presence of prostate cancer. racy of performing such biopsies, although the number of
However, a normal PSA, in association with a lesion that biopsies (ranging from 1 to 6 cores) and where to take
seems to occupy just the seminal vesicles may indicate a them has not been standardized. Wymenga et al.15 per-
primary process. If this is the case, further investigation formed 2 biopsies at the junction of the seminal vesicles
is warranted. More common benign lesions, such as sem- prior to radical prostatectomy. Eighty-three out of 138
inal vesicle cysts, as well as uncommon lesions, such as had positive biopsies and the accuracy was 91%. PSA,
papillary adenomas, cystadenomas, fibromas, myomas, clinical stage, and Gleason score were all predictive of SV
and schwannomas have been reported.1–6 Primary involvement. Okihara et al.16 performed SV staging
tumors of the vesicles include adenocarcinoma, rhab- biopsies in 244 men with prostate cancer and found 31%
domyosarcoma, squamous cell carcinoma, myosarcoma, positive. The number of cores taken varied with their
hemangiosarcoma, cystadenocarcinoma phyllodes, and clinical indication for the biopsies. Ninety underwent RP
seminoma.7 and no false positive biopsies were encountered. Stone
described a technique where 6 seminal vesicle biopsies
were taken, 3 from each side at a separate setting.13 With
DIAGNOSIS AND EVALUATION
this technique, 15% of patients were found to have vesi-
Biopsy is warranted when a suspicious lesion of the vesi- cle involvement. Linzer and associates7 compared the 6
cles is encountered and can be most easily accomplished biopsy method (n = 222) to a cohort of 187 men who had
by the transrectal ultrasound-guided route. Additional radical prostatectomy and developed indications for vesicle
evaluation will depend on the pathology, with asympto- biopsy (Table 33-1). Univariate and multivariate analyses
matic benign lesions requiring no further testing. demonstrated a PSA > 10 ng/ml, Gleason score ≥ 7,

555
556 Part V Prostate Gland and Seminal Vesicles

and clinical stage T2b (1992 AJCC) independently for predicting seminal vesicle involvement. While nomo-
predicted for involvement of the vesicles.7 Based on these grams are predictive of seminal vesicle involvement, they
data, the following indications for seminal vesicle biopsy are not a substitute for biopsy. In planning a patient’s
were developed: PSA > 10 ng/ml or stage T2b or greater treatment, the confirmed presence of vesicle involvement
or Gleason score ≥ 7. In contrast, Fowler found no will influence further diagnostic evaluation and the even-
advantage in performing a seminal vesicle biopsy in con- tual treatment decision.
junction with the prostate biopsy.17 He performed only Endorectal coil magnetic resonance imaging (MRI)
one biopsy that was taken at the time of the initial has also been used to detect extracapsular extension and
prostate biopsy. These data suggest that if seminal vesicle seminal vesicle invasion.20 The MRI has a high specificity
staging is to be performed, at least 4 biopsies should be when it detects SV involvement, but a sensitivity of <50%
taken and they should not be performed at the time of is suggesting that it often misses smaller tumors.21
the primary prostate biopsy. Pathologic confirmation, by biopsy will most likely be
Tables or nomograms have also been used to predict required if the MRI is suggestive of vesicle involvement.
extraprostatic disease.18–20 Penson et al.19 used the Partin If the urologist aggressively stages the high-risk
tables to assess the probability of extraprostatic extension prostate cancer patient using the seminal vesicle biopsy,
in 1162 men from the CapSure database. The calculated then a diagnosis of extraprostatic extension can be made
receiver operating characteristics curve area was 0.726 in advance of definitive therapy. Advance knowledge of
seminal vesicle involvement may preclude recommenda-
tions for standard radical prostatectomy, external beam
irradiation, or brachytherapy. In addition, patients with
seminal vesicle involvement are at increased risk for
pelvic lymph node metastases.12,13 One-third of men with
biopsy detected seminal vesicle involvement will have
metastases to the pelvic lymph nodes. These patients
should probably undergo pelvic lymph node dissection
prior to considering definitive therapy.12,13,16

TREATMENT OPTIONS FOR PATIENTS WITH


SEMINAL VESICLE INVOLVEMENT
The treatment of patients with seminal vesicle involve-
ment from prostate cancer will depend on whether it was
discovered following prostatectomy or if it was identified
by biopsy prior to definitive therapy. The literature seems
to indicate that the former is the more common scenario.
Figure 33-1 Transrectal ultrasound guided biopsy of the The options for treating patients following prostatectomy
seminal vesicles. include observation, adjuvant radiation therapy, or hor-

Table 33-1 Likelihood of Encountering Prostate Cancer Involving the Seminal Vesicles either by Trus-Guided
Seminal Vesical Biopsy (SVB, n = 222) Prior to Radiation Therapy or Following Radical Prostatectomy
(RP, n = 187)
Clinical Positive SVB in SV Involvement
Characteristics Radiation Patients in RP Patients p-Value

PSA 10–20 ng/ml 11/63 (17%) 6/39 (15%) 0.78

PSA > 20 ng/ml 17/53 (32%) 22/165 (13%) 0.98

T2b 19/96 (20%) 14/66 (21%) 0.82

T2c 11/37 (30%) 2/13 (15%) 0.31

Gleason score 7–10 23/62 (37%) 8/40 (20%) 0.06

From Linzer DG, Stock RG, Stone NN, et al: Urology 1996; 48:757–761.
Chapter 33 Seminal Vesicles: Diagnosis, Staging, Surgery, and Management 557

monal therapy. The argument for treatment is the high with mixed results. The lack of randomized data compar-
recurrence rate, while the argument for observation and ing adjuvant versus no treatment have hampered decision-
deferred therapy is the lack of randomized studies indi- making. Choo et al.31 analyzed 125 patients with a
cating an advantage of one over the other. positive resection margin or pT3 disease of whom 73
The choice to administer some forms of adjuvant were treated with adjuvant radiation therapy and 52 were
therapy results form the high relapse rate. Sofer et al.22 followed expectantly. Radiation therapy was delivered a
assessed the outcomes of 812 men of whom 106 (13%) median of 3.4 months post-RP with a 4-field technique
were found to have seminal vesicle involvement follow- to a dose of 60 to 66 Gy. The clinical target volume
ing radical prostatectomy. At an average follow-up of 48 (CTV) was limited to the prostate bed, and if the seminal
months follow-up, 53% of the entire cohort were free of vesicles were pathologically involved, they were also
disease (PSA < 0.4 ng/ml). In an analysis of prognostic included in the CTV. The treatment volume included a
variables only patients with a PSA > 10 ng/ml showed a 1-cm margin around the CTV. A Cox proportional haz-
higher recurrence rate (70% versus 30%, p < 0.001). The ards model of relapse-free probability versus time
5-year likelihood of being disease free with seminal vesi- demonstrated adjuvant radiation therapy (0.22, p = 0.0008),
cle involvement was 35%. PSA preprostatectomy (1.022, p = 0.029) and seminal
Van den Ouden et al.23 performed radical prostatec- vesicle involvement (2.03, p = 0.09) as important vari-
tomy on a group of 83 men with T3 prostate cancer. Of ables. Unfortunately, there was no independent seminal
these patients, 64 (77%) were found with extracapsular vesicle analysis.
extension and/or growth into the seminal vesicles. At a Patients with seminal vesicle involvement are at risk
median follow-up of 52 months, 19% demonstrated local for local and systemic recurrence. The exact incidence of
recurrence and 57% PSA relapse. Actuarial biochemical each and whether local failure presages systemic relapse
progression at 5 years for the pT3G1-2 patients was 61% has never been fully defined. Gibbons et al.32 reported a
and for the pT3G3 group 100%. Van Poppel et al.24 44% local failure rate following radical prostatectomy in
described 110 men who had radical prostatectomy for T3 patients with seminal vesicle involvement. In a 15-year
prostate cancer. Five-year PSA-free survival for the follow-up study, up to 83% of patients have been
entire group was 40%. Lerner et al.25 reported on 812 reported with local recurrence if the radical prostatec-
patients with clinical T3 treated at the Mayo Clinic. Of tomy specimen contained pT3 disease.33 In looking for
the 812 patients, 290 (35.7%) had pT3c (seminal vesicle local recurrence, Medini et al.34 analyzed 40 men who
involvement) disease. PSA-free survival (>0.2 ng/ml) at 5 were found to have elevated PSA 9 to 96 months post-
years for the entire cohort was 41% and at 10 years was prostatectomy. Of the 40 patients, 25 had a positive sur-
30%. Ten-year cause specific for the pT3c group was gical margin and 6 had involvement of the seminal
60%. Several other reports of men who underwent radi- vesicles. Twenty-eight were found to have recurrent local
cal prostatectomy with seminal vesicle involvement disease as determined by transrectal biopsy.
demonstrate similar results26–30 (Table 33-2). Zeitman et al.35 performed a review of residual disease
Adjuvant radiation therapy has been used in the set- after radical surgery or radiation therapy in patients with
ting of positive margins and seminal vesicle involvement prostate cancer. He noted a 12% to 68% risk of local

Table 33-2 Comparison of Current Series of Patients with Seminal Vesicle Implant to Biochemical Freedom
from PSA Failure Following Radical Prostatectomy with Vesicle Involvement
Study Number Definition Rate (%) Years

Catalona and Bigg26 86 >0.6 32 5

D’Amico et al.27 39 >0.2 5 2

Kupelian et al.28 60 >0.2 20 6

Sofer et al.22 66 >0.4 35 5

Trapasso et al.29 93 >0.4 56 5

Zeitman et al.30 12 >0.2 4 4

Van den Ouden et al.23 83 2 increases above 0.1 29 5

Current 32 ASTRO 74 7
558 Part V Prostate Gland and Seminal Vesicles

Figure 33-2 A, Axial image showing two implant needles at “C1” and “c1” positions within
the seminal vesicles. The bladder with Foley balloon can be seen just above. Arrow points to
anterior rectal wall. B, Longitudinal image of implant needle in anterior seminal vesicle wall.
The bladder is above and to the left. The arrow points to the posterior wall of the SV. A Mick
applicator will be used to deposit two seeds, one proximal and one just above prostate base.

recurrence, which was associated with a poorer prognosis. prostate D90/SV2 D90 ranged from 19% to 88%
In a review of patients with T2 to T3 disease receiving (median 52%).
radiation therapy, between 27% and 100% were found A multimodality treatment program has been devel-
to have positive biopsies, including 27% treated with oped to address these high-risk prostate cancer patients
EBRT plus AU-198 seeds and 28% with EBRT plus with biopsy proven SV involvement.41,42 The protocol
I-125 seeds.36,37 consists of three parts: neoadjuvant and concomitant
Planning a radiation treatment field that adequately hormonal therapy, partial palladium-103 implant (plan-
boosts dose to encompass disease that has extended into ning dose 83 to 90 Gy) with seeds placed in the prostate
the seminal vesicles has not been addressed. While dose and seminal vesicles followed 2 months later with 45 Gy
escalation trials have been performed on high-risk of conformal EBRT, which includes a 1.5-cm margin
prostate cancer patients and have demonstrated an around the prostate and seminal vesicles (Figure 33-3).
advantage to doses in excess of 75 Gy, no studies have High-risk patients undergo routine evaluation, which
specifically addressed these higher doses in the seminal includes bone and CT scanning. Seminal vesicle biopsies
vesicles.38,39 The problem in adequately targeting the identify those patients with vesicle involvement (Figure
seminal vesicles with the higher doses and sparing the 33-4A,B). Laparoscopy can exclude the 25% to 35%,
rectum, bladder base, and ureters has probably prevented who may harbor micrometastatic disease in the pelvic
an adequate treatment to this region. Centers that choose lymph nodes. Treatment is begun with 3 months of com-
to treat high-risk prostate cancer with a combination of plete androgen blockade. Preimplant prostate volume is
brachytherapy and beam irradiation risk under treatment determined with an extra 10 cc added to account for the
if the disease has extended into the seminal vesicles. seminal vesicles. The real-time method of seed implanta-
Stock et al.40 have shown that the implant provides very tion where total activity is identified by nomogram and
little radiation dose to the seminal vesicles if the implant planning done in the operating room is used.43–45 The
is limited to the prostate. The most proximal 20% of patient is brought to the OR and placed in the lithotomy
seminal vesicle tissue (SV1) received a median of 35% of position. The probe is advanced to the base of the bladder
the prostate dose while the next cephalad 20% (SV2) until the tips of the seminal vesicles are no longer visible.
received just 3%. With external radiation dose of 45 Gy The probe is then retracted until the seminal vesicles
in this situation, adequate treatment of the seminal vesi- appear under the bladder base. The seminal vesicles are
cle extension would be inadequate. In the same study, contoured at 5-mm intervals, which is continued when
Stock et al.40 demonstrated in a small cohort of 5 patients the prostate is reached to encompass both the prostate
with seminal vesicle tumor how to boost the SV dose base and seminal vesicles. The contouring is continued to
with an implant technique developed for this purpose the prostate apex. The physicist can then match these
(Figure 33-2A and B). The postimplant prostate D90/SV1 structures, as well as urethra and rectum, in order to per-
D90 ranged from 63% to 97% (median 80%) and the form the intraoperative planning (Figure 33-5A,B).
Chapter 33 Seminal Vesicles: Diagnosis, Staging, Surgery, and Management 559

Stage T3c prostate cancer

Laparoscopic pelvic lymph node dissection

Node negative Node positive

3 months LHRHa plus antiandrogen Hormonal therapy

partial Pd-103 implant to prostate and seminal vesicles


Dose 100 Gy (NIST 99)

2 month break
continue HT

45 Gy conformal EBRT to prostate and SV


continue HT till end of radiation

Figure 33-3 Flow chart of treatment protocol for high-risk prostate cancer patients with
biopsy proven seminal vesicle involvement.

Figure 33-4 A, Pretreatment biopsy: seminal vesicle with lamina propria extensively
infiltrated with prostatic adenocarcinoma (Gleason’s pattern 4 + 4, total score 8). The
seminal vesicle epithelium shows mild nuclear pleomorphism and atypia, which is a normal
degenerative finding (arrow) (H/E, 200×). B, Posttreatment biopsy: seminal vesicle that is
negative for prostatic adenocarcinoma. Focally, the seminal vesicle glands contain
intraluminal secretions (arrow). Endothelium in a small vessel shows nuclear atypia
consistent radiation effect (arrowhead) (H/E, 200×).

Once the intraoperative planning is complete, the sources. With the use of intraoperative planning software
peripheral needles are placed. The implant is performed (VariSeed 7.0, Varian, Palo Alto, CA) in an interactive
in two phases, with placement of the peripheral needles fashion, the plan continually evolves as each seed is
and sources first, followed by the interior needles and placed. The Mick Applicator (Mick Radionuclear
560 Part V Prostate Gland and Seminal Vesicles

Figure 33-5 A, Axial image from planning computer (VariSeed, Varian, Palo Alto, CA)
demonstrating intended needle and seen positions in seminal vesicles with isodose
contours superimposed. Center isodose contour represents 100% of prescription (100 Gy
palladium-103). B, 3D representation of completed implant showing the prescription dose
cloud covering the prostate and proximal seminal vesicles.
Chapter 33 Seminal Vesicles: Diagnosis, Staging, Surgery, and Management 561

Figure 33-6 A, Postimplant CT image of seminal vesicle seed implant with bladder above
rectum below. The 80% and 100% isodose contours encompass the vesicles with very little
dose distributed to bladder or rectum. B, Coverage of SV at base of prostate.

Instruments, Mount Vernon, NY) permits each seed to vesicle doses and to aid in the external beam planning
be placed individually, maximizing the ability to conform (Figure 33-6A,B). Two months postimplant 45 Gy of
the radiation dose cloud to the prostate and seminal vesi- conformal external beam is given, limited to the prostate
cles (see Figure 33-5B). and seminal vesicles with a margin of 1.5 cm. The hor-
Postimplant dosimetry is performed 1 month after the monal therapy is continued till the end of the external
implant is used in part to confirm prostate and seminal beam (total length of time 9 months). Routine prostate
562 Part V Prostate Gland and Seminal Vesicles

100 100
90
Percent free from PSA failure

Percent free from PSA failure


80
70
60
50
Gleason 8−10
40
30 Gleason −7
20
Survival Function
Censored 10 p = 0.128 Gleason 4−6
0
0 3 5 8 0
0 3 5 8
Years
Years
Figure 33-7 Kaplan-Meier estimates of PSA freedom from
Figure 33-8 PSA freedom from failure for Gleason score 4 to
failure defined as being free from 3 consecutive PSA rises
6 (100%), 7 (67%), and 8 to 10 (60%). Although the
above a nadir (ASTRO). Of the 32 patients treated, 8 have
differences were not significant, there does appear to be
failed. The 7-year freedom from failure is 74%. The median
worse outcome for patients with Gleason score 8 to 10 in the
PSA for these 24 is <0.1 ng/ml.
seminal vesicles.

and seminal vesicle biopsies were performed 2 years after a continued state of androgen deprivation. Only one
completion of treatment (see Figure 33-4A,B). Biopsies patient still had a testosterone level below 50 ng/dl while
were repeated in the case of rising PSA. To date, no the rest had values above 200. Therefore, it is not likely
patients have demonstrated evidence of local recurrence. that these favorable results are a reflection of persistent
The results of this multimodality approach have been hormonal therapy effects. It is likely that the short-
highly effective and safe.41,42 An update of previously term hormonal therapy benefits these patients, probably
published data continues to demonstrate the efficacy of from a combination of cytoreduction during the neoad-
this approach in 32 men with biopsy confirmed seminal juvant phase and additive effects during the treatment
vesicle involvement followed a minimum of 3 years. phase. There is both animal model and clinical data to
Initial PSA ranged from 4 to 88 ng/ml (mean 22.2, support this concept.46–49 There may be no benefit in
median 16.3), Gleason score was 4 to 6 in 9 (28.1%), 7 in continuing the hormone therapy beyond the end of radi-
13 (40.6%), and 8 to 9 in 10 (31.3%). Clinical stage was ation if all of the local tumor can be eradicated with the
T1c to T2a in 3 (9.4%), T2b in 3 (9.4%), T2c in 18 shorter-term treatment and high radiation doses.
(56.2%), and T3 in 8 (25%). Follow-up was a mean of 5 Testosterone production will recover in most men
years (2.5 to 8). PSA failure was defined as three consec- treated with luteinizing hormone releasing hormone
utive rises above the nadir (ASTRO). Eight patients have agonists if given for less than a year.49 In contrast, longer-
met the criteria for failure yielding a 7-year actuarial term administration of these agents is likely to result in
freedom from biochemical failure of 74% (Figure 33-7). permanent hypogonadism. The improvement in results
Neither presenting PSA nor stage predicted for bio- with the multimodality approach cannot be explained on
chemical outcome. There was a trend for higher Gleason the basis of prolonged hormonal therapy usage noted in
score and worse outcome (Figure 33-8). trials where androgen deprivation has been administered
Overall morbidity with this regimen has been accept- for a prolonged period.49 In fact, almost all of the patients
able. No patients have experienced a grade 3 or grade 4 rec- had a return in serum testosterone levels, suggesting that
tal injury. One patient (3%) with a history of prior TURP persistent androgen deprivation does not explain these
has minor stress incontinence. Testosterone levels have favorable results. It is likely that this regimen eradicates
returned to above 200 ng/dl in the majority of patients. all of the local disease, which was evident by the negative
The data from the multimodality approach suggest biopsies, especially in those patients with PSA progres-
that there may be an advantage with this approach over sion. In contrast, when seminal vesicle involvement was
radical prostatectomy (see Table 33-2). There are several found in the prostatectomy specimen, local relapse was
possible explanations for these favorable results. The quite common.22–29 The difference in PSA-free relapse
implant patients were treated with 9 months of hormonal between these data and the prostatectomy data implies
therapy and the current PSA readings might result from that difference may be accounted for by the persistence
Chapter 33 Seminal Vesicles: Diagnosis, Staging, Surgery, and Management 563

of residual adenocarcinoma following an inadequate dissection in the men with localized carcinoma of the
prostate resection. It is not unreasonable to believe that prostate. J Urol 1995; 154:1393–1396.
residual disease would remain following prostatectomy in 14. Terris MK, McNeal JE, Freiha FS, et al: Efficacy of
patients with seminal vesicles involvement. transrectal ultrasound-guided biopsies in the detection of
seminal vesicle invasion by prostate cancer. J Urol 1993;
149:1035–1039.
SUMMARY 15. Wymenga LF, Duisterwinkel FJ, Groenier K, Mensink
HJ: Ultrasound-guided seminal vesicle biopsies in prostate
Primary tumors of the seminal vesicles are uncommon. cancer, Prostate Cancer Prostatic Dis 2000; 3:100–106.
Extension of prostate cancer into the vesicles should be 16. Okihara K, Kamoi K, Lane RB, et al: Role of systematic
identified prior to consideration for definitive therapy. A ultrasound-guided staging biopsies in predicting
monotherapy approach to management of this disease extraprostatic extension and seminal vesicle invasion in
will most likely fail. A combined therapeutic approach men with prostate cancer. J Clin Ultrasound 2002;
either with radiation or surgery should be considered for 30:123–131.
this difficult disease. 17. Pandey P, Fowler JE, Seaver LE, et al: Ultrasound guided
seminal vesicle biopsies in men with suspected prostate
cancer. J Urol 1995; 154:1798–1801.
18. Roach M, Chen A, Song J, et al: Pretreatment
References
prostate-specific antigen and Gleason score predict
the risk of extracapsular extension and the risk of
1. Latchamsetty KC, Elterman L, Coogan CL: Schwannoma failure following radiotherapy in patients with clinically
of a seminal vesicle. Urology 2002; 60:515. localized prostate cancer. Semin Urol Oncol 2000;
2. Iqbal N, Zins J, Klienman GW: Schwannoma of the 18:108–114.
seminal vesicle. Conn Med 2002; 66:259–260. 19. Penson DF, Grossfeld GD, Li YP, et al: How well does
3. Patel B, Gujral S, Jefferson K, et al: Seminal vesicle cysts the Partin nomogram predict pathological stage after
and associated anomalies. BJU Int 2002; 90:265–271. radical prostatectomy in a community based population?
4. Berger AP, Bartsch G, Horninger W: Primary Results of the cancer of the prostate strategic urological
rhabdomyosarcoma of the seminal vesicle. J Urol 2002; research endeavor. J Urol 2002; 167:1653–1657.
168:643. 20. D’Amico AV, Whittington R, Malkomicz SB, et al:
5. Yanagisawa N, Saegusa M, Yoshida T, Okayasu I: Critical analysis of the ability of the endorectal coil
Squamous cell carcinoma arising from a seminal vesicular magnetic imaging scan to predict pathologic stage, margin
cyst: possible relationship between chronic inflammation status, and post-operative prostate specific antigen failure
and tumor development. Pathol Int 2002; 52:249–253. in patients with clinically organ confined prostate cancer.
6. Tabata K, Irie A, Ishii D, et al: Primary squamous J Clin Oncol 1995; 14:1770–1777.
cell carcinoma of the seminal vesicle. Urology 2002; 59:445. 21. Cornud F, Flam T, Chauveinc L, et al: Extraprostatic
7. Linzer DG, Stock RG, Stone NN, et al: Seminal vesicle spread of clinically localized prostate cancer: factors
biopsy: accuracy and implications for staging of prostate predictive of pT3 tumor and of positive endorectal MR
cancer. Urology 1996; 48:757–761. imaging examination results. Radiology 2002;
8. Peabody JO, Korman HJ, Amin MB: Miscellaneous 224:203–210.
tumors of the genitourinary tract. In Vogelzang NJ, 22. Sofer M, Savoie M, Kim SS, et al: Biochemical and
Scardino PT, Coffey DS (eds): Comprehensive Textbook pathological predictors of the recurrence of prostatic
of Genitourinary Oncology, pp 1152–1158. Baltimore, adenocarcinoma with seminal vesicle invasion. J Urol
Williams and Wilkens, 1996. 2003; 169:153–156.
9. Mukamel E, deKernion JB, Hannah J, et al: The 23. Van den Ouden D, Hop WCJ, Schroder FH: Progression
incidence and significance of seminal vesicle invasion in and survival of patients with locally advanced prostate
patients with adenocarcinoma of the prostate. Cancer caner (T3) treated with radical prostatectomy as
1987; 59:1535–1538. monotherapy. J Urol 1998; 160:1392–1397.
10. Vallencien G, Bochereau G, Wetzel O, et al: Influence of 24. Van Poppel H, Goethuys H, Callewaert P, et al:
preoperative positive seminal vesicle biopsy on the staging Radical prostatectomy can provide a cure for well
of prostate cancer. J Urol 1994; 152:1152–1156. selected clinical stage T3 prostate cancer. Eur Urol
11. Allepuz Lossa C, Sanz A, Velez JI, et al: Seminal vesicle 2000; 38:372–379.
biopsy in prostate cancer staging. J Urol 1995; 25. Lerner SE, Blute MI, Zinke H: Extended experience with
154:1407–1411. radical prostatectomy for clinical stage T3 prostate
12. Stock RG, Stone NN, Ianuzzi C, et al: Seminal vesicle cancer: outcome and contemporary morbidity. J Urol
biopsy and laparoscopic pelvic lymph node dissection. 1995; 154:1447–1452.
Implications for patient selection in the radiotherapeutic 26. Catalona WJ, Bigg SW: Nerve-sparing radical
management of prostate cancer. Int J Radiat Oncol Biol prostatectomy: evaluation of results after 250 patients.
Phys 1995; 33:815–821. J Urol 1990; 143:538–544.
13. Stone NN, Stock RG, Unger P: Indications for seminal 27. D’Amico A, Whittington R, Malkowicz S: A multivariate
vesicle biopsy and laparoscopic pelvic lymph node analysis of clinical and pathological factors that predict for
564 Part V Prostate Gland and Seminal Vesicles

prostate specific antigen failure after radical prostatectomy 39. Hanks GE, Hanlon AL, Epstein B, et al: Dose response in
for prostate cancer. J Urol 1995; 154:538–548. prostate cancer with 8–12 years follow-up. Int J Radiat
28. Kupelian P, Kathcher J, Levin H, et al: Correlation of Oncol Biol Phys 2002; 54:427–435.
clinical and pathologic factors with rising prostate-specific 40. Stock RG, Lo YC, Gaildon MS, Stone NN: Does
antigen profiles after radical prostatectomy alone for prostate brachytherapy treat the seminal vesicles? A dose-
clinically localized prostate cancer. Urology 1996; response histogram analysis of seminal vesicles in patients
48:249–260. undergoing combined Pd-103 prostate implantation and
29. Trapasso JG, deKernion JG, Smith RB, et al: The external beam irradiation. Int J Radiat Oncol Biol Phys
incidence and significance of detectable levels of serum 1999; 45:385–389.
PSA after radical prostatectomy. J Urol 1994; 41. Stone NN, Stock RG: Prostate brachytherapy: treatment
152:1816–1825. strategies. J Urol 1999; 162:421–426.
30. Zeitman AL, Edelstein RA, Coen JJ, et al: Radical 42. Stock RG, Stone NN: Preliminary toxicity and prostate-
prostatectomy for adenocarcinoma of the prostate: the specific antigen response to a phase I/II trial of
influence of preoperative and pathologic findings on neoadjuvant hormonal therapy, Pd-103 brachytherapy,
biochemical disease free outcomes. Urology 1994; and 3D conformal external beam irradiation in the
43:828–833. treatment of locally advanced prostate cancer.
31. Choo R, Hruby G, Hong J, et al: Positive resection Brachytherapy 2002; 1:2–10.
margin and/or pathologic T3 adenocarcinoma of 43. Stone NN, Hong S, Lo YC, et al: Comparison of
prostate with undetectable postoperative prostate- intraoperative dosimetric implant representation to
specific antigen after radical prostatectomy: to post-implant dosimetry in patients receiving prostate
irradiate or not? Int J Radiat Oncol Biol Phys 2003; 52: brachytherapy. Brachytherapy 2003; 2:17–25.
674–680. 44. Stone NN, Stock RG: Brachytherapy for prostate cancer:
32. Gibbons RP, Cole BS, Richardson G, et al: Adjuvant Real time three dimensional interactive seed implantation.
radiotherapy following radical prostatectomy: results and Tech Urol 1995; 1:72–80.
complications. J Urol 1986; 135: 65–68. 45. Stock RG, Stone NN, Lo YC, et al: Post-implant
33. Jewett HJ, Eggleston JC, Yawn DH: Radical dosimetry for I-125 prostate implants: definitions and
prostatectomy in the management of carcinoma of the factors affecting outcome. Int J Radiat Oncol Biol Phys
prostate: probable causes of some therapeutic failures. 2000; 48: 899–906.
J Urol 1972; 107:1034–1040. 46. Cahlon O, Stock RG, Kollmeier M, Stone NN:
34. Medini E, Medini I, Reddy PK, Levitt SH: Hormonal therapy, brachytherapy and external beam
Delayed/salvage radiation therapy in patients with irradiation in the treatment of high risk prostate cancer,
elevated prostate specific antigen levels after radical post-treatment and PSA outcomes. Bracytherapy 2003;
prostatectomy. Cancer 1996; 78:1254–1259. 2:P-72.
35. Zeitman AL, Shipley WU, Willett CG: Residual disease 47. Zeitman AL, Nakfoor BM, Prince EA, et al: The effect of
after radical surgery or radiation therapy for prostate androgen deprivation and radiation therapy on an
cancer. Cancer 1993; 71:959–969. androgen-sensitive murine tumor: an in vitro and in vivo
36. Scardino PT, Wheeler TM: Local control of study. Cancer J Sci Am 1997; 3:31–36.
prostate cancer with radiotherapy: frequency and 48. Pilepich MV, Winter K, John MJ, et al: Phase III
prognostic significance of positive results of radiation therapy oncology group (RTOG) trial 86-10 of
postirradiation prostate biopsy. Monogr Natl Cancer androgen deprivation adjuvant to definitive radiotherapy
Inst 1988; 7:95–103. in locally advanced carcinoma of the prostate. Int J Radiat
37. Schellhammer PF, El-Mahdi AM, Higgins AM, et al: Oncol Biol Phys 2001; 50:1243–1252.
Prostate biopsy after definitive treatment by interstitial 49. Bolla M, Collette L, Blank L, et al: Long-term results
125 iodine implant or external beam radiation therapy. with immediate androgen suppression and external
J Urol 1987; 137:897–901. irradiation in patients with locally advanced prostate
38. Pollack A, Zagers GK, Starkschall G: Prostate cancer cancer (an EORTC study): a phase III randomized trial.
radiation dose response: results of the M.D. Anderson Lancet 2002; 360:103–106.
phase II randomized trial. Int J Radiat Oncol Biol Phys
2002; 53:1097–1105.
C H A P T E R

34 Testis Tumors: Diagnosis


and Staging
Avrum Jacobson, MDCM, and Paul H. Lange, MD

DIAGNOSIS
such cases urgent referral to a specialist is crucial. In
Introduction
addition, physicians must understand that GCTs are high
Testicular germ cell tumors (GCTs) typically strike men among the differential diagnoses for a retroperitoneal
in the prime of their lives. It is the most common malig- mass of unknown primary in a young man. All such men
nancy in men aged 20 to 35, and the second most com- should, at the very least, undergo a thorough genital
mon malignancy in men aged 35 to 39.1 On a positive examination and have testicular serum markers drawn.
note, however, GCTs have become a paradigm of the
potential of translational cancer research. Advancements
Presentation
in imaging, tumor markers, surgical techniques, and most
importantly, the introduction of cisplatin-based The most common presentation of GCTs is that of a
chemotherapy have drastically impacted morbidity and painless testicular mass. However, although only 10%
mortality, improving overall survival from 10% in the present with an acute painful scrotum,6 some pain has
1970s to 90% today.2 Despite these advances, 20% of been reported in up to 32%.7 Neither pain nor testicular
patients who present with metastasis still succumb to tenderness should not dissuade one from making the
their disease.3 diagnosis of malignancy. A history of trauma should also
Delay in diagnosis continues to be a major concern. not decrease the index of suspicion, as 4% of GCT
Bosl et al.4 reported a median delay from onset of symp- patients give such a history.7 It is possible that either the
toms to pathologic diagnosis of 85 days. In 285 cases ini- trauma draws their attention to their testicular abnor-
tially seen by primary care physicians, only 56% were mality, or that the tumor increases the sensitivity of the
suspected of malignancy, despite testicular signs being testis to a degree of trauma that would otherwise not
noted in 92%. A British study reported even more have brought the patient to medical attention.
abysmal results with a mean delay in presentation to Occasionally, pain prevents a proper assessment of the
medical care of 5.5 months.5 In both studies, delay was testis. If, in such cases, epididymitis or orchitis is diag-
associated with higher stage, and in the later study, mor- nosed, the patient should be treated and followed very
tality doubled from 8% to 16% if delay was greater than closely for reexamination. If malignancy can still not be
6 months.4,5 In our own institution, we continue to see ruled out, an ultrasound should be obtained. In the vast
rare young men referred by outside physicians who have majority of cases, however, the key to diagnosis is a well-
been diagnosed by retroperitoneal mass biopsy without executed genital examination.
ever having had a testicular examination. Clearly, there is The physical examination of the testes is best per-
a great need for educational programs. The general pub- formed with the patient standing and the physician
lic should be indoctrinated in the need for regular self- seated facing him. The testis is palpated between the
examination of the testis, as well as periodic physical thumb and the first 2 fingers. The normal testis should
examination performed by a physician. The physicians be examined first to establish a baseline. The normal
must be aware that any mass within the testicle should be testis should be homogeneous, freely mobile, and distinct
considered malignant until proven otherwise, and that in from the epididymis and other cord structures. Any area

567
568 Part VI Testis

within the testis itself, which appears fixed, firm, or even when histology demonstrates only pure seminoma,
nodular, should be considered highly suspicious. the patient should be treated according to NSGCT pro-
Seventy percent of GCTs are discovered by self-exam- tocols. AFP elevation has been associated with other
ination and an additional 15% are discovered by physi- malignant and benign processes. These include pancre-
cian examination.7 Patients may also seek attention for atic, gastric, and lung cancer,12 as well as liver disease,
symptoms of disseminated disease. At diagnosis, 50% are pregnancy, ataxia telangiectasia, and tyrosenemia.12,13
found to have metastasis; signs and symptoms of these, However, in the clinical context of GCTs, these other
however, are found in approximately 10%.4 Patients may diagnosis are rarely a source of confusion.
complain of generalized weakness, fatigue, and weight HCG is a biologically active hormone, secreted by the
loss. Abdominal pain, back pain, GI disturbances, or placental syncytiotrophoblasts, which is responsible for
lower extremity edema can arise secondary to retroperi- maintaining the corpus luteum. Elevated serum concen-
toneal adenopathy, and pulmonary metastasis may mani- trations have been reported in hepatic, pancreatic, gas-
fest as dyspnea, cough, or hemoptysis. Manifestations of tric, pulmonary, breast, renal, and bladder tumors, as well
other sites of metastasis include headaches, seizures, or as multiple myeloma.12 Levels >10,000 IU/l, however,
other palpable masses, most commonly in the left supra- are seen almost exclusively in pregnancy, gestational dis-
clavicular area. Other presenting symptoms include orders, or GCTs.14 Forty percent to 60% of patients with
infertility and a 3% incidence of gynecomastia,7 which is GCTs will have an elevated hCG. This includes essen-
believed to be secondary to an estrogen/testosterone tially all patients with choriocarcinoma, 80% of those
imbalance caused by the effect of human chorionic with embryonal carcinoma, and 10% to 25% of those
gonadotropin (hCG) on Leydig cells. with seminoma.10
LDH is elevated in a wide spectrum of illnesses and,
thus in GCTs, LDH has not been shown to have a strong
Serum Tumor Markers
predilection for certain histologies. Its usefulness in diag-
Tumor markers can be considered to be biologic attrib- nosis is therefore somewhat limited. Nonetheless, LDH
utes of malignant cells that help to distinguish them from has been shown to reflect tumor burden15 and be an
normal cells. These may be uniquely identified with cer- important indictor of prognosis.16,17 Routine LDH levels
tain neoplasms or may be normal constituents, which, in are therefore recommended in all GCT patients.18,19
the face of a tumor, are expressed in abnormal locations Other markers, which have been proposed for GCTs,
and/or quantities, or display abnormal functions. In include placental alkaline phosphatase (PLAP) and
GCTs, perhaps more than any other tumor, the serum neuron-specific enolase (NSE). PLAP, although widely
tumor markers, including α-fetoprotein (AFP), hCG, accepted as a reliable histologic marker for seminoma,
and lactate dehydrogenase (LDH), play a key role in all has largely been abandoned as a serum marker. Although
aspects of clinical management. Their role in diagnosis is elevated in 50% to 72% of seminoma,20–22 specificity is
perhaps best illustrated in the 5% to 7% of GCTs that poor. Elevated levels can be detected in smokers and in
present with extragonadal primaries.8 These lesions often other malignancies, including lung, ovary, breast, and
present as poorly differentiated carcinomas of unknown gastrointestinal.23,24 Even among nonsmokers, positive
origin; positive immunohistochemical staining for AFP predictive value has been shown to be less than 50%.24
or hCG, or elevated serum levels, can confirm the diag- NSE, which has been found to be a useful marker in neu-
nosis. In testicular primaries, serum markers retain a crit- roendocrine tumors, initially showed promise as a marker
ical diagnostic role. Nonseminomatous GCTs (NSGCT) in GCTs, especially seminoma. Further investigation,
present with elevated AFP or hCG in 85% of cases (AFP however, failed to demonstrate clinical utility25, and
alone in 40%, hCG alone in 50% to 60%).9 Seminoma, investigational interest has waned.
on the other hand, by definition, never presents with ele-
vated AFP, but detectable hCG is present in 10% to 25%
Imaging
of cases.10 In the context of a testicular mass, elevated
markers are practically pathognomonic, and can, to a Scrotal ultrasound (U/S) is the current standard for testis
degree, predict histology. imaging. It is essential for suspicious (as opposed to def-
AFP is the dominant serum protein of the early inite) testicular masses found on physical examination
embryo and although detectable at birth, levels fall by and also for those patients in whom physical examination
age 1 to the minimal level seen in adults (<10 mg/dl). is limited either by pain or by a hydrocele. In addition, in
Trophoblastic cells, which are present in embryonal car- patients who present with extragonadal GCTs, U/S may
cinoma, teratocarcinoma, and yolk sac tumor, are respon- reveal an occult testis primary. U/S is generally easy to
sible for its production. These elements are never present obtain, noninvasive, inexpensive, and accurate. Newer
in pure choriocarcinoma or seminoma.11 This has impor- high frequency transducers (5 to 10 MHz) can detect
tant ramifications, as in the presence of a positive AFP, lesions as small as 1 to 2 mm. U/S can also distinguish
Chapter 34 Testis Tumors: Diagnosis and Staging 569

cystic from solid masses with 100% accuracy and Thus, in general, MRI provides good imaging of testicu-
intratubular from extratubular testicular with 99% accu- lar neoplasms but has not been shown to have any advan-
racy.26 Cyst can be seen within the tunica albuginea but tage over the cheaper and more easily accessible scrotal
are uncommon, especially among younger men, within ultrasound, an exception to this is possibly cryptorchid
the testis itself. In men older than 60, testicular cysts can (especially intraabdominal) testes.
be seen in 8% to 10%; these occur near the mediastinum
at the superior/lateral aspect of the testis and must fulfill
STAGING
all criteria of simple cyst.26 Solid lesions within the testes,
Introduction
that are malignant, cannot be distinguished absolutely
from those that are benign for reasons that we will now Prior to 1997, there was no consensus on staging of tes-
discuss. ticular GCTs. Most staging systems were based on the
The normal testis is uniformly heterogeneous on U/S. model introduced by Boden and Gibb31 in 1951, which
Seminomas typically appear as well-defined hypoechoic divided the disease into 3 stages: I, limited to the testis,
masses within the substance of the testis, without calcifi- II, spread to the retroperitoneum, and III, spread beyond
cation or cystic components. NSGCTs, on the other the retroperitoneum. Other systems generally kept this
hand, are typically inhomogeneous hypoechoic, normoe- basic format, subdividing stage II based on the size of the
choic, or hyperechoic masses, with a higher incident of retroperitoneal adenopathy. As well, different systems
cystic components and echogenic foci.27–29 U/S cannot were intended for seminoma and nonseminoma, and for
distinguish histology among NSGCT but has been shown clinical staging and pathologic staging.12,32–35 The
to distinguish correctly seminoma from nonseminoma in plethora of systems, each with its own subtleties led to
70% of cases.29 In addition, U/S does not provide accu- confusion and difficulty in comparing data from various
rate local staging information. The tunica albuginea is institutions. In 1997, the American Joint Committee on
poorly seen and even in seminoma where more precise Cancer (AJCC) introduced an internationally accepted
borders are present, only 44% of patients are staged accu- consensus classification.36 This system, which will be dis-
rately. In NSGCT, this figure drops to 8%.29 cussed in detail later, provided much needed uniformity.
Parenchymal changes may occur in infection,
inflammation, vascular insult, or trauma, which may
Staging Modalities
mimic a tumor on ultrasound. Infective changes, on
appropriate therapy, should improve within 2 weeks. Testis tumors generally spread initially through lym-
However, severe orchitis with microabscesses may not phatic channels, which follow the embryologic origin of
resolve and may appear identical to necrosis and hem- the testis to the retroperitoneum. The primary landing
orrhage. Small infarcts and intratesticular hematomas site of right testicular tumors is intraaortocaval, at the
may also mimic tumor and be slow to resolve.26 In level of the second lumbar vertebra, and the primary
these settings, one must use clinical judgment and landing site of left tumors is paraaortic (i.e., to the left
explore surgically if any doubt remains; keeping in side of the aorta), just below the renal hilum.37
mind that excision of a diseased benign testis is cer- Hematologic dissemination can also occur, and is mani-
tainly preferable than retaining tumor. fested most often as lung metastasis; however, more
The testis can also be well visualized with MRI. The advanced disease can present with involvement of the
normal testis displays homogeneous signal intensity on liver, bone, or brain. The AJCC recommends radi-
both T1 and T2 weighted images. The hallmark of a ographic assessment of the chest, abdomen, and pelvis.36
tumor is loss of high signal intensity on T2. On T1, in Routine bone scan and brain imaging are generally
contrast, tumors can be difficult to distinguish from nor- unnecessary unless clinically indicated, as these locations
mal parenchyma. After gadolinium, however, brisk het- are extremely rarely the sole metastatic site, and their
erogeneous enhancement may be seen, providing clear presence is often accompanied by symptoms. The cur-
visualization of the lesion.30 NSGCT are markedly het- rent standard for imaging the chest, abdomen, and pelvis
erogeneous and can be differentiated from seminoma in is CT scan of the abdomen and pelvis, and plain CXR.
almost all cases. Differentiating NSGCT from CT chest should be performed if the CXR is abnormal or
hematoma, and seminoma from benign lesions, however if CT abdomen reveals metastasis.
can be difficult or even impossible. Also, despite good Although CT scan has become the imaging modality
visualization of the tunica albuginea, local staging of choice for the retroperitoneum, the ideal lymph node
remains poor. Partial volume averaging and similar size criterion has not been firmly established. Using the
intensities between the tumor and tunica albuginea conventional, but arbitrary, cutoff of 1 cm, 22% to 44%
makes assessment of invasion difficult. Finally, the medi- of metastasis can be missed.38 As expected, as smaller
astinum testis can be difficult to see, and local tumor sizes are applied, sensitivity improves and specificity
extension into this structure is therefore inaccurate.30 worsens. Stomper et al.39 evaluated various size criteria;
570 Part VI Testis

at 5, 10, and 15 mm, sensitivity was found to be 88%, cal meandering collaterals often accompany obstruction
73%, and 58% respectively, and specificity was found to of the lymphatic system.44 Although it has been sug-
be 44%, 60%, and 76% respectively. A study out of Indiana gested that patients with stage I GCTs undergo lym-
University examined transaxial lymph node diameter as a phangiogram prior to initiating watchful waiting,43 in
continuous variable between 0 and 25 mm. They found 174 patients with a negative work up (including negative
that the highest overall accuracy was reached at a diame- CT and tumor markers), lymphangiogram found only
ter of 8 mm. At that level, sensitivity was 66.7% and 4% unsuspected metastases and another 2% had false
specificity was 94.4%. Separate analysis revealed that positive findings.45 Today, there is little role for lym-
enlarged nodes within the expected landing zone were phography in the management of testicular GCTs.
more likely to represent true metastasis. Using a cutoff of PET scan demonstrates uptake of the glucose analog
10 mm outside the predicted landing zone and 3 mm 18-fluoro-2 deoxyglucose, and relies on the fact that
within, a sensitivity and specificity of 90.7% and 53.3%, many tumors have a high rate of glycolysis. In GCTs,
respectively, were attained.40 however, PET scan has not been widely investigated. In
Other modalities, which have been utilized to evaluate a German study, PET slightly out performed CT scan in
the retroperitoneum, include ultrasound, MRI, lym- 50 GCT patients staged at initial diagnosis. Sensitivity
phography, positron emission tomography (PET) scan, was 87% versus 73%, with equal specificity of 94%. CT
and laparoscopic lymph node dissection. Ultrasound scanners in that study, however, were not standardized,
examination of the retroperitoneum is often limited and pick up on PET scan of small positive lymph nodes
either by bowel gas or by patient obesity. Even in cases was still poor.46 In another European study, PET scan
where a good quality examination can be obtained, ultra- upstaged 3 of 31 patients, although management
sound is markedly inferior to CT scan. Subtle lymph remained unchanged.47 At present, PET scanning for
node metastases are generally impossible to diagnose, initial staging remains mainly investigational, but clearly
and even nodes between 2 and 2.5 cm are accurately further study is warranted. Parenthetically, in assessing
found in only 60%.26 Ultrasound is generally uninforma- postchemotherapy residual masses, PET still holds some
tive unless bulky lymphadenopathy is present, and even promise for differentiating viable tumor, teratoma, and
then, CT is preferable for more precise anatomic infor- necrosis.
mation and for follow-up comparison. MR, on the other The gold standard for staging of the retroperitoneum
hand, provides excellent imaging of the retroperitoneum is a formal retroperitoneal lymph node dissection.
with overall accuracy of approximately 80%. Studies, Despite all available modalities, 23% to 30% of clinical
however, have shown no additional benefit over CT.41,42 stage I patients are found to harbor retroperitoneal dis-
One clear advantage is the ability of MR to distinguish ease.48–51 In high risk patients, this number increases to
lymph node from vessel in the absence of contrast. In approximately 50%.50,51 Recently, specialized centers
patients in whom intravenous contrast is ill advised, MR have attempted to lessen operative morbidity by per-
is a sound alternative. forming the dissection laparoscopically. As a staging pro-
Bipedal lymphangiogram has largely been replaced cedure, outcomes have been excellent, with, to our
with CT scan. Although imaging of retroperitoneal knowledge, no retroperitoneal recurrences.52,53 However,
nodes is fair, with a positive predictive value of 83% and although it is clear that open RPLND is therapeutic in
negative predictive value of 69%,43 lymphography is addition to diagnostic, the therapeutic benefit of laparo-
invasive and, as such, has inherent morbidity. scopic RPLND remains unproven. This question arises
Complications include fever, local pain, wound infection, because the laparoscopic groups have treated their stage
lymphadenitis, and pulmonary dysfunction. As well, reac- IIA patients with 2 cycles of chemotherapy, a practice
tive changes can increase difficulty of future surgery, and that is not common after RPLND, as after surgery alone
imaging above L2 is poor, as at that level many lymphatic these patients have been shown to have recurrence rates
vessels coalesce into the thoracic duct. The only advan- of only 8% to 20%.54 In our institution, we are convinced
tage of lymphography over other imaging is that abnor- that the laparoscopic dissection can exactly mimic the
mal architecture can be detected in the absence of open technique. Therefore, we do not routinely adminis-
enlargement. Normally the lymphatic sinusoids pick up ter adjuvant therapy for our stage IIA group but remain
contrast well, while follicles pick up very little. This pro- watchful to our results and other groups in that.
duces a characteristic appearance of homogeneously dis- Finally, what staging modality is best for chest imag-
tributed droplets. Replacement of a node by tumor can ing? Although CT is obviously more sensitive than CXR,
produce a filling defect, or compression of sinusoidal sys- this does not always equate with being a superior test.
tem can produce an inhomogeneous contrast scattered The issue with CT of the chest is that there is a high inci-
throughout the node, termed “foamy.” In addition, typi- dence in the general population of small lesions seen on
Chapter 34 Testis Tumors: Diagnosis and Staging 571

CT, which are entirely benign. The morbidity of finding Intrapelvic, external iliac, and inguinal nodes are consid-
such lesions can, at times, be considerable; either requiring ered regional only after scrotal or inguinal surgery prior
excision for pathologic diagnosis or leading to incorrect to presentation with a testicular tumor. All other nodes
staging and treatment. One study found that in are considered distant and are staged within the M clas-
42 patients, with a negative abdominal CT, only 4.4% sification. For pathologic staging, it is critical that the
had chest involvement. All were diagnosed on both CXR pathologist carefully examines and liberally samples the
and CT scan, while CT scan picked up 3 additional specimen, including cystic, fibrotic, hemorrhagic,
lesions were found to be benign. In patients with positive necrotic, and solid areas. The number of lymph nodes
abdominal CT scan, on the other hand, the rate of pul- involved with tumor should be recorded, as should any
monary metastasis was considerably higher at 40%. In evidence of extranodal involvement.
this group, CT chest picked up an additional 12.5% of
patients missed on CXR alone. In addition, 3 cases of
Distant Metastasis
extrapulmonary metastasis were identified as well.55
The International Germ Cell Cancer Collaborative
Group (IGCCCG), a panel of leading experts from
1997 American Joint Committee On Cancer Staging
around the world, retrospectively studied prognostic fac-
System
tors in 5662 patients with metastatic testicular GCT, all
The 1997 AJCC staging of testis tumors depends on the of whom received cisplatin (or carboplatin) containing
determination of T (tumor), N (node), M (metastases), chemotherapy regiments. Based on this work, such
and S (serum tumor markers), categories. Although the patients could be categorized into good, intermediate, or
TNM classification is commonly used among various poor prognostic groups. The presence of liver, bone,
malignancies, the S category is unique to testis tumor and brain, or other nonpulmonary visceral metastasis, was
is a reflection of the importance of serum markers in the shown, in NSGCT, to be a poor prognostic factor with a
management of this disease. 5-year survival rate of 47%. In seminoma, although no
poor risk group was found, the most important prognostic
factor was again the presence of nonpulmonary visceral
Primary Tumor
metastases, placing patients in the intermediate prognosis
Histologic evaluation of the radical orchiectomy speci- group with a 5-year survival of 72% (Table 34-3).57 This
men is required for the T classification. The tumor information was integrated into the staging system by
should be sampled extensively, including all grossly subdividing the M1 category into M1a and M1b (see
diverse areas, as well as the junction of tumor and the Table 34-1).
nonneoplastic testis, and at least one section remote from
the tumor. Whenever possible, sections should include
Serum Tumor Markers
overlying tunica albuginea.
New to this system, is the introduction of lymphvas- For staging purposes, AFP, hCG, and LDH should be
cular invasion into staging (Table 34-1). This modifica- performed immediately after orchiectomy and, if ele-
tion assists the clinician to better differentiate stage I vated, repeated serially after orchiectomy to assess for
patients at high risk of harboring undetected retroperi- persistent elevation. In the absence of disease, markers
toneal metastasis (stage IB) from those at lower risk should decline according to their half-lives: <7 days for
(stage IA) (Table 34-2); if lymphvascular invasion is pres- AFP and <3 days for hCG. The subdivisions of the S
ent, 50% are found on RPLND to be understaged stage category (see Table 34-1) were adapted from the
II, while if lymphvascular invasion is absent, this drops to IGCCCG,57 and play an extremely important role, not
only 18% to 23%.49,56 Another factor also commonly only in prognosis, but also in the management of testic-
used in the same fashion is the percent embryonal carci- ular GCTS. Patients with no other evidence of metastatic
noma in the specimen; however, pathologic tumor type is disease with persistent marker elevation after orchiec-
inherently not a staging factor, and indeed, is not tomy are labeled stage 1S (see Table 34-2). These
included in the TNMS system. patients are almost always found to have disseminated
disease and are better treated with chemotherapy than
surgery.58 Patients with S2 or S3 marker levels with evi-
Regional Lymph Nodes
dence of only regional nodal disease are upstaged to
Interaortocaval, paraaortic, paracaval, preaortic, precaval, stage III, and again are better treated with chemother-
retroaortic, and retrocaval lymph nodes, as well as nodes apy, and, in the case of S3, perhaps even with more
along the spermatic vein, are considered regional. aggressive regiments.
572 Part VI Testis

Table 34-1 2002 AJCC TNMS Staging Classification


Primary Tumor (pT) Pathologic

Clinical pNX Regional lymph nodes cannot be assessed

Tumor stage is generally determined after pN0 No regional lymph node metastasis
orchiectomy at which time a pathologic stage is
assigned pN1 Metastasis with a lymph node mass 2 cm or less
in greatest dimension and less than or equal to 5
Pathologic nodes positive, none more than 2 cm in greatest
dimension
pTX Primary tumor cannot be assessed (if no radical
orchiectomy has been performed, TX is used) pN2 Metastasis with a lymph node mass more than
2 cm but not more than 5 cm in greatest
pT0 No evidence of primary tumor (e.g., histologic dimension; or more than 5 nodes positive, none
scar in testis) more than 5 cm; or evidence of extranodal
extension of tumor
pTis Intratubular germ cell neoplasia (carcinoma in
situ) pN3 Metastasis with a lymph node mass more than
5 cm in greatest dimension
pT1 Tumor limited to testis and epididymis without
vascular/lymphatic invasion; tumor may invade Distant Metastasis (M)
into tunica albuginea but not into tunica
vaginalis MX Distant metastasis cannot be assessed

pT2 Tumor limited to the testis and epididymis with M0 No distant metastasis
vascular/lymphatic invasion, or tumor extending
through the tunica albuginea with involvement M1 Distant metastasis
of the tunica vaginalis
M1a Nonregional nodal or pulmonary metastasis
pT3 Tumor invades the spermatic cord with or
without vascular/lymphatic invasion M1b Distant metastasis other than to nonregional
lymph nodes and lungs
pT4 Tumor invades the scrotum with or without
vascular/lymphatic invasion Serum Tumor Markers (S)

Regional Lymph Nodes (N) SX Markers studies not available or not performed

Clinical S0 Marker study levels within normal limits

NX Regional lymph nodes cannot be assessed S1 LDH < 1.5 × N and

N0 No regional lymph node metastasis hCG (mIU/ml) < 5000 and

N1 Metastasis within a lymph node mass 2 cm or AFP (ng/ml) < 1000


less in greatest dimension; or multiple lymph
S2 LDH 1.5 − 10 × N or
nodes, none more than 2 cm in greatest
dimension HCG (mIU/ml) 5000 − 50,000 or
N2 Metastasis with a lymph node mass more than AFP (ng/ml) 1000 − 10,000
2 cm but not more than 5 cm in greatest
dimension; or multiple lymph nodes, any one S3 LDH > 10 × N or
mass greater than 2 cm but not more than 5 cm
in greatest dimension HCG (mIU/ml) > 50,000 or

N3 Metastasis with a lymph node mass more than AFP (ng/ml) > 10,000
5 cm in greatest dimension
Chapter 34 Testis Tumors: Diagnosis and Staging 573

Table 34-2 Staging Classification (Stage Groups)


Stage grouping

Stage 0 pTis N0 M0 S0

Stage I pT1-4 N0 M0 SX

Stage IA pT1 N0 M0 S0

Stage IB pT2-4 N0 M0 S0

Stage IS Any pT/Tx N0 M0 S1-3

Stage II Any pT/Tx N1-3 M0 SX

Stage IIA Any pT/Tx N1 M0 S0-1

Stage IIB Any pT/Tx N2 M0 S0-1

Stage IIC Any pT/Tx N3 M0 S0-1

Stage III Any pT/Tx Any N M1 SX

Stage IIIA Any pT/Tx Any N M1a S0-1

Stage IIIB Any pT/Tx N1-3 M0 S2

Stage IIIB Any pT/Tx Any N M1a S2

Stage IIIC Any pT/Tx N1-3 M0 S3

Stage IIIC Any pT/Tx Any N M1a S3

Stage IIIC Any pT/Tx Any N M1b Any S

Table 34-3 International Germ Cell Consensus Classification


Nonseminoma Seminoma

Good prognosis
Testis/retroperitoneal primary Any primary site

And And
No nonpulmonary visceral metastasis No nonpulmonary visceral metastasis

And And
Good markers Nomal AFP, any HCG, any LDH

AFP < 1000 ng/ml and

HCG < 5000 IU/l (1000 ng/ml) and

LDH < 1.5 upper limit of normal

5-year PFS 89% 5-year PFS 82%

5-year survival 92% 5-year survival 86%

Continued
574 Part VI Testis

Table 34-3—cont’d
Nonseminoma Seminoma

Intermediate prognosis

Testis/retroperitoneal primary Any primary site

And And
No nonpulmonary visceral metastasis Nonpulmonary visceral metastasis

And And
Intermediate markers Nomal AFP, any HCG, any LDH

AFP > 1000 ng/ml and <10,000 ng/ml or

HCG > 5000 IU/l and <50,000 IU/l or

LDH > 1.5 × N and <10 × N

5-year PFS 75% 5-year PFS 67%

5-year survival 80% 5-year survival 72%

Poor prognosis

Mediastinal primary

Or
Nonpulmonary visceral metastasis

Or
Poor markers

AFP > 10,000 ng/ml or

HCG > 50,000 IU/l or

LDH > 10 × upper limit of normal

5-year PFS 41%

5-year survival 48%

REFERENCES 7. Kennedy BJ, Schmidt JD Winchester DP, et al: National


survey of patterns of care for testis cancer. Cancer 1987;
1. Richie JP: Detection and treatment of testicular cancer. 60:1921–1930.
CA Cancer J Clin 1993; 43:151–175. 8. Bokemeyer C, Hartmann JT, Fossa SD, et al: Extragonadal
2. Richie JP: Advances in the diagnosis and treatment of germ cell tumors: relation to testicular neoplasia and
testicular cancer. Cancer Invest 1993; 11(6):670–675. management options. APMIS 2003; 111:49–63.
3. McCaffrey JA, Bajorin DF, Motzer RJ, et al: Risk 9. Small EJ, Torti FM: Testes. In Abeloff MD, Armitage JO,
assessment for metastatic testis cancer. Urol Clin NA Lichter AS, Niederhuber JE (eds) Abeloff Clinical
1998; 25(3):389–395. Oncology, 2nd edition. Philadelphia, PA, Churchill
4. Bosl GJ, Goldman A, Lange PH, et al: Impact of delay in Livingston, 2000.
diagnosis on clinical stage of testicular cancer. Lancet 10. Klein EA: Tumor markers in testis cancer. Urol Clin
1981; 2:970–973. North Am 1993; 20:67–73.
5. Nikzas D, Champion AE, Fox M: Germ cell tumours of 11. Javadpour N: Significance of elevated serum
testis: prognostic factors and results. Eur Urol alphafetoproteins (AFP) in seminoma. Cancer 1980;
18:242–247. 45:2166.
6. Richie JP, Steele GS: Neoplasms of the testis. In Walsh PC, 12. Richie JP: Neoplasms of the testis. In Walsh PC, Retik
Retik AB, Vaughan ED, Wein A (eds) Campbell’s Urology, AB, Stamey TA, Vaughan ED (eds). Campbell’s Urology,
8th edition. Philadelphia, PA, WB Saunders, 2002. 6th edition. Philadelphia, PA, WB Saunders, 1992.
Chapter 34 Testis Tumors: Diagnosis and Staging 575

13. Bloomer JR, Waldmann TA, McIntire KR, et al: Serum 33. Doornbos JF, Hussey DH, Johnson DE: Radiotherapy for
alpha-fetoprotein levels in patients with nonneoplastic pure seminoma of the testis. Radiology 1975;
liver disease. Gastroenterology 1973; 65:530. 116:401–404.
14. Bower M, Rustin GJS: Serum tumor markers and their 34. Ball D, Barrett A, Peckham MJ: The management
role in monitoring germ cell cancers of the testis. In of metastatic seminoma testis. Cancer 1982;
Vogelzang NJ, Scardino PT, Shipley WU, Coffey DS 50:2289–2294.
(eds): Comprehensive Textbook of Genitourinary 35. Crawford ED, Smith RB, DeKernion JB: Treatment of
Oncology, 2nd edition. Philadelphia, PA, Lippincott advanced seminoma with preradiation chemotherapy.
Williams and Wilkins, 1999. J Urol 1983; 129:752–756.
15. Boyle LE, Samuels ML: Serum LDH activity and 36. Fleming ID, Cooper JS, Henson DE, et al. (eds): Testis.
isoenzyme patterns in nonseminomatous germinal In American Joint Committee on Cancer (AJCC) Cancer
(NSG) testis tumors. Proc Am Soc Clin Oncol 1977; Staging Manual, 5th edition. Philadelphia, PA,
18:278. Lippincott-Raven, 1997.
16. Mencel PJ, Motzer RJ, Mazumdar M, et al: Advanced 37. Donohue JP, Zachary JM, Maynard BR: Distribution of
seminoma; treatment results, survival, and prognostic nodal metastasis in nonseminomatous testis cancer. J Urol
factor in 142 patients. J Clin Oncol 1994; 12:120–126. 1982; 128:315–320.
17. Stoter G, Bosl GJ, Droz JP, et al: Prognostic factors in 38. Hilton S, Herr HW, Teitcher JB, et al: CT detection of
metastatic germ cell tumors. Porg Clin Biol Res 1990; retroperitoneal lymph node metastasis in patients with
357:313–319. clinical stage I testicular nonseminomatous germ cell
18. National Comprehensive Cancer Network Practice cancer: assessment of size and distribution criteria. AJR
Guidelines in Oncology, Vol 1, Testicular Cancer. 1997; 169(2):521–525.
Rockledge, PA, 2003. 39. Stomper PC, Fung CY, Socincki MA, et al: Detection of
19. Laguna MP, Pizzocaro G, Klepp O, et al: EAU guidelines retroperitoneal metastasis in early stage nonseminomatous
on Testicular Cancer. Eur Urol 2001; 40:102–110. testicular cancer: analysis of different CT criteria. AJR
20. Albrecht W, Bonner E, Jeschke K, et al: PLAP as a 1987; 149:1187–1190.
marker for germ cell tumors. In Jones NG, Appleyard I, 40. Leibovitch I, Foster RS, Kopecky KK, et al: Improved
Harnden P, Joffe JK (eds): Germ Cell Tumours IV. accuracy of computerized tomography based clinical
London, John Libbey & Co, 1998. staging in low stage nonseminomatous germ cell cancer
21. Koshida K, Nishino A, Yamamoto H, et al: The role of using size criteria of retroperitoneal lymph nodes. J Urol
alkaline phosphatase isoenzymes as a tumor marker for 1995; 154:1759–1763.
testicular germ cell tumors. J Urol 1991; 146:57. 41. Ellis JH, Bies JR, Kopecky KK, et al: Comparison of
22. Lange PH, Millan JL, Stigbrand T, et al: Placental NMR and CT imaging in the evaluation of metastatic
alkaline phosphatase as a tumor marker for seminoma. retroperitoneal lymphadenopathy from testicular
Cancer Res 1982; 42:3244. carcinoma. J Comput Assist Tomogr 1984; 8(4):709–719.
23. Muensch HA, Maslow WC, Azama F, et al: Placental-like 42. Hogeboom WR, Hoekstra HJ, Mooyaart EL, et al:
alkaline phosphatase. Reevaluation of the tumor marker Magnetic resonance imaging of retroperitoneal lymph
with exclusion of smokers. Cancer 1986; 58:1689. node metastases of nonseminomatous germ cell tumours
24. Nielsen OS, Muntro AJ, Duncan W, et al: Is placental of the testis. J Surg Oncol 1993; 19:429–437.
alkaline phosphatase (PLAP) a useful marker for 43. Bussar-Maatz R, Weissbach L: Retroperitoneal lymph
seminoma? Euro J Cancer 1990; 26(10):1049–1054. node staging of testicular tumours. BJU 1993;
25. Gross AJ, Dieckmann KP: Neuron-specific enolase: a 72:234–240.
serum marker in malignant germ-cell tumors? Euro Urol 44. Casellino RA: Lymphography. In Pollack HM,
1993; 24(2):277–278. McClennan BL (eds): Pollack Clinical Urography, 2nd
26. Benson CB: The role of ultrasound in diagnosis and staging edition. Philadelphia, PA, WB Saunders, 2000.
of testicular cancer. Semin Urol 1988; 6(3):189–202. 45. Wishnow KI, Johnson DE, Tenney D: Are
27. Richie JP, Birnholz J, Garnick MB: Ultrasonography as a lymphangiograms necessary before placing patients with
diagnostic adjunct for the evaluation of masses in the nonseminomatous testicular tumors on surveillance?
scrotum. Surg Gyn Obs 1982; 154:695–698. J Urol 1989; 141:1133–1135.
28. Schwerk WB, Schwerk WN, Rodeck G: Testicular 46. Cremerius U, Wildberger JE Borchers H, et al: Does
tumors: prospective analysis of real-time US patterns and positron emission tomography using 18-fluoro-2-
abdominal staging. Radiology 1987; 164:369–374. deoxyglucose improve clinical staging of testicular cancer?
29. Marth D, Scheidegger J, Studer UE: Ultrasonography of Results of a study of 50 patients. Urology 1999;
testicular tumors. Urol Int 1990; 45(4):237–240. 54:900–904.
30. Oyen R, Verellen S, Drochmans A, et al: Value of MRI in 47. Hain SF, O’Doherty MJ, Timothy AR, et al:
the diagnosis and staging of testicular tumors. JBR BTR Fluorodeoxyglucose PET in the initial staging of germ
1993; 76:84–89. cell tumours. Eur J Nucl Med 2000; 27(5):590–594.
31. Boden G, Gibb R: Radiotherapy and testicular neoplasms. 48. Donohue JP, Thornhill JA, Foster RS: Retroperitoneal
Lancet 1951; 2:1195. lymphadenectomy for clinical stage A testis cancer
32. Maier JG, Sulak MH: Radiation therapy in malignant (1965–1989): modifications of technique and impact on
testis tumors. Cancer 1973; 32:1217–1226. ejaculation. J Urol 1993; 149:237–243.
576 Part VI Testis

49. Klepp O, Olsson AM, Henrikson H, et al: Prognostic 54. Richie JP, Kantoff PW: Is adjuvant chemotherapy
factors in clinical stage I nonseminomatous germ cell necessary for patients with stage B1 testicular cancer?
tumors of the testis: multivariate analysis of a J Clin Oncol 1991; 9:1393–1396.
prospective multicenter study. Swedish-Norwegian 55. See WA, Hoxie L: Chest staging in testis cancer patients:
testicular cancer group. J Clin Oncol 1990; imaging modality selection based upon risk assessment as
8:509–518. determined by abdominal computerized tomography scan
50. Albers P, Siener R, Kliesch S, et al: Risk factors for relapse results. J Urol 1993; 150:874–878.
in clinical stage I nonseminomatous testicular germ cell 56. Hoskin P, Dilly S, Easton D, et al: Prognostic factors in
tumors: results of the German testicular cancer study stage I nonseminomatous germ cell testicular tumors
group trial. J Clin Oncol 2003; 21:1505–1512. managed by orchiectomy and surveillance: implications for
51. Hermans BP, Sweeney CJ, Foster RS, et al: Risk of adjuvant chemotherapy. J Clin Oncol 1986; 4:1031–1036.
systemic metastases in clinical stage I nonseminomatous 57. The International Germ Cell Collaborative Group:
germ cell testis tumor managed by retroperitoneal lymph International germ cell consensus classification: a
node dissection. J Urol 2000; 163:1721–1724. prognostic factor-based staging system for metastatic
52. Janetschek G, Hobisch A, Peschel R, et al: Laparoscopic germ cell cancers. J Clin Oncol 1997; 15(2):594–603.
retroperitoneal lymph node dissection. Urology 2000; 58. Davis BE, Herr HW, Fair WR, et al: The management of
55:136–140. patients with nonseminomatous germ-cell tumors of the
53. Nelson JB, Chen RN, Bishoff JT, et al: Laparoscopic testis with serologic disease only after orchiectomy. J Urol
retroperitoneal lymph node dissection for clinical stage I 1994; 152:111–114.
nonseminomatous germ cell testicular tumors. Urology
1999; 54:1064–1067.
C H A P T E R

35Seminoma: Management
and Prognosis
Michael A. S. Jewett, MD, FRCSC, FACS,
Rishikesh Pandya, MCh, DNB, MS, and
Padraig Warde, MB, BCh, BAO

Testicular cancer is uncommon and accounts for only 1% lack of apparent serious morbidity with adjuvant RT, the
to 2% of all cancers in North America, but it is the most lack of long-term follow-up in surveillance studies, and
common solid malignancy in men 20 to 35 years of age.1 the increased cost of surveillance have all dissuaded most
The vast majority (98%) are primary germ cell tumors clinicians from abandoning the traditional treatment
(GCTs). Of GCTs, approximately 60% are seminomas approach.3 Stage II group patients with small bulk
and most usually present as an asymptomatic testicular retroperitoneal lymphadenopathy have a high probability
mass.1,2 The identification of prognostic factors in of long-term disease control with RT.4 Stage II group
patients with both early and advanced disease has helped patients with large masses and stage III group patients
to refine management strategies for these patients. The are managed by CT.
management of patients with testicular tumors is signifi-
cantly affected by histology and disease extent.
EPIDEMIOLOGY
Treatment results with seminoma have been good for
many years because of the relatively low metastatic rate The age distribution of testicular cancer is similar in all
and high radiosensitivity. More recent advances in Caucasian populations. There is a small peak in early
chemotherapy (CT) have improved the cure rate to childhood around 2 years of age, with rates then remain-
>95% overall. The postorchidectomy management of the ing low until 15 years of age.5 There is a second peak in
stage I group seminoma patients is focused on reducing young adults around 25 to 40 years of age and the rate
the side effects of therapy. Treatment options include then declines with a small peak again between 65 and 75
surveillance, adjuvant radiation therapy (RT), retroperi- years of age. Testicular cancers occurring in childhood
toneal lymphadenectomy, and adjuvant CT. Adjuvant and in the young adult years are usually GCTs, while
retroperitoneal RT remains the treatment of choice in those occurring after age 65 are principally nongerm cell
most centers. The success of surveillance in stage I non- malignancies, mainly lymphomas. Nonseminomatous
seminomatous germ cell testis tumors, the establishment tumors are more common in childhood and in the 15-to-30
of curative CT for advanced disease, and the improve- age group, while seminomas are seen more frequently in
ments in computed tomography for staging have led to slightly older (25 to 45 years) patients. In 2003, it is esti-
reexamination of the standard treatment approach in mated that there will be approximately 7500 new cases
some centers. Also, while the acute morbidity of the rel- and probably 300 deaths due to testicular cancer in the
atively low-dose RT used in this setting is minimal, there U.S.6 The current incidence of testicular cancer in the
are reports of impaired spermatogenesis, increased rates white U.S. population is 6:100000 males per year and in
of gastrointestinal symptoms and peptic ulceration on Canada it is 4:100000.7,8 The cumulative lifetime risk of
long-term follow-up, and increasing concern regarding developing a GCT is 0.2%.5 The incidence of testicular
the possible induction of second malignancies by RT. tumors is rising but the reasons are not well understood.
However, the low relapse rates following radiation, the The incidence rate has doubled in the past 30 years, and

577
578 Part VI Testis

while most patients present with early-stage curable dis- relation between germ cell cancers and these genital
ease, the continued rising incidence of these tumors pres- abnormalities.
ents a challenge. Weir et al.2 reported that the incidence
of testicular GCT (TGCT) has risen in Ontario by 60%
SYMPTOMS
between 1964 and 1996. Seminomas have increased by
72% and nonseminomas by 45%. From SEER data, the The commonest mode of presentation is a painless
overall incidence of TGCT rose over 44% from 3.35 to swelling of the testis but a few patients complain of pain
4.84 per 100,000 men between 1973 to 1978 and 1994 to or heaviness in the affected side. A hard, nontender testis
1998. Among white men, the incidence rose 52% from mass that cannot be transilluminated is diagnostic of a
3.69 in 1973 to 1978 to 5.62 per 100,000 men in 1994 to testicular cancer until proven otherwise. Occasionally,
1998. Among black men, the overall incidence of TGCT presentation with loss of libido or infertility leads to
rose 25% from 0.83 in 1973 to 1978 to 1.04 per 100,000 detection of a testicular mass. Acute presentations with
men in 1994 to 1998.1. The incidence in England and symptoms resembling torsion, hematospermia, varico-
Wales was 5.4 in 1997 compared with 2.9 per 100,000 cele, or thrombosis of the pampiniform plexus are rare.
person-years in 1971.9 Similar increases in incidence Nipple tenderness or breast swelling is noted in approxi-
have been reported in other populations with European mately 5% of patients. Back pain as a result of metastatic
ancestry, including Australia, New Zealand, and Europe lesion is a very rare presentation.
itself.10 Geographically, the highest incidence of testicu- Any history of cryptorchidism, orchiopexy, or any
lar cancers is seen in Denmark (8.4 per 100,000 men per other inguinal or scrotal surgeries should be elicited, as
year) and Switzerland (8.8 per 100,000 per year).5 It is these may be relevant to etiology or pattern of metasta-
known that the incidence is lower in nonwhites com- sis. When RT is being considered, the presence of
pared to whites. Low incidence rates are seen in other inflammatory bowel disease (IBD), previous abdominal
ethnic groups, such as Americans of Chinese and or pelvic surgeries, or any other intraabdominal diseases
Japanese descent. However, a high incidence of testicular should be ruled out.
cancer is seen in some nonwhite populations, such as the
Maoris in New Zealand and Native Americans.
PRIMARY SURGERY AND STAGING
Radical inguinal orchiectomy is usually the initial treat-
ETIOLOGY
ment that is both diagnostic of the tumor type and ther-
Cryptorchidism affects 0.7% of men and is the only con- apeutic. It is curative in approximately 75% of patients
dition that has been definitely associated with an with clinical stage I disease. An intraoperative excisional
increased risk of testicular cancer.7,11 The mechanisms are biopsy of the testicular mass for frozen-section evalua-
unknown. Pure seminoma is the most common tumor tion after delivery of the testis from the scrotum may pre-
histology observed in cryptorchid testes.11 Impalpable vent unnecessary orchiectomy in the small proportion of
cryptorchid testes may be detected with 91% and 95% patients who are ultimately found to have benign testic-
accuracy by ultrasound and CT, respectively. The pattern ular masses, but it is not routinely performed if the mass
of nodal metastasis may be different from that of scrotal is large or clinically malignant in appearance.
primary tumors with a much higher incidence of pelvic Measurement of the serum tumor markers alpha-
nodal involvement.12 There is ongoing research to define fetoprotein (AFP), the beta subunit of human chorionic
the genetics of susceptibility to testicular cancer. An gonadotropin (β-hCG), and lactate dehydrogenase
international consortium has been established to collect (LDH) play an important role in the diagnosis, treat-
pedigrees of patients and their families who are thought ment, and establishment of prognosis. A detectable AFP
to have increased susceptibility. A gene on the X chromo- is an indication of nonseminomatous elements, which
some (Xp27) has been identified. Patients with a family mandates treatment as a nonseminoma. Up to 40% of
history of testis cancer, undescended testes, and bilateral patients with pure seminomas have low but detectable
testicular cancers are being studied to identify further sus- levels of (β−hCG) and >200 ng/ml of (β−hCG) suggests
ceptibility genes.13 There may be a relation between metastatic disease or nonseminomatous elements.16
estrogenic exposure in utero and testicular cancer, and LDH is a useful marker in cases of advanced seminoma
there is evidence that environmental pollutants with and therapy can be monitored by serial measure-
estrogenic or antiandrogenic activity result in hormonal ments.17,18 Placental alkaline phosphatase is nonspecific
disruption and are responsible for the steadily increasing and has not generally been found useful in the manage-
incidence of testicular cancer.14,15 ment of seminoma.18
The increased incidence of germ cell cancers among The half-lives of AFP and β-hCG are 5 days and 24
men with testicular atrophy, testicular dysgenesis, cryp- hours, respectively. The tumor markers are measured
torchidism, and infertility suggests a common etiologic serially, immediately before or at the time of radical
Chapter 35 Seminoma: Management and Prognosis 579

orchidectomy. If elevated, measurements should be Classical seminoma is usually seen in the fourth decade
repeated until the level(s) normalize or plateau (the later of life. The typical histologic picture is sheets of rela-
indicates residual metastatic disease). tively large cells with clear cytoplasm and densely
Staging investigations should include a chest x-ray and staining nuclei with 10% to 15% of them showing syn-
a CT scan of the abdomen and pelvis. Patients with cytiotrophoblastic elements and 20% showing lympho-
retroperitoneal lymphadenopathy should also have a CT cytic infiltration. β-hCG levels are proportionate to the
scan of the chest and a bone scan. extent of syncytiotrophoblastic cells.
Clinical staging of testis tumors is now generally Anaplastic seminoma also occurs in the fourth decade.
done according to the 2002 UICC TNM staging system This histologic subtype may indicate a poorer prognosis
(see staging chart as Table 35-1). Seminoma is clinically but this is not supported by the results with surveil-
confined to the testis at diagnosis (T1–4, N0, M0, or lance.27 Increased mitotic activity, microinvasion, nuclear
stage I group) in approximately 75% of men. The dis- pleomorphism, and cellular anaplasia are its typical fea-
ease is characterized by an orderly, predictable spread tures. β-hCG levels are elevated in 36%, 25% present
pattern from the testis to paraaortic lymph nodes at or with higher stage disease, and almost half of these
below the renal hilar and then to distant sites, including patients have extragonadal extension of the primary
mediastinal and supraclavicular lymph nodes, lung, and tumor.
bone. Approximately 20% of patients have involvement Spermatocytic seminoma is a histologic variant that
of regional lymph nodes at presentation (any T, N1-3, occurs in the elderly, around the sixth decade. They are
M0, or stage II group), most commonly paraaortic large multinodular fleshy gelatinous and hemorrhagic
nodes. Distant metastases are evident at diagnosis tumors. Under the microscope they have solid sheets of
(any T, Any N, M1, or stage III group) in only 5% of cells interrupted by pseudoglandular patterns. Nests of
patients. cells in edematous stroma with occasional lymphocytic
Nonstandard surgical approaches (scrotal violations), infiltrates can be seen. Spermatocytic seminomas do
including scrotal orchiectomy, open testicular biopsy, and not metastasize, so radical orchiectomy is an adequate
fine needle aspiration, have historically been condemned treatment.
as potentially complicating further treatment and com- Warde et al.28 showed that on univariate analysis,
promising patient prognosis.19 Capelouto et al.20 showed tumor size (relapse-free rate or RFR: = 4 cm 87% ver-
that although statistically significant differences were sus >4 cm 76%; p = 0.003), rete testis invasion (RFR:
found in the local recurrence rate among the scrotal vio- 86% when absent versus 77% when present, p =
lation and inguinal group studies, the overall local recur- 0.003), and the presence of SVI (small vessel invasion)
rence rates were small (2.9% versus 0.4%, respectively) (RFR: 86% when absent versus 77% when present, p =
and did not occur in stage I group seminoma patients. 0.038) were predictive of relapse.28 On multivariate
There were no statistical differences in distant recur- analysis, tumor size (= 4 cm) and invasion of the rete
rences or survival rates in all groups analyzed. Patients testis remained important predictors for relapse.
with scrotal violation who did not receive prophylactic Thus, the size of the primary tumor and rete testis
local therapy fared as well as those who did. Patients with invasion are important prognostic factors for relapse
stage I disease and scrotal violation should not necessar- in patients with stage I seminoma when managed with
ily be disqualified from surveillance protocols or sub- surveillance.
jected to adjuvant local therapy.
PATTERNS OF SPREAD
PATHOLOGY
The N staging of the testicular tumor depends on the
Testicular Intraepithelial Neoplasia
number and size of involved retroperitoneal lymph
Testicular intraepithelial neoplasia (TIN), previously nodes. Spread is primarily lymphatic and is predictable.
referred to as carcinoma-in-situ (CIS), is the precursor Surgical mapping studies by Donohue et al.,29 Weissbach
to all TGCTs except spermatocytic seminoma.21 and Boedefeld,30 and other workers have defined the pat-
Furthermore, virtually all cases of TIN in postpubertal terns of metastasis in terms of its landing sites or stations.
men will progress to invasive cancer if given sufficient For right-sided tumors, the first station is the interaorto-
time.22 The incidence of TIN in the contralateral testis caval nodes, then the precaval and preaortic; for left-
of men with a unilateral GST is approximately 5%, sided, first station is paraaortic and preaortic and then the
which approximates the incidence of second contralateral interaortocaval nodes. The presence of more caudal
testicular tumors.23–26 metastatic nodes is generally seen in high volume disease
There are three histologic subtypes of seminoma: clas- or with aberrant lymphatic drainage. Contralateral
sical (70% to 85%), anaplastic (10% to 30%), and sper- spread is more common from right-sided tumors than
matocytic (2% to 12%). left, especially in high volume disease.
580 Part VI Testis

Table 35-1 Staging (UICC 2002)


DEFINITIONS

Pathologic Primary Tumor (T)(1)

■ pTX Primary tumor cannot be assessed (if no radical orchiectomy has been Notes
performed, TX is used) 1. Except for pTis and pT4,
extent of primary tumor is
■ pT0 No evidence of primary tumor (e.g., histologic scar in testis) classified as radical orchiectomy.
TX may be used for other
■ pTis Intratubular germ cell neoplasia (carcinoma in situ) categories in the absence of
radical mechiactomy.
■ PT1 Tumor limited to the testis and epididymis without vascular/
lymphatic invasion; tumor may invade into the tunica albuginea
but not the tunica vaginalis

■ pT2 Tumor limited to the testis and epididymis with vascular/


lymphatic invasion, or tumor extending through the tunica
albuginea with involvement of the tunica vaginalis

■ PT3 Tumor invades the spermatic cord with or without vascular/lymphatic


invasion

■ pT4 Tumor invades the scrotum with or without vascular/lymphatic invasion

Clinical Primary Tumor (T)

■ Tumor stage is generally determined after orchiectomy at which time a pathologic stage is assigned.

Pathologic Regional Lymph Nodes (N) Clinical Regional Lymph Nodes (N)
■ NX Regional lymph nodes cannot be
■ pNX Regional lymph nodes cannot be assessed assessed
■ pN0 No regional lymph node metastasis ■ N0 No regional lymph node metastasis
■ pN1 Metastasis with a lymph node mass 2 cm or ■ N1 Metastasis with a lymph node mass 2 cm
less in greatest dimension and less than or equal or less in greatest dimension; or
to 5 nodes positive, none more than multiple lymph nodes, none more
2 cm in greatest dimension than 2 cm in greatest dimension
■ pN2 Metastasis with a lymph node mass more than 2 cm ■ N2 Metastasis with a lymph node mass
but not more than 5 cm in greatest dimension; or more than 2 cm but not more than
more than 5 nodes positive, none more than 5 cm; 5 cm in greatest dimension; or multiple
or evidence of extranodal extension of tumor lymph nodes, any one mass greater
than 2 cm but not more than 5 cm
■ pN3 Metastasis with a lymph node mass more
in greatest dimension
than 5 cm in greatest dimension
■ N3 Metastasis with a lymph node mass
more than 5 cm in greatest dimension
Clinical Pathologic Distant Metastasis (M)

■ ■ MX Distant metastasis cannot be assessed

■ ■ M0 No distant metastasis

■ ■ M1 Distant metastasis

■ ■ M1a Non-regional nodal or pulmonary metastasis

■ ■ M1b Distant metastasis other than to non-regional lymph nodes and lungs

Biopsy of metastatic site performed ■ Y ■ N

Source of pathologic metastatic specimen_____


Chapter 35 Seminoma: Management and Prognosis 581

Table 35-1 Staging—cont’d


Serum Tumor Murder (S) (N indicates the upper limit of normal for the LDH assay)

■ ■ SX Marker studies not available or not performed

■ ■ S0 Marker study levels within normal limits

■ ■ S1 LDH < 1.5 × N AND

hCG (mIu/ml) < 5000 AND

AFP (ng/ml) < 1000

■ ■ S2 LDH 1.5–10 × N OR

hCG (mIu/ml) 5000–50,000 OR

AFP (ng/ml) 1000–10,000

■ ■ S3 LDH > 10 × N OR

hCG (mIu/ml) > 50,000 OR

AFP (ng/ml) > 10,000

Clinical Pathologic Stage Grouping Notes


Additional Descriptiors
■ ■ 0 pTis N0 M0 S0 Lymphatic Vessel Invasion (L)

■ ■ I pT1-4 N0 M0 SX LX Lymphatic vessel invasion cannot


be asscesed
■ ■ IA pT1 N0 M0 S0
L0 No lymphatic vessel invasion
■ ■ IB pT2 N0 M0 S0
L1 Lymphatic vessel invasion
pT3 N0 M0 S0
Venous Invasion (V)
pT4 N0 M0 S0
VX Venous invasion cannot be assumed
■ ■ IS Any pT/Tx N0 M0 S1–3
V0 No venous invasion
■ ■ II Any pT/Tx N1–3 M0 SX
V1 Microscoic venous invasion
■ ■ IIA Any pT/Tx N1 M0 S0
V2 Macroscopic venous invasion
Any pT/Tx N1 M0 S1

■ ■ IIB Any pT/Tx N2 M0 S0

Any pT/Tx N2 M0 S1

■ ■ IIC Any pT/Tx N3 M0 S0

Any pT/Tx N3 M0 S0

■ ■ III Any pT/Tx Any N M1 SX

■ ■ IIIA Any pT/Tx Any N M1a S0

Any pT/Tx Any N M1a S1

■ ■ IIIB Any pT/Tx N1–3 M0 S2

Any pT/Tx Any N M1a S2

Continued
582 Part VI Testis

Table 35-1 Staging—cont’d


■ ■ IIIC Any pT/Tx N1-3 M0 S3

Any pT/Tx Any N M1a S3

Any pT/Tx Any N M1b Any S


Residual Tumor (R)
■ RX Presence of residual tumor cannot be assessed
■ R0 No residual tumor
■ R1 Microscopic residual tumor
■ R2 Macroscopic residual tumor

Additional Descriptors

For identification of special cases of TNM or pTNM


classifications, the “m” suffix and “y,” “r,” and “a”
prefixes are used. Although they do not affect the stage
grouping, they indicate cases needing separate analysis.

■ m suffix indicates the presence of multiple primary


tumors in a single site and is recorded in parentheses:
pT(m)NM.

■ y prefix indicates those cases in which classification is


performed during or following initial multimodality
therapy. The cTNM or pTNM category is identified by
a “y” prefix. The ycTNM or ypTNM categories the extent
of tumor actually present at the time of that examination.
The “y” categorization is not an estimate of tumor prior to
multimodality therapy.

■ r prefix indicates a recurrent tumor when staged after a


disease-free interval, and is identified by the “r”
prefix rTNM.

■ a prefix designates the stage determined at autopsy:


aTNM. Indicate on diagram primary tumor and regional
nodes involved.
Prognostic Indicators (if applicable)

Involvement of inguinal, pelvic, and iliac lymph nodes


PREVENTION AND EARLY DETECTION
is uncommon except in cases of (i) prior pelvic or abdom-
inal surgery where the normal lymphatic drainage may be The identification of TIN as a precursor of TGCT has
disturbed, (ii) seminomas arising in a cryptorchid testis, raised the possibility that the development of invasive
(iii) congenital anomalies of the genitourinary tract, (iv) testicular cancer can be prevented by treating TIN. The
bulky paraaortic lymphadenopathy, and (v) possibly post- incidence of TIN in the general population is low at
scrotal orchiectomy with incision of the tunica albuginea 0.7%; however, the diagnosis should be considered in
and with tumor invasion of tunica vaginalis or the lower high-risk patients, including those with a history of cryp-
third of the epididymis.31–33 torchidism, presumed extragonadal GCT, androgen
Chapter 35 Seminoma: Management and Prognosis 583

insensitivity syndrome, with intersex syndromes or gery. The optimal follow-up program for surveillance has
gonadal dysgenesis, and in patients with contralateral not been determined and has evolved over time.
GCTs.34,35 It has been suggested that men with a uni- Abdominal radiation to the paraaortic lymph nodes is
lateral tumor should undergo biopsy of the contralateral highly effective with rare in-field relapse.42 Acute side
testis, preferably at the time of ipsilateral orchiectomy effects are modest although long-term follow-up has
with the aim of identifying and treating TIN before revealed a slight increase in secondary malignancy.43,44
progression to invasive disease.36 Those with a small, Primary chemotherapy for high-risk patients is not
soft contralateral testis and severe oligospermia or widely practiced in North America, in part because
aspermia are at a higher risk that appears to justify strong prognostic factors have not been defined.45
biopsy if treatment is going to be recommended. Men
with a normal testicular biopsy can be reassured that
Role of Surveillance
their risk of a contralateral tumor is <1%.23 Those with
a positive biopsy should be considered for testicular RT Surveillance is feasible in patients with seminoma
using a fractionated dose of 20 Gy that has been because of the predictable recurrence pattern, the avail-
reported to eradicate TIN and prevent the development ability of high quality abdominal imaging, which allows
of invasive cancer.22 Leydig cell function with andro- recurrences to be detected at an early stage, and the
gen production appears to be preserved in most effectiveness of chemotherapy at curing even patients
patients.35,37 with advanced disease. Surveillance is a safe alternative to
Testicular self-examination has been advocated by adjuvant RT in stage I seminoma. It avoids unnecessary
many for the early detection of invasive tumors but its treatment and treatment-induced morbidity in the
usefulness is unproven. There is no evidence to indi- majority of patients and does not compromise cure.
cate that a screening program would be of benefit; Newly diagnosed patients at Princess Margaret Hospital
however, there is a need for education about the early (PMH) are usually offered both surveillance and imme-
signs and symptoms of testicular cancer to reduce delay diately RT, and most opt for surveillance. Nevertheless,
at presentation. Patients with unilateral TGCTs and surveillance is not yet an accepted alternative to RT in
cryptorchidism should practice transmissible spongi- many centers for several reasons. It requires a commit-
form encephalopathy (TSE). An ultrasound can be ment by both patient and physician to long-term inten-
done for early detection and intervention if a tumor is sive monitoring with regular physical examination, chest
suspected. x-ray, CT imaging of the abdomen and pelvis, and meas-
Testicular microlithiasis has been considered a pre- urement of serum LDH, AFP and β-hCG. A number of
cursor of testicular cancer. The term testicular prospective nonrandomized studies of surveillance have
microlithiasis refers to the high intensity signals that are been conducted over the past 15 years. The largest
recorded on ultrasound, where microcalcifications are reported series are the PMH and Danish Testicular
within the tubules. 38–40 Patients with testicular Carcinoma Study Group’s (DATECA) trials.46 In the
microlithiasis found on ultrasound along with associ- PMH study of 241 patients, 5-year actuarial relapse-free
ated risk factors, such as infertility or cryptorchidism, survival was 86% with a median follow-up of 7.3 years.3
may require close follow-up considering its association In the DATECA study, 19% of patients had failed with a
with testicular neoplasia.39 median follow-up of 48 months.41 Other studies with
greater than 36-month follow-up have reported similar
relapse rates. Site of relapse was similar in all surveillance
MANAGEMENT OF CLINICAL STAGE I (T1–4,
studies with the vast majority recurring in the paraaortic
N0, M0) GROUP
and interaortocaval nodes, 33 of 37 (89%) and 41 of 49
Stage I seminoma represents the most common presen- (82%) in the PMH and DATECA studies, respectively.
tation of the disease, representing 70% to 80% of all new While the median time to relapse ranged from 12 to 18
cases.3,41 For the past 20 years, clinical experience with months, recurrences have been reported as late as 9 years
surveillance has demonstrated that, after orchiectomy, from diagnosis.47,48 This implies the need for ongoing
survival is equivalent to the outcome with adjuvant RT, follow-up over an extended period. Patients who choose
which remains the standard of care. With surveillance, surveillance but are poorly compliant with follow-up may
many patients can avoid unnecessary therapy but require compromise their outcome by delaying the diagnosis of
close follow-up, and a proportion will progress and recurrence until a late stage when more intensive therapy
require further treatment. For this latter reason, it is is necessary or cure is not possible. Hao et al.49 described
important to identify potential prognostic factors for two deaths among poorly compliant patients with non-
occult metastatic disease so that patients at high risk of seminomatous testicular cancers on surveillance, but
relapse can be treated with adjuvant treatment after sur- none among patients who attended regularly for follow-up.
584 Part VI Testis

Finally, at least one economic analysis has suggested that relapse.48 Among 57 patients who had no adverse prog-
surveillance may be more costly to the health care budget nostic factors (age > 34, tumor size < 6 cm, and no lym-
than immediate RT, although the costs of managing treat- phvascular invasion), tumor relapse was 6% at 5 years. In
ment-induced second malignancies and the psychologic the DATECA study, primary tumor size was the only risk
and social costs to patients were not considered.50 factor. Risk of relapse at 4 years was 6%, 18%, and 36%
Prognostic factors for relapse have been identified.51 A among patients with tumors <3 cm, 3 to 6 cm, and >6 cm,
list of studies comparing these prognostic factors is pro- respectively.46 In a series published by the group at Royal
vided in Table 35-2. In the PMH series, only age (≤34 Marsden, only the presence of lymph vascular space
years) and tumor size (>6 cm) were associated with involvement was associated with relapse (9% versus 17%).52

Table 35-2 Prognostic Factors for Relapse in Stage I Seminoma Patients on Surveillance
Series Relapse (%) Factor Strata Relapse (%)

Horwich, 1992, n = 103 18 SVI* No 10

Yes 20

von der Masse, 1993, n = 261 20 Size <3 cm 6

3–5.9 cm 18

>6 cm 36

Histology Spermatocytic 0

Classical 16

Anaplastic 33

Necrosis No 14

Yes 23

Rete testis No 14

Yes 23

Warde, 1997, n = 201 15 Size <6 cm 12

>6 cm 33

Age <34 9

>34 21

SVI No 14

Yes 31

18 Size <4 cm 13

Worde et al. 1999, n = 638 >4 cm 24

Rete testis No 14

Yes 24

SVI No 14

Yes 23

*SVI, Small vessel invasion by tumor.


Chapter 35 Seminoma: Management and Prognosis 585

Effective surveillance for seminoma implies effica- consequence, the side effects that arise months or years
cious therapy for patients who relapse. In the DATECA after the treatment is finished may have long-term con-
and PMH series, most patients were treated with sequences. Chronic gastrointestinal symptoms may
retroperitoneal RT. Second relapse post-RT was 19% in develop, and an increased incidence of peptic ulceration
the PMH series and 11% in the DATECA series. particularly after abdominal radiation doses in the range
Although this relapse rate is higher than de novo treated of 30 to 45 Gy has been reported.55,56 The germinal
patients, it is important to recognize that this represents epithelium of the testis is one of the most sensitive tis-
a subset of patients who have already failed surveillance. sues in the body to ionizing radiation, and doses as low
Overall failure for the entire cohort is similar to upfront as 20 cGy (<1% of the dose that is usually used to
RT, and almost 100% of patients are cured regardless of treat seminoma) are sufficient to transiently elevate
choice of therapy postorchiectomy. gonadotropins and reduce sperm counts.57,58 Even with
An additional concern about surveillance is the ability shielding of the remaining testis, the scatter radiation
to detect retroperitoneal disease at a small volume dose results in a significant risk of infertility in previously
(<5 cm) such that recurrence can be managed with RT fertile men.59 However, the most dire consequence of
without the need for cytotoxic chemotherapy. In the radiation for seminoma may be the increased risk of sec-
PMH experience, a similar number of patients managed ond malignant tumors within the irradiated volume many
with surveillance (13 of 241) and adjuvant RT (10 of 254) years after treatment. Radiation-induced tumors must be
ultimately required chemotherapy as part of their treat- distinguished from the development of a second GCT,
ment regimen. It is difficult to determine the incidence of which occurs in about 5% of patients with seminoma and
this phenomenon from the reported literature, as in many reflects an underlying predisposition.25,26 The largest
centers chemotherapy is routinely used at relapse; also in study of second malignant non-GCTs in patients with
many isolated cases, patients had known contraindications. testicular cancer was a cooperative effort involving over
28,000 patients from 16 population-based cancer reg-
istries.44 The actuarial excessive risk of developing a sec-
Role of Adjuvant Radiation Therapy for Dose
ond malignancy increased progressively with time from
Reduction
diagnosis of testicular cancer, and was 18% at 25 years.
The traditional management of patients with stage I The risk was the same for seminoma and nonseminoma-
seminoma following radical orchiectomy is with RT tous tumors. RT was associated with a 2- to 6-fold
directed to the paraaortic lymph nodes below the increased risk of leukemia, as well as an increased risk of
diaphragm and the ipsilateral common and external iliac solid tumors of the gastrointestinal and genitourinary
lymph nodes to the level of the mid-obturator foramen. tracts. Chemotherapy that included alkylating agents was
The inguinal lymph nodes may or may not be included associated with a 7- to 10-fold excessive risk of leukemia.
depending on the estimated risk of inguinal involvement.
The radiation dose is typically 25 to 30 Gy in 15 to 20
Role of Modified Adjuvant Radiation Therapy
daily fractions. Patients experience fatigue and mild gas-
trointestinal upset during therapy that is usually readily Given these concerns about abandoning adjuvant RT
controlled with 5-hydroxytryptamine antagonists for entirely in patients with clinical stage I seminoma in
nausea and antimotility agents for cramping and diar- favor of surveillance, alternate management strategies
rhea. These side effects typically resolve within a few have emerged that aim to reduce the toxicity of RT by
weeks of completing RT, although fatigue may occasion- reducing either the radiation dose or the volume that is
ally persist longer. Numerous reports have documented treated. A radiation dose of 25 Gy in 20 daily fractions
long-term disease control rates of approximately 95% controls microscopic seminoma in paraaortic lymph
following radiation.27,53,54 Recurrences within the irradi- nodes with a probability approaching 100%. The dose-
ated volume are extremely rare if treatment is planned response relationship for seminoma below this level is
and delivered appropriately, and follow-up imaging of unknown, although there are isolated reports of in-field
the abdomen and pelvis is therefore not required. The recurrences after fractionated doses of 15 and 21 Gy.60,61
few recurrences that do arise typically involve the medi- There is experience in the United Kingdom using a radi-
astinal or supraclavicular lymph nodes or the lung ation dose of 20 Gy in 8 daily fractions. The treatment
parenchyma and can usually be detected with a combina- was well tolerated, and there was only one in-field recur-
tion of regular physical examination and chest x-ray. The rence among 263 patients. It is difficult to compare these
majority of these patients are salvaged with chemother- results to more conventional dose-fractionation schemes
apy administered at the time of relapse, and overall cure because of differences in daily fraction size (2.5 Gy ver-
rates approach 100%. sus 1.25 to 2 Gy, respectively). Total dose and fraction
While the acute side effects of radiation that develop size both influence the biologic effectiveness of a course
during treatment are usually self-limiting and of minimal of RT. The Medical Research Council of the United
586 Part VI Testis

Kingdom (MRC, UK) has conducted a phase III ran- the remaining testis and preserve fertility without the
domized study comparing high- and low-dose radiation requirement for ongoing pelvic surveillance.67
schedules with equal fraction size (30 Gy in 15 fractions
versus 20 Gy in 10 fractions). The results of this study,
Role of Primary Chemotherapy
which are not yet available, should better define the low-
est dose that is necessary to reliably control microscopic The effectiveness of multiagent cisplatin-containing
deposits of seminoma. However, small to modest reduc- chemotherapy in advanced seminoma has lead to at least
tions in dose of this magnitude may not necessarily trans- 4 studies of adjuvant chemotherapy with one or two
late into a reduced risk of infertility or second courses of single-agent carboplatin in patients with stage I
malignancy, especially if treatment is administered in an disease as an alternative to RT or surveillance.68–70
otherwise conventional fashion to encompass the Median follow-up was 24 to 74 months. Only 2 of the
paraaortic and ipsilateral pelvic lymph nodes. 160 patients developed a recurrence. Treatment was well
Paraaortic relapse accounts for 85% of recurrences in tolerated without significant acute morbidity. There was
seminoma patients on surveillance, whereas recurrence minimal disruption of normal lifestyle and fertility did
in the ipsilateral iliac lymph nodes is seen in <10% of not appear to be compromised.68,71,72 However, notwith-
patients.46,48,52 In addition, surgicopathologic series of standing these promising results, the use of chemother-
patients with clinical stage I nonseminomatous testicular apy in this setting must be approached cautiously. The
cancer have demonstrated ipsilateral common iliac nodal experience is small, and the follow-up remains short.
involvement in only about 10% of cases.29 One of the Late recurrences may yet arise and the long-term toxic-
most important factors determining the radiation dose to ity of this treatment, particularly with respect to the
the remaining contralateral testis, and therefore the risk induction of leukemia, is not known.44 The MRC, UK is
of infertility, is the distance from the inferior edge of the presently comparing a single cycle of carboplatin to adju-
radiation field to the scrotum.59 With this knowledge, vant RT in a randomized study that should help to clar-
several investigators have proposed limiting the radiation ify the relative efficacy and toxicity of these treatments.
fields to treat only the paraaortic nodes.42,62–64 An MRC, Several studies have reported that adjuvant chemother-
UK, randomized phase III study comparing conventional apy yields higher cure rates in relapsed surveillance
paraaortic and ipsilateral pelvic radiation to paraaortic patients compared with those who underwent adjuvant
RT alone in 478 patients was recently reported, and RT , reinforcing the value of adjuvant chemotherapy for
showed no difference in disease-free survival between the stage I seminoma.
two arms.42 There were four iliac lymph node recur-
rences in patients who received paraaortic irradiation
MANAGEMENT OF CLINICAL STAGE II (ANY
alone, and none in patients treated with paraaortic and
T, ANY N, M0) GROUP
ipsilateral pelvic radiation. Sperm counts after treatment
were significantly higher in the paraaortic group. Only about 20% of patients have clinically evident
This study is likely to significantly influence the stan- involvement of paraaortic lymph nodes at diagnosis and
dard of RT practice for patients with stage I seminoma, are classified as having stage II disease. The number of
in that a greater proportion will receive paraaortic radia- patients is too small to mount phase III clinical trials of
tion alone. However, the small risk of iliac lymph node management, and treatment decisions are generally
recurrence remains problematic. The other usual sites of based on reports from single institutions where patients
recurrence after paraaortic RT alone are the inguinal, have been treated in a uniform fashion.54,73–76
mediastinal, and supraclavicular lymph nodes and the The most important prognostic factor in stage II
lung parenchyma.42,63 These areas are easily followed by seminoma is the bulk of the retroperitoneal tumor, meas-
physical examination and chest x-ray, but regular CT ured as the axial diameter of the largest lymph node or
imaging may be necessary to detect pelvic recurrence at lymph node mass visible on CT scan. Patients who are
an early stage. The advantage of paraaortic RT alone is staged according to the 2002 UICC TNM classification
therefore diminished, particularly in comparison to sur- are stratified into 3 groups reflecting nodal masses of <2
veillance. A compromise may be to irradiate the paraaor- cm (stage IIA), 2 to 5 cm (stage IIB), and >5 cm (stage
tic and ipsilateral common iliac lymph nodes by IIC). Approximately 70% of stage II patients have small
positioning the inferior border of the radiation fields at bulk retroperitoneal disease at presentation with lymph
the lower aspect of the sacroiliac joints.65 This encom- nodes that are <5 cm.76
passes the lymph nodes that are typically removed at Lymph node size was the only factor that predicted
lymphadenectomy in patients with nonseminomatous recurrence in 80 patients with stage II seminoma treated
tumors.66 The external iliac and inguinal nodes are not with RT at PMH.76 The 5-year RFR in patients with
treated but are unlikely to harbor occult metastases. This nodal disease of <5 cm was 89%, compared to 44% in
approach has the potential to reduce the scatter dose to patients with bulkier disease. In total, 16 of 80 patients
Chapter 35 Seminoma: Management and Prognosis 587

treated with radiation developed recurrence, most com- the PMH series), the ease with which supraclavicular
monly in mediastinal or supraclavicular lymph nodes, lymph nodes can be followed clinically, the availability
lung, or bone. Thirteen patients were treated with of effective salvage chemotherapy, the possibility of
chemotherapy at relapse, and 9 were free of disease at compromising bone marrow reserve for subsequent
last follow-up. The other 4 patients plus 2 additional chemotherapy should it be necessary, and the potential
patients who did not receive salvage chemotherapy died for radiation-induced cardiac toxicity.54,78,79
of progressive seminoma. In contrast, no recurrences or Patients with gross retroperitoneal lymphadenopa-
deaths were seen in the 19 patients who received initial thy require follow-up imaging of the abdomen after
chemotherapy, even though 14 had nodal masses >5 cm treatment until complete regression of disease has
in size. These results support the continued use of pri- occurred. Residual retroperitoneal masses are fre-
mary RT in stage II patients with small bulk lym- quently seen that may either regress slowly over time
phadenopathy. However, the high failure rate following or remain stable. A stable persistent mass often repre-
RT in patients with bulky retroperitoneal disease, the sents fibrosis or necrosis and only the minority contain
fact that not all patients with recurrence were salvaged, active tumor.80–82 However, the possibility of a non-
and the apparently better outcome of similar patients seminomatous component to explain the persistence
who were treated with chemotherapy at diagnosis man- needs to be kept in mind even in patients whose pri-
dates primary chemotherapy instead of radiation in this mary tumors show pure seminoma.76 The Memorial
population. Sloan-Kettering Cancer Center (MSKCC) has the
Tumor bulk should not be the only parameter used to largest reported experience with the management of
decide on treatment of retroperitoneal disease in patients residual retroperitoneal masses in seminoma.82
with stage II seminoma. Other patient and tumor-related The most common sites of recurrence following RT
factors should also be considered. Lymph node masses in stage II patients with small bulk lymph node metas-
that are situated laterally may necessitate irradiating a tases are mediastinal or supraclavicular nodes, lung, and
large volume of one or both kidneys or the liver in order bone.76 Most relapsing patients are cured with
to adequately encompass the tumor. The same situation chemotherapy, which underscores the importance of reg-
may arise in cases of abnormal anatomy, such as with ular follow-up with clinical examination and chest x-ray
horseshoe or pelvic kidney. These patients are better after radiation. CT imaging of the abdomen and pelvis is
treated with chemotherapy because of an unacceptably not necessary after complete resolution of abdominal dis-
high risk of radiation toxicity. Patients in whom RT and ease. In the PMH series, 4 of the 16 patients who
chemotherapy are contraindicated or the diagnosis is recurred had bone metastases, and 3 of these 4 presented
uncertain should be considered for retroperitoneal with spinal cord compression as the first sign of recur-
lymph node dissection.77 rence.76,83 Therefore, all patients with unexplained back
RT offers a high probability of controlling metasta- pain require a bone scan to exclude metastases, and those
tic seminoma in lymph nodes <5 cm in size. The treat- with new onset neurologic deficits require urgent imag-
ment volume includes the gross tumor, as well as the ing of the spine with magnetic resonance imaging or
paraaortic and ipsilateral common and external iliac myelography.
lymph nodes. The radiation dose is typically 25 Gy in
20 daily fractions plus a boost of a further 10 Gy in 5 to
MANAGEMENT OF CLINICAL STAGE III (ANY
8 fractions to the gross lymphadenopathy. A CT scan
T, ANY N, ANY M) GROUP
with the patient in the treatment position is used to
ensure that the gross tumor is adequately encompassed Approximately 20% of testicular seminoma patients have
by the radiation fields and that the minimal possible metastatic disease; 15% in the regional lymph nodes and
volume of kidney and liver is irradiated. The contralat- 5% in distant organ metastasis (stage III).84 Multiagent
eral iliac lymph nodes may also be treated in cases cisplatin-based chemotherapy is effective treatment for
where lymphadenopathy in the low paraaortic area is patients with distant metastases at diagnosis (stage III),
deemed to increase the risk of these nodes being and even those with very advanced disease are frequently
involved by tumor. However, this is probably of most cured.85 The most important factor predicting survival of
concern in patients with bulky retroperitoneal lym- patients with stage III seminoma is the presence or
phadenopathy, who are better treated with primary absence of nonpulmonary visceral metastases. The
chemotherapy as discussed previously.33 Adjuvant radi- International Germ Cell Consensus Group reported that
ation of supraclavicular lymph nodes in patients with stage III patients with nonpulmonary visceral metastases
stage II disease has been recommended by some, had a 67% 5-year disease-free survival, compared to 82%
although it is not justified on a routine basis in view of in patients with only lung or lymph node involvement.83
the low risk of isolated supraclavicular recurrence (5% Liver or brain metastases, which are rarely seen in patients
of patients with retroperitoneal nodal masses >5 cm in with seminoma, carried a particularly poor prognosis
588 Part VI Testis

with reported disease-free survivals of 50% and 57%, >50 cGy may preclude recovery of spermatogenesis.93
respectively. Scrotal shielding does limit this somewhat but cannot
The evolution of chemotherapy for metastatic semi- assure protection in all patients. In men who recover
noma has paralleled the evolution of chemotherapy for spermatogenesis after RT for seminoma, there is no evi-
nonseminomatous GCTs. Cisplatin and carboplatin are dence of an increased incidence of genetic anomalies
the most active single agents and are responsible for the among their offspring.93 Limiting the RT field to
high cure rates, particularly when combined with other paraaortic and common iliac has reduced the concern
drugs. Historically, the most commonly used regimens regarding post-RT infertility. Elevated LH values with
consisted of cisplatin plus vinblastine, cyclophosphamide, normal testosterone values have been observed after
actinomycin-D and bleomycin (VAB-6), or cisplatin plus infradiaphragmatic irradiation.94
vinblastine and bleomycin (PVB).81,86 The vinblastine in
the PVB regimen was replaced by etoposide (BEP) with
no difference in outcome but a significant reduction in Second Malignancy
neuromuscular toxicity.87 Investigators at the MSKCC Patients with seminoma are at higher risk of later sec-
later showed that 4 cycles of cisplatin and etoposide (EP) ondary cancers in addition to an increased risk of con-
produced equivalent long-term disease-free survival to tralateral GCTs; they are at risk for other neoplasms,
bleomycin-containing regimens without the risk of pul- some of which may be related to treatment and others
monary toxicity associated with bleomycin.88 EP is now related to genetic/environmental exposures unrelated to
the most widely used regimen for initial treatment of treatment.44,95–97 Cancers that may arise secondary to
metastatic seminoma. Salvage treatment for patients who treatment are an important consideration for possible
develop progressive disease after first-line chemotherapy surveillance versus radiation for stage I seminoma. Travis
most often consists of combinations of cisplatin, ifos- et al.44 reported on the risk of second cancers among
famide, and vinblastine.89 High-dose chemotherapy with 28,843 long-term survivors of testicular cancer, of which
autologous bone marrow transplantation should be con- 15,000 had seminoma. Overall, seminoma patients had a
sidered for patients who fail more conventional therapy. 42% increased rate of second cancers. Risks were signif-
There is no role for routine surgery or RT in the man- icantly elevated among patients treated initially by RT
agement of stage III seminoma, although there may be but not chemotherapy although the absolute number in
specific indications in individual patients. Surgery should the chemotherapy cohort was relatively small (n = 560).
be considered for residual clinical or radiographic abnor- Subsequent reanalysis of these data also suggests that
malities in the retroperitoneum or at other sites follow- chemotherapy also elevated the risk of leukemia.43
ing chemotherapy, extrapolating from the experience Specific cancers that were detected at higher than
with nonseminomatous tumors.90 Surgery and RT may expected rates included leukemia, colon, renal, gastric,
be useful in achieving rapid tumor debulking in situations pancreatic, and bladder. Overall risk of second non–germ
where normal tissue function is compromised, such as cell malignancy was 18.2% at 25 years. Ruther et al.98
with brain metastases or spinal cord compression.91,92 assessed second malignancies among 839 patients with 4-
year follow-up. Among this cohort, of whom the vast
EFFECTS OF TREATMENT majority had RT , an excess number of renal cancers were
Acute noted. Although it is difficult to determine to what
degree these incidence rates would drop by active sur-
With the low doses of RT used in seminoma, acute compli- veillance, most investigators believe that a significant
cations are minor in most patients. Mild nausea and vomit- proportion of these cases are iatrogenic in nature.
ing are common. A small proportion of patients require
regular antiemetics and are unable to complete daily tasks
Psychologic Toxicity
while receiving RT. Diarrhea develops in a minority of
patients. An increased incidence of peptic ulceration in The vast majority of men with seminoma suffer no psy-
patients treated with 30 to 45 Gy has been reported. chologic sequelae from their illness.99 Some studies sug-
gest that these patients have an improved outlook on life
compared to their state prior to the detection of testicu-
Chronic
lar cancer and to that of age-matched controls. Isolated
Late Gonadal Toxicity
patients, however, suffer severe psychologic symptoms in
Germinal epithelium is highly radiosensitive and the domains of sexuality, parenthood, distress, and social
although the contralateral testis is not directly located in upheaval. Data of the psychologic distress of surveillance
the field, scatter dose can be significant and profoundly versus up-front active therapy are few. One study suggests
diminish spermatogenesis. A radiation dose between 20 that men on surveillance have fewer sexual problems than
and 50 cGy may produce temporary aspermia, and doses men on active treatment.100
Chapter 35 Seminoma: Management and Prognosis 589

Loss of one or both testes may cause psychologic dis- The largest reported series is from the MSKCC with
tress related to the feeling of decreased masculinity and 55 cases; 32 (58%) underwent complete resection and 23
changed body image. Testicular prostheses are helpful in had multiple biopsies only. In 27 patients the mass was >3
many cases, especially bilateral tumors; however, many cm in diameter and 8 of these had residual tumor.82 Of
unilaterally orchiectomized patients frequently regard the 8 with positive histologic findings 6 were seminomas,
them as not necessary. In spite of residual treatment and complete resection was possible in 6, 4 seminomas, and 2
disease-related morbidity (Raynaud’s phenomena, periph- teratomas. By CT scan characteristics well-defined lesions
eral sensory neuropathy, dry ejaculate, and hypogonadism) were more likely to be completely resected (78%) than
in one-third of patients, most patients cope satisfactorily poorly defined masses (44%). Unfortunately, the inter-
with the long-term posttreatment distress and do not vals from the completion of chemotherapy to surgery and
report a major reduction of health-related quality of life. the pretreatment sizes were not reported. Ravi et al.104
reported similar findings in 107 patients, in whom 43 had
residual tumor. Nineteen underwent surgery and in 11
Fertility
patients who had well-defined masses >3 cm in diameter,
In testicular cancer patients, the overall paternity rates 6 had positive histology. Flechon et al.105 reported that
are probably reduced by 15% to 30% compared with the viable tumor cells were noted only in residual masses 3 cm
normal population. After standard RT or standard or greater (13%) and 50% of residual masses disappeared
chemotherapy most patients are oligo or azoospermic for during surveillance.
6 to 12 months, but sperm production recovers within A recent report has suggested that the routine use of
the following 2 years.101 The risk adapted treatment of tes- RT in this setting is of little value, in part reflecting the
ticular cancer preserves fertility in many patients, such as low incidence of active tumor in residual masses and the
the use of paraaortic fields instead of RT to paraaortic and possibility of nonseminomatous elements that are less
iliac region, increased use of surveillance protocols, the radio-responsive.106 While it appears that surgery should
development of nerve-sparing retroperitoneal lym- be reserved for postchemotherapy residual masses >3 cm
phadenectomies, which are now being practiced laparo- in diameter and those <3 cm should be observed, it is also
scopically, reduction of number of chemotherapy cycles evident that surgery may be complicated due to excessive
and the use of spermatogenesis saving cytostatics. fibrosis following the chemotherapy and that there is a risk
Infertility and subfertility are major issues in the health- of perioperative mortality. Laparoscopic technique is
related quality of life in the young testicular cancer sur- being used at many centers for nonseminomas in an
vivor. Pretreatment cryopreservation of the semen attempt to reduce the morbidity of the open retroperi-
technique should be used to offer an option for parent- toneal lymph node dissection. The advantages of
hood to these patients. Assisted reproductive technique decreased pain, shorter hospital stay and convalescence,
can be offered in pretreatment or posttreatment cases hav- and superior cosmesis make laparoscopic retroperitoneal
ing low sperm count.102 Rectal probe electroejaculation or lymph node dissection a promising procedure for
sperm aspiration techniques can be used in patients with choice.107 Further studies are needed as it may be safer to
emission problems of the ejaculate posttreatment.103 follow masses, even >3 cm, to determine if they will
decrease in size. The natural history of residual masses in
seminoma is not well defined. It may be one of gradual
MANAGEMENT OF RESIDUAL MASSES
shrinkage unless nonseminomatous elements or a second
Residual retroperitoneal masses remain in 20% to 80% primary are present.
of cases after chemotherapy and the management
remains controversial. Different approaches have been
OTHER ISSUES AND MANAGEMENT
reported, including surgery, RT, and surveillance.
OF UNUSUAL CASES
Surgery is technically more difficult in seminomas com-
Patients With High hCG
pared to nonseminomas after chemotherapy because of
extensive fibrosis. Complete excision is often difficult and β-hCG > 200 ng/ml are suggestive of metastatic disease
only biopsies are performed. The percent of viable semi- and/or there may be nonseminomatous elements as the
noma is low at 10% to 20% and viable disease only seems cause.16 Thus, the higher levels of β-hCG normally
to present in well-delineated residual masses 3 cm or should arouse a suspicion for nonseminomatous tumors.
greater. RT has been proposed for postchemotherapy Weissbach and Busser-Maatz108 (in a large prospective
residual masses, although it may increase the risk of a sec- study of seminomas) found 31% of patients with β-hCG
ond solid tumor and decrease hematologic tolerance to secreting tumors were more likely to have metastases in
salvage chemotherapy in cases of relapse. Surveillance is retroperitoneal lymph nodes. In seminoma, β-hCG is an
an option based on the spontaneous regression of resid- indicator of tumor burden rather than a sign of tumor
ual masses. aggressiveness. A high hCG with negative imaging
590 Part VI Testis

studies is suggestive of retroperitoneal or distant occult bility of relapse when the standard radiation fields are
metastatic disease. used. Unusual patterns of relapse have been observed in
patients managed by surveillance, confirming concerns
regarding abnormal lymphatic pathways.115 For these
Bilateral Tumors
reasons, postorchiectomy surveillance in stage I and
The reported incidence of bilateral synchronous or chemotherapy in stage II have usually been recom-
metachronous testicular germ cell tumors is 0.5% to 7%. mended. Retroperitoneal lymphadenectomy is another
Approximately 5% are bilateral.25,26,109 Several studies option for patients unwilling to follow the surveillance
have reported an increasing incidence possibly due to program. This approach has been reported to be both
increased overall survival and earlier age of onset.109,110 safe and effective in selected cases where it was per-
Frequent examination of the remaining testis after treat- formed although surgeons must be aware of the potential
ment of a unilateral tumor, including self-examination by for anomalous vasculature.116,117
patients, is important to detect a second GCT early when
small and confined to the testis. Bilateral inguinal
Inflammatory Bowel Disease
orchiectomy has been the standard management in this
situation. However, patients then require life-long Preexisting morbid conditions like IBD have to be taken
androgen replacement therapy, which may be associated into consideration when RT is planned. Radiation leads
with sexual dysfunction, mood swings, and a general to increased gastrointestinal bleeding, peptic ulceration,
impairment of quality of life. Partial orchidectomy with and long-term incidence of stenosis.
preservation of some normal testicular tissue and andro-
gen production has been proposed as an alternative to
SEMINOMA IN PATIENTS WITH
orchiectomy.111–113 Heidenreich and coworkers reported
IMMUNOSUPPRESSION AND HUMAN
52 metachronous and 17 synchronous bilateral testicular
IMMUNODEFICIENCY VIRUS INFECTION
germ cell tumor patients who underwent an organ-spar-
ing approach. As expected, approximately 60% had semi- Testicular neoplasms appear to be 20 to 50 times more
noma, 20% embryonal carcinoma, 15% mature prevalent among patients on immunosuppression com-
teratoma, and 8% mixed GCT. The mean tumor diame- pared to the general population and in patients who are
ter was 15 mm. Eighty-two percent of patients had asso- seropositive for human immunodeficiency virus
ciated TIN and the residual testis was treated with 18 Gy (HIV).118,119 The interpretation of staging investiga-
of local radiation. On mean follow-up of 91 months (3 to tions for seminoma can be difficult in the later patients
191 months) 98.6% patients had no evidence of disease because of the benign lymphadenopathy that is fre-
and only 1 had died of disease. There was no local relapse quently associated with HIV infection. A compilation of
in 46 patients who received local radiation. Testosterone the reported experience with testicular tumors in HIV-
levels were normal in 85% of patients, and clinical positive patients showed a higher than expected fre-
hypogonadism occurred in about 10%. For tumors of 20 quency of stage II disease (58% versus 20%).119 This
mm and less, tumor enucleation may be a reasonable may be attributable at least in part to the false assign-
alternative to bilateral orchiectomy. ment of patients with HIV to stage II because of benign
paraaortic lymphadenopathy. However, it is also possi-
ble that seminoma may have a more aggressive clinical
Horseshoe or Ectopic Kidney
course in HIV-positive patients. The majority of
Horseshoe kidney occurs in approximately 1 in 400 of patients described in the literature have received stan-
the general population. There is an association between dard treatment. RT and chemotherapy appear to be well
renal fusion abnormalities and cryptorchidism, which in tolerated except in patients with very advanced
turn is associated with an increased incidence of testicu- immunosuppression. 119 Most patients are cured.
lar neoplasms.114 There are two main problems in the Overall survival is usually determined by the severity of
management of GCTs associated with horseshoe or immunosuppression and the complications of the
pelvic kidney. The first is related to the technical prob- acquired immunodeficiency syndrome (AIDS) rather
lem of delivery of RT in patients with seminoma. In a than by seminoma.119,120
number of cases of horseshoe kidney, a large part of the Following renal transplant there are cases reported of
renal parenchyma lies within the standard radiation vol- seminomas.49,121,122 In such patients stages I and II have
ume and directly overlies the regional lymph nodes. The usually been treated with postoperative RT, but the con-
delivery of a standard radiation dose would be associated cerns regarding potential damage to the transplanted
with an unacceptable risk of radiation nephritis. The sec- kidney may dictate surveillance. Short-term follow-up of
ond problem is related to the possible abnormalities in these patients does not suggest a higher risk of relapse,
lymphatic drainage of the testis and therefore the possi- but the available data are limited.
Chapter 35 Seminoma: Management and Prognosis 591

THE NONCOMPLIANT PATIENT mended, and a 5-year survival of 73% has been reported
with the use of cisplatin + vincristine + methotrexate +
As the cure rate for patients with stages I and II semino-
bleomycin + actinomycin D + cyclophosphamide +etopo-
mas approaches 100%, problems with patient compli-
side (POMB/ACE) chemotherapy.128
ance with treatment or follow-up recommendations may
affect the outcome more than choice of therapy. Every
large cancer center has experience with patients refusing SUMMARY
conventional therapy or failing to attend for follow-up
Testicular seminoma is highly curable with currently
with eventual death due to disease. Although Hao et al.49
available treatments. There is good evidence that
reported that compliance with clinical evaluations in
patients with stage I disease can be managed equally well
patients with nonseminomas was 61.5% in year 1 and
after radical orchiectomy with either adjuvant RT or sur-
35.5% in year 2, compliance with CT was only 25% and
veillance. Current areas of investigation in stage I
11.8% in years 1 and 2, respectively. The compliance
patients include optimization of the RT treatment tech-
with this surveillance program was poor but this study
nique to minimize toxicity and the use of 1 or 2 cycles of
was too small to demonstrate whether poor compliance
single-agent carboplatin instead of RT or surveillance.
adversely affects overall survival; other studies show no
Patients with small bulk stage II disease are treated effec-
relation to survival. Patient education regarding the diag-
tively with RT. Patients with bulky stage II or stage III
nosis, available treatment options and outcome in testis
disease should receive 4 cycles of etoposide and cisplatin
tumors, and need for regular evaluation should be given
(EP) or equivalent chemotherapy. Overall, more than
high priority.
95% of patients with seminoma are cured with this strat-
egy. The current challenge for clinicians is to maintain
MANAGEMENT OF SEMINOMA IN CRYPTORCHID high cure rates while minimizing toxicity and individual-
TESTES izing therapy to the specific social, emotional, and eco-
nomic circumstances of each patient. It is important to
The management of seminoma presenting in cryptorchid
continue to study the long-term consequences of current
testis depends on the location of the primary tumor and
treatment strategies, particularly with respect to serious
the extent of the disease. The awareness of orchiopexy
late side effects that might ultimately compromise the
early in childhood appears to have led to a significant
longevity of patients with seminoma diagnosed and
drop in the incidence of patients presenting with tumors
treated decades earlier. The treatment objectives not only
in uncorrected cryptorchids. Giving similar doses of radi-
include long-term survival but should include a near nor-
ation and adjusting the size of the paraaortic and pelvic
mal quality of life. It is important that modifications in
radiation fields to cover the known extent of disease can
surgery, radiation, and chemotherapy, as well as advanced
achieve comparable survival.123
care to improve the quality of life in terms of sexual func-
tion, fertility, and problems of hormone replacement be
EXTRAGONADAL GERM CELL TUMORS integrated into current management strategies as they
become available, and that the doctrine established as a
Extragonadal germ cell tumors (EGCTs) have a similar
result of years of successful treatment not present an
histology to testicular GCTs but are found in other parts
insurmountable barrier to changes that may enhance the
of the body, in the absence of a testicular mass. They
therapeutic ratio and improve overall patient outcome.
account for 1% to 5% of all GCTs and, like testicular
GCTs, tend to occur in young men, although the median
age of presentation is 5 to 10 years older than with testic- REFERENCES
ular GCTs.124 Ten percent of adult EGCTs occur in
women, usually as ovarian dysgerminomas. In infants, 1. McGlynn KA, et al: Trends in the incidence of testicular
EGCTs are more common than testicular primary tumors germ cell tumors in the United States. Cancer 2003;
(usually sacrococcygeal teratomas).125,126 An increased 97(1):63–70.
incidence of EGCTs is seen with Klinefelter’s syndrome.126 2. Weir HK, Marrett LD, Moravan V: Trends in the
Overall, patients with extragonadal GCTs (especially incidence of testicular germ cell cancer in Ontario by
histologic subgroup, 1964–1996. CMAJ 1999;
NSGCTs) have a worse prognosis than patients with tes-
160(2):201–205.
ticular primaries. In a recent review of published results of 3. Warde P, Jewett MA: Surveillance for stage I testicular
chemotherapy for mediastinal tumors, 28 of 37 (76%) seminoma: is it a good option? Urol Clin North Am
patients with seminomatous histology were disease free in 1998; 25(3):425–433.
comparison to 90 of 204 patients (44%) with nonsemino- 4. Milosevic MF, Gospodarowicz M, Warde P:
matous tumors.127 The use of aggressive regimens for Management of testicular seminoma. Semin Surg Oncol
patients with nonseminomatous tumors has been recom- 1999; 17(4):240–249.
592 Part VI Testis

5. Parkin DM, Muir CS: Cancer Incidence in Five (carcinoma in situ)—surgical aspects. APMIS 2003;
Continents. Comparability and quality of data. IARC Sci 111(1):64–68 [Discussion 68-9].
Publ 1992; 120:45–173. 25. Wanderas EH, Fossa SD, Tretli S: Risk of a second germ
6. Parker SL, et al: Cancer statistics, 1997. CA Cancer cell cancer after treatment of a primary germ cell cancer
J Clin 1997; 47(1):5–27. in 2201 Norwegian male patients. Eur J Cancer 1997;
7. Brown N: Cryptorchidism—a significant risk factor for 33(2):244–252.
testicular cancer. Br J Gen Pract 1991; 41(348):305. 26. Osterlind A, et al: Risk of bilateral testicular germ cell
8. Prener A, Engholm G, Jensen OM: Genital anomalies cancer in Denmark: 1960–1984. J Natl Cancer Inst 1991;
and risk for testicular cancer in Danish men. 83(19):1391–1395.
Epidemiology 1996; 7(1):14–19. 27. Warde P, et al: Stage I testicular seminoma: results of
9. Power DA, et al: Trends in testicular carcinoma in adjuvant irradiation and surveillance. J Clin Oncol 1995;
England and Wales, 1971–1999. BJU Int 2001; 13(9):2255–2262.
87(4):361–365. 28. Warde P, et al: Prognostic factors for relapse in stage I
10. Huyghe E, Matsuda T, Thonneau P: Increasing seminoma managed by surveillance: a pooled analysis.
incidence of testicular cancer worldwide: a review. J Urol J Clin Oncol 2002; 20(22):4448–4452.
2003; 170(1):5–11. 29. Donohue JP, Zachary JM, Maynard BR: Distribution of
11. Abratt RP, Reddi VB, Sarembock LA: Testicular cancer nodal metastases in nonseminomatous testis cancer.
and cryptorchidism. Br J Urol 1992; 70(6):656–659. J Urol 1982; 128(2):315–320.
12. Klein FA, et al: Inguinal lymph node metastases from 30. Weissbach L, Boedefeld EA, FTTTS Group:
germ cell testicular tumors. J Urol 1984; 131(3): Localization of solitary and multiple metastases in stage
497–500. II nonseminomatous testis tumor as basis for a modified
13. Rapley EA, et al: Localisation of susceptibility genes for staging lymph node dissection in stage I. J Urol 1987;
familial testicular germ cell tumour. APMIS 2003; 138:77–82.
111(1):128–133 [Discussion 33-5]. 31. Gauwitz MD, Zagars GK: Treatment of seminoma
14. Swerdlow AJ, et al: Risks of breast and testicular cancers arising in cryptorchid testes. Int J Radiat Oncol Biol
in young adult twins in England and Wales: evidence on Phys 1992; 24(1):153–159.
prenatal and genetic aetiology. Lancet 1997; 32. Li YX, et al: Seminoma arising in corrected and
350(9093):1723–1728. uncorrected inguinal cryptorchidism: treatment and
15. Klotz LH: Why is the rate of testicular cancer prognosis in 66 patients. Int J Radiat Oncol Biol Phys
increasing? CMAJ 1999; 160(2):213–214. 1997; 38(2):343–350.
16. Ruther U, et al: Role of human chorionic gonadotropin 33. Mason MD, et al: Inguinal and iliac lymph node
in patients with pure seminoma. Eur Urol 1994; involvement in germ cell tumours of the testis:
26(2):129–133. implications for radiological investigation and
17. Edler von Eyben F, et al: Serum lactate dehydrogenase for therapy. Clin Oncol (R Coll Radiol) 1991;
isoenzyme. 1. An early indicator of relapse in patients 3(3):147–150.
with testicular germ cell tumors. Acta Oncol 1995; 34. Giwercman A, et al: Evidence for increasing incidence of
34(7):925–929. abnormalities of the human testis: a review. Environ
18. Nielsen OS, et al: Is placental alkaline phosphatase Health Perspect 1993; 101(Suppl 2):65–71.
(PLAP) a useful marker for seminoma? Eur J Cancer 35. Giwercman A, et al: Current concepts of radiation
1990; 26(10):1049–1054. treatment of carcinoma in situ of the testis. World J Urol
19. Herr HW, Sheinfeld J: Is biopsy of the contralateral 1994; 12(3):125–130.
testis necessary in patients with germ cell tumors? J Urol 36. Rorth M, et al: Carcinoma in situ in the testis. Scand
1997; 158(4):1331–1334. J Urol Nephrol Suppl 2000; 205:166–186.
20. Capelouto CC, et al: A review of scrotal violation in 37. Petersen PM, et al: Endocrine function in patients
testicular cancer: Is adjuvant local therapy necessary? treated for carcinoma in situ in the testis with irradiation.
J Urol 1995; 153(3 Pt 2):981–985. APMIS 2003; 111(1):93–98 [Discussion 98-9].
21. Skakkebaek NE, et al: Carcinoma-in-situ of the testis: 38. Freeman A, Rowbotham C, Parkinson MC: The
possible origin from gonocytes and precursor of all types prevalence of testicular microlithiasis in an asymptomatic
of germ cell tumours except spermatocytoma. Int population of men 18 to 35 years old. J Urol 2003;
J Androl 1987; 10(1):19–28. 169(4):1474.
22. Giwercman A, von der Maase H, Skakkebaek NE: 39. Holm M, et al: The prevalence of testicular
Epidemiological and clinical aspects of carcinoma in situ microlithiasis in an asymptomatic population of men 18
of the testis. Eur Urol 1993; 23(1):104–110 [Discussion to 35 years old. J Urol 2002; 168(3):1108 [author reply
111-4]. 1108-9].
23. Dieckmann KP, Loy V: Prevalence of contralateral 40. Peterson AC, et al: The prevalence of testicular
testicular intraepithelial neoplasia in patients with microlithiasis in an asymptomatic population of men 18
testicular germ cell neoplasms. J Clin Oncol 1996; to 35 years old. J Urol 2001; 166(6):2061–2064.
14(12):3126–3132. 41. Steele GS, et al: The National Cancer Data Base report
24. Dieckmann KP, Classen J, Loy V: Diagnosis and on patterns of care for testicular carcinoma, 1985–1996.
management of testicular intraepithelial neoplasia Cancer 1999; 86(10):2171–2183.
Chapter 35 Seminoma: Management and Prognosis 593

42. Fossa SD, et al: Optimal planning target volume for 60. Lester SG, Morphis JG II, Hornback NB: Testicular
stage I testicular seminoma: a medical research council seminoma: analysis of treatment results and failures. Int
randomized trial. Medical Research Council Testicular J Radiat Oncol Biol Phys 1986; 12(3): 353–358.
Tumor Working Group. J Clin Oncol 1999; 17(4):1146. 61. Dosoretz DE, et al: Megavoltage irradiation for pure
43. Travis LB, et al: Treatment-associated leukemia testicular seminoma: results and patterns of failure.
following testicular cancer. J Natl Cancer Inst 2000; Cancer 1981; 48(10):2184–2190.
92(14):1165–1171. 62. Sultanem K, et al: Para-aortic irradiation only appears to
44. Travis LB, et al: Risk of second malignant neoplasms be adequate treatment for patients with stage I
among long-term survivors of testicular cancer. J Natl seminoma of the testis. Int J Radiat Oncol Biol Phys
Cancer Inst 1997; 89(19):1429–1439. 1998; 40(2):455–459.
45. Fiveash J, Sandler HM, Controversies in the 63. Kiricuta IC, Sauer J, Bohndorf W: Omission of the
management of stage I seminoma. Oncology (Huntingt) pelvic irradiation in stage I testicular seminoma: a study
1998; 12(8):1203–1212 [Discussion 1212-21]. of postorchiectomy paraaortic radiotherapy. Int J Radiat
46. von der Maase H, et al: Surveillance following Oncol Biol Phys 1996; 35(2):293–298.
orchidectomy for stage I seminoma of the testis. Eur J 64. Read G, Johnston RJ: Short duration radiotherapy in
Cancer 1993; 29A(14):1931–1934. stage I seminoma of the testis: preliminary results of a
47. Michael H, et al: The pathology of late recurrence of prospective study. Clin Oncol (R Coll Radiol) 1993;
testicular germ cell tumors. Am J Surg Pathol 2000; 5(6):364–366.
24(2):257–273. 65. Thomas GM: Alternative management options to
48. Warde P, et al: Prognostic factors for relapse in stage I radiation therapy for stage I and IIA testicular
testicular seminoma treated with surveillance. J Urol seminoma. Int J Radiat Oncol Biol Phys 1994;
1997; 157(5):1705–1709 [Discussion 1709-10]. 28(2):547–548.
49. Hao D, et al: Compliance of clinical stage I 66. Donohue JP, et al: Primary retroperitoneal lymph
nonseminomatous germ cell tumor patients with node dissection in clinical stage A non-seminomatous
surveillance. J Urol 1998; 160(3 Pt 1):768–771. germ cell testis cancer. Review of the Indiana
50. Sharda NN, Kinsella TJ, Ritter MA: Adjuvant radiation University experience 1965–1989. Br J Urol 1993;
versus observation: a cost analysis of alternate 71(3): 326–335.
management schemes in early-stage testicular seminoma. 67. Schmidberger H, et al: Radiotherapy in stages IIA and
J Clin Oncol 1996; 14(11):2933–2939. IIB testicular seminoma with reduced portals: a
51. Warde P, et al: Long term outcome and cost in the prospective multicenter study. Int J Radiat Oncol Biol
management of stage I testicular seminoma. Can J Urol Phys 1997; 39(2):321–326.
2000; 7(2):967–972 [Discussion 973]. 68. Dieckmann KP, et al: Adjuvant carboplatin treatment for
52. Horwich A, et al: Surveillance following orchidectomy seminoma clinical stage I. J Cancer Res Clin Oncol
for stage I testicular seminoma. Br J Cancer 1992; 1996; 122(1):63–66.
65(5):775–778. 69. Aparicio J, et al: Multicenter study evaluating a dual
53. Fossa SD, Aass N, Kaalhus O: Radiotherapy for policy of postorchiectomy surveillance and selective
testicular seminoma stage I: treatment results and adjuvant single-agent carboplatin for patients with
long-term post-irradiation morbidity in 365 patients. clinical stage I seminoma. Ann Oncol 2003;
Int J Radiat Oncol Biol Phys 1989; 16(2):383–388. 14(6):867–872.
54. Dosmann MA, Zagars GK: Post-orchiectomy 70. Reiter WJ, et al: Twelve-year experience with two
radiotherapy for stages I and II testicular seminoma. Int courses of adjuvant single-agent carboplatin therapy for
J Radiat Oncol Biol Phys 1993; 26(3):381–390. clinical stage I seminoma. J Clin Oncol 2001; 19(1):
55. Hamilton C, et al: Radiotherapy for stage I seminoma 101–104.
testis: results of treatment and complications. Radiother 71. Steiner H, et al: Long-term experience with carboplatin
Oncol 1986; 6(2):115–120. monotherapy for clinical stage I seminoma: a
56. Hamilton C, et al: Radiotherapy for stage I seminoma retrospective single-center study. Urology 2002; 60(2):
testis: results of treatment and complications. Radiother 324–328.
Oncol 1986; 6(2):115. 72. Dieckmann K-P, et al: Adjuvant carboplatin treatment
57. Centola GM, et al: Effect of low-dose testicular for seminoma clinical stage I. J Cancer Res Clin Oncol
irradiation on sperm count and fertility in patients 1996; 122:63–66.
with testicular seminoma. J Androl 1994; 15(6): 73. Tsatalpas P, et al: Diagnostic value of 18F-FDG positron
608–613. emission tomography for detection and treatment
58. Joos H, et al: Endocrine profiles after radiotherapy in control of malignant germ cell tumors. Urol Int 2002;
stage I seminoma: impact of two different radiation 68(3):157–163.
treatment modalities. Radiother Oncol 1997; 74. Hultenschmidt B, et al: Results of radiotherapy for 230
43(2):159–162. patients with stage I-II seminomas. Strahlenther Onkol
59. Fraass BA, et al: Peripheral dose to the testes: the design 1996; 172(4):186–192.
and clinical use of a practical and effective gonadal 75. Vallis KA, et al: Radiotherapy for stages I and II
shield. Int J Radiat Oncol Biol Phys 1985; testicular seminoma: results and morbidity in 238
11(3):609–615. patients. Br J Radiol 1995; 68(808):400–405.
594 Part VI Testis

76. Warde P, et al: Management of stage II seminoma. J Clin clinical aspects. APMIS 1998; 106(1):24–34 [Discussion
Oncol 1998; 16(1):290–294. 34-6].
77. Warszawski N, Schmucking M: Relapses in early-stage 95. Moller H, et al: Incidence of second primary cancer
testicular seminoma: radiation therapy versus following testicular cancer. Eur J Cancer 1993; 29A(5):
retroperitoneal lymphadenectomy. Scand J Urol Nephrol 672–676.
1997; 31(4): 355–359. 96. van Leeuwen FE, et al: Second cancer risk following
78. Hanks GE, Peters T, Owen J: Seminoma of the testis: testicular cancer: a follow-up study of 1909 patients.
long-term beneficial and deleterious results of J Clin Oncol 1993; 11(3):415–424.
radiation. Int J Radiat Oncol Biol Phys 1992; 97. Travis LB, Curtis RE, Hankey BF: Second malignancies
24(5):913–919. after testicular cancer. J Clin Oncol 1995; 13(2):533–534.
79. Lederman GS, et al: Cardiac disease after mediastinal 98. Ruther U, et al: Second malignancies following pure
irradiation for seminoma. Cancer 1987; 60(4):772–776. seminoma. Oncology 2000; 58(1):75–82.
80. Loehrer PJ Sr, et al: Chemotherapy of metastatic 99. Caffo O, et al: Quality of life after radiotherapy for
seminoma: the Southeastern Cancer Study Group early-stage testicular seminoma. Radiother Oncol 2001;
experience. J Clin Oncol 1987; 5(8):1212–1220. 59(1):13–20.
81. Fossa SD, et al: The treatment of advanced metastatic 100. Tinkler SD, Howard GC, Kerr GR: Sexual morbidity
seminoma: experience in 55 cases. J Clin Oncol 1987; following radiotherapy for germ cell tumours of the
5(7):1071–1077. testis. Radiother Oncol 1992; 25(3):207–212.
82. Herr HW, et al: Surgery for a post-chemotherapy 101. Brennemann W, et al: Pretreatment follicle-stimulating
residual mass in seminoma. J Urol 1997; 157(3):860–862. hormone: a prognostic serum marker of spermatogenesis
83. International Germ Cell Cancer Collaborative Group: status in patients treated for germ cell cancer. J Urol
International germ cell consensus classification: a 1998; 159(6):1942–1946.
prognostic factor-based staging system for metastatic 102. Hallak J, et al: Investigation of fertilizing capacity of
germ cell cancers. J Clin Oncol 1997; 15(2):594–603. cryopreserved spermatozoa from patients with cancer.
84. Porcaro AB, et al: Management of testicular seminoma J Urol 1998; 159(4):1217–1220.
advanced disease. Report on 14 cases and review of the 103. Rosenlund B, et al: In-vitro fertilization and
literature. Arch Ital Urol Androl 2002; 74(2):81–85. intracytoplasmic sperm injection in the treatment of
85. Gholam D, et al: Advanced seminoma—treatment results infertility after testicular cancer. Hum Reprod 1998;
and prognostic factors for survival after first-line, 13(2):414–418.
cisplatin-based chemotherapy and for patients with 104. Ravi R, et al: The management of residual masses after
recurrent disease: a single-institution experience in 145 chemotherapy in metastatic seminoma. BJU Int 1999;
patients. Cancer 2003; 98(4):745–752. 83(6): 649–653.
86. Stanton GF, et al: VAB-6 as initial treatment of patients 105. Flechon A, et al: Management of post-chemotherapy
with advanced seminoma. J Clin Oncol 1985; residual masses in advanced seminoma. J Urol 2002;
3(3):336–339. 168(5):1975–1979.
87. Williams MP, Husband JE, Heron CW: Intrathoracic 106. Duchesne GM, et al: Radiotherapy after chemotherapy
manifestations of metastatic testicular seminoma: a for metastatic seminoma—a diminishing role. MRC
comparison of chest radiographic and CT findings. Am Testicular Tumour Working Party. Eur J Cancer 1997;
J Roentgenol 1987; 149(3):473–475. 33(6):829–835.
88. Mencel PJ, et al: Advanced seminoma: treatment results, 107. Ogan K, et al: Laparoscopic versus open retroperitoneal
survival, and prognostic factors in 142 patients. J Clin lymph node dissection: a cost analysis. J Urol 2002;
Oncol 1994; 12(1):120–126. 168(5):1945–1949 [Discussion 1949].
89. Miller KD, et al: Salvage chemotherapy with vinblastine, 108. Weissbach L, Bussar-Maatz R: hCG-positive seminoma.
ifosfamide, and cisplatin in recurrent seminoma. J Clin Eur Urol 1993; 23(Suppl 2):29–32.
Oncol 1997; 15(4):1427–1431. 109. Patel SR, Richardson RL, Kvols L: Synchronous and
90. Toner GC, et al: The management of testicular cancer in metachronous bilateral testicular tumors. Mayo Clinic
Victoria, 1988–1993. Urology Study Committee of the experience. Cancer 1990; 65(1):1–4.
Victorian Co-operative Oncology Group. Med J Aust 110. Coogan CL, et al: Bilateral testicular tumors:
2001; 174(7):328–331. management and outcome in 21 patients. Cancer 1998;
91. Mackey JR, Venner P: Seminoma with isolated central 83(3):547–552.
nervous system relapse, and salvage with craniospinal 111. Heidenreich A, Weissbach L, Holt W, et al: Organ
irradiation. Urology 1998; 51(6):1043–1045. sparing surgery for malignant germ cell tumor of the
92. Bokemeyer C, et al: Treatment of brain metastases in testis. J Urol 2001; 166(6):2161–2165.
patients with testicular cancer. J Clin Oncol 1997; 112. Richie JP: Organ sparing surgery for malignancy
15(4):1449–1454. germ cell tumor of the testis. J Oncl 2003; 21(1):87.
93. Senturia YD, Peckham CS, Peckham MJ: Children 114. Li, FP, Fraumeni JF: Testicular cancers in children:
fathered by men treated for testicular cancer. Lancet epidemiologic characteristics. J Natl Cancer Inst 1972;
1985; 2(8458):766–769. 48(6):1575–1581.
94. Petersen PM, Skakkebaek NE, Giwercman A: Gonadal 115. Elyan SA, Reed DH, Ostrowski MJ, et al: Problems in
function in men with testicular cancer: biological and the management of testicular seminoma associated with a
Chapter 35 Seminoma: Management and Prognosis 595

horseshoe kidney. Clin Oncol (R Coll Radiol) 1990; 123. Sham JS, Choy P, Chan KW et al: Seminoma of
2(3):163–167. normally-descended and cryptorchid testis. Eur J Surg
116. Key DW, Moyad R, Grossman HB: Seminoma Oncol 1990; 16(1):33–36.
associated with crossed fused renal ectopia. J Urol 1990; 124. Shirdasani RA: Extragonadal germ-cell tumours. In
143(5):1015–1016. Scher H, Raghavan D, Leibel S, Lange P (eds):
117. Sogani PC, Whitmore WF Jr: Retroperitoneal Principles and Practice of Genitourinary Oncology,
lymphadenectomy for germ cell tumor of testis in p 751. Philadelphia, Lippincott-Raven Publishers,
association with horseshoe kidney. Urology 1981; 1997.
18(5):446–452. 125. Mann JR, Pearson D, Barrett A, et al: Results of the
118. Lyter DW, Bryant J, Thackeray R, et al: Incidence of United Kingdom Children’s Cancer Study Group’s
human immunodeficiency virus-related and nonrelated malignant germ cell tumor studies. Cancer 1989;
malignancies in a large cohort of homosexual men. J Clin 63(9):1657–1667.
Oncol 1995; 13(10):2540–2546. 126. Nichols CR, Heereina NA, Palma C, et al: Klinefelter’s
119. Leibovitch I, Baniel J, Rowland RA, et al: Malignant syndrome associated with mediastinal germ cell
testicular neoplasms in immunosuppressed patients. neoplasms. J Clin Oncol 1987; 5(8):1290–1294.
J Urol 1996; 155(6):1938–1942. 127. Droz JP, Horwich A: Extragonadal germ cell tumours. In
120. Timmerman JM, Northfelt DW, Small EJ: Malignant Vogelzang NJ, Scardino P (eds): Comprehensive
germ cell tumors in men infected with the human Textbook of Genitourinary Oncology. Baltimore,
immunodeficiency virus: natural history and results of Williams & Wilkins, 1995.
therapy. J Clin Oncol 1995; 13(6):1391–1397. 128. Bower M, Brock C, Holden L, et al: POMB/ACE
121. Dieckmann KP, Due W, Offermann G: Testicular chemotherapy for mediastinal germ cell tumours. Eur
seminoma in an immunosuppressed renal transplant J Cancer 1997; 33(6):838–842.
recipient. Br J Urol 1989; 63(5):549–550. 129. Worde P, Specht L, von der Masse H, et al: Prognostic
122. Villalona-Calero MA, Ducker T, Holasek M, et al: factors for relapse in Stage I seminoma managed by
Management of testicular seminoma following organ surveillance (abstract). J Urol 1999; 161:158.
transplantation. Med Pediatr Oncol 1992; 20(4):338–340.
C H A P T E R

36 Nonseminomatous Germ Cell Testis


Tumors: Management and Prognosis
Randall G. Rowland, MD, PhD

The survival rates for patients with testis cancer have monary lesion or back pain from large retroperitoneal
improved dramatically over the last 30 years. This has adenopathy.
been especially true for nonseminomatous germ cell testis Physical examination of the testicle still plays a major
tumors (NSGCTT). There are several factors involved role in the evaluation of the patient. It is important to dif-
in this improvement in survival: improved diagnostic ferentiate a mass in the testicle versus the epididymis or
tools, such as serum markers; better imaging studies; cord structures.
improved surgical techniques; effective chemotherapeu- Scrotal ultrasound is perhaps the best and most read-
tic agents; multimodal therapy when appropriate; and ily available imaging study for suspected GCTTs.
increased public awareness. Ultrasound scans facilitate the location of scrotal masses,
The high rate of cure of NSGCTT has allowed us as well as giving us information on the homogeneity or
to concentrate on reducing the morbidity and mortal- heterogeneity of the mass.
ity of the treatments, while maintaining the high rate of Scrotal ultrasounds may give some insight regarding
efficacy. Fertility can even be preserved in the majority the cell type(s) in a mass. Pure seminomas tend to be very
of patients using modified templates and nerve-sparing homogeneous masses (Figure 36-1), while mixed GCTTs
techniques during radical retroperitoneal lymph node may have a heterogeneous echo pattern (Figure 36-2).
dissection (RPLND). The finding of cystic structures in a mass suggests the
This chapter will focus on the diagnoses, staging, presence of teratoma (Figure 36-3).
treatments, and outcomes of NSGCTT. Serum markers are also helpful in making the
diagnosis of testis cancer. Overall approximately
70% of patients with testis cancer will have elevated
DIAGNOSIS
alpha-fetoprotein (AFP) and/or HCG levels in their
Germ cell testis tumors (GCTT) are the most common blood. The clinical test for HCG is a radioim-
solid tumors in males from the age of 20 to 35 years.1 munoassay that measures the beta chain of the HCG
GCTTs have been reported in patients from infants to and is designated B-HCG. Yolk sac cell tumors pro-
age 89 years. NSGCTTs account for approximately 60% duce AFP with rare exception. HCG is made by
of all GCTTs.2 syncytio-trophoblasts, which are present in all cases of
Any mass in the testicle has to be considered a poten- choriocarcinoma and in about 10% of cases of pure
tial GCTT until proven otherwise. GCTTs must also seminoma.
be considered as a possibility in patients with scrotal Lactate dehydrogenase (LDH) is sometimes used as a
pain and/or swelling, the presence of a hydrocele, or measure of tumor bulk; however, the author does not
a hematocele after relatively minor scrotal trauma. find this text useful in clinical practice. Placental alkaline
Production of human chorionic gonadotropin (HCG) phosphatase (PLAP) is present in up to 84% of patients
by some tumors causes patients to present with gyneco- with seminoma. PLAP is falsely elevated in a high per-
mastia or breast tenderness. Metastases of GCTT have centage of smokers, which could negate the value of the
made some patients present with hemoptysis from pul- marker.3

596
Chapter 36 Nonseminomatous Germ Cell Tests Tumors: Management and Prognosis 597

Figure 36-1 Sagittal ultrasound of testis with a central mass, which was a seminoma. Note
the homogeneity of the mass.

RADICAL ORCHIECTOMY VERSUS PARTIAL Transscrotal needle biopsy of testicular masses is dis-
ORCHIECTOMY couraged for two reasons. First, there can easily be a sam-
pling error that would not give a true representation of
The standard approach for making the definitive diag-
the pathology present. Second, there is a possibility for
nosis of testicular cancer has been the radical orchiec-
cross-contamination for the testicle lymphatics with the
tomy performed by removing the testicle using an
scrotal/inguinal lymphatics.
inguinal approach. This approach helps prevent cross-
contamination of the testicular lymphatics to the
inguinal lymphatics. The vas deferens and the gonadal
PATHOLOGY
vessels are ligated and divided at the level of the internal
inguinal ring. Histologic study of the orchiectomy specimen should
An organ-sparing approach has been advocated by include documenting the cell type(s) present in the tumor
some authors.4,5 In this approach, the testicle is still and the T stage of the tumor. Extensive sampling of the
delivered through an inguinal incision, except the mass in testis should be performed to allow a careful and thor-
the testicle is excised. The testis is reconstructed and ough determination of the cell types that are present.
returned to the scrotum. This technique is a benefit in T stage can be accurately assessed by careful observation
patients with a solitary testis or an intratesticular lesion and extensive histologic sampling.
that is questionable with respect to its being a testicular Tumors can be intratubular (CIS) or invasive. The cell
tumor. The down-side of a local excision is the potential types of germ cell tumors that occur are shown in Table
for leaving a finger of a lesion or a satellite lesion behind. 36-1. Approximately 40% of patients present with pure
598 Part VI Testis

Figure 36-2 Sagittal ultrasound of testis with a mass, which was a mixed germ cell tumor.
Note the more heterogeneous character of the mass.

seminoma and 60% present with nonseminomatous cell taken to evaluate the contralateral testis since bilateral syn-
elements either as a pure cell type or more commonly as chronous or metachronous testicular tumors do occur.
mixed cell type tumors.2 The determination of cell types
present in a testicular tumor is very important in terms of
Serum Markers
treatment. Pure seminomas, including spermatocytic
seminomas, are treated with a different approach than AFP and HCG should be rechecked after orchiectomy in
tumors with nonseminomatous elements. all patients, but especially in those patients with elevated
Table 36-2 shows the American Joint Commission on serum markers prior to orchiectomy. If the tumor was
Cancer’s (AJCC) definitions of the pathologic T stages of localized in the testis only, orchiectomy should cause
testicular tumors.6 the markers to return to normal. If tumor remains, the
markers may not return to normal.
When following the serum marker level, the time after
CLINICAL STAGING
orchiectomy and the serum half-life of each marker must
Once the histologic diagnosis of testicular cancer is docu- be considered. The serum half-life of AFP is approxi-
mented, the patient should have clinical staging performed mately 5 days and that of HCG is about 1 day. Markers
to allow the formulation of a treatment plan to best fit the that either fall at a lower than expected rate or rising are
patient’s needs. Clinical staging involves assessment of the indicative of persistent disease.
patient using tumor markers before and after orchiectomy, When LDH is used as a marker, it is usually consid-
radiologic imaging studies of the chest, abdomen and ered in terms of its percentage or number of times the
pelvis, and physical examination to document any normal value. LDH has a half-life of approximately
adenopathy or abdominal masses. Care should also be 3 days.
Chapter 36 Nonseminomatous Germ Cell Tests Tumors: Management and Prognosis 599

Figure 36-3 Sagittal ultrasound of testis with two adjacent masses. Note the hypoechoic
areas, which correspond to cystic areas of teratoma.

Table 36-1 Germ Cell Testicular Tumors Table 36-2 Pathologic Stage of The Primary Tumor (T)
Precursor lesions Intratubular germ cell neoplasia (CIS) pTX Primary tumor cannot be assessed

Tumors of one Seminoma pT0 No evidence of primary tumor (e.g., histologic


histologic type Spermatocytic seminoma scar in testis)
Embryonal carcinoma
Yolk sac tumor (endodermal pTis Intratubular germ cell neoplasia (carcinoma in situ)
sinus tumor)
Choriocarcinoma PT1 Tumor limited to the testis and epididymis without
Teratoma vascular/lymphatic invasion; tumor may invade into
Mature the tunica albuginea but not the tunica vaginalis
Immature
pT2 Tumor limited to the testis and epididymis with
With malignant component(s)
vascular/lymphatic invasion, or tumor extending
Monodermal variants
through the tunica albuginea with involvement
Carcinoid tumor
of the tunica vaginalis
Primitive neuroectodermal
tumor PT3 Tumor invades the spermatic cord with or without
vascular/lymphatic invasion
Tumors of more Mixed germ cell tumors
than one (specifically individual types) pT4 Tumor invades the scrotum with or without
histologic type vascular/lymphatic invasion

These T stages are based on the pathologic findings from


orchiectomy specimens.
600 Part VI Testis

Imaging Studies Physical Examination


The goal of imaging studies is to detect any nodal or vis- Patients should be carefully examined for adenopathy
ceral tumor involvement. Based on the known patterns of (inguinal, axillary, or cervical). The physician needs to
spread of testicular tumors by lymphatics and rarely by check for neck, abdominal, or scrotal masses. The con-
vasculature, the studies need to cover the chest, both pul- tralateral testis should be examined with great care for
monary parenchyma and the mediastinum, the abdomen, any possible intratesticular masses. Scrotal ultrasound
again the viscera and the retroperitoneal lymph nodes, is again very helpful in evaluating the contralateral
and the pelvis. testis.
The chest was initially evaluated by standard chest
x-rays and whole lung tomograms. With the advent of
Clinical Stage Designations
computed axial tomography (CT) and subsequent
improvement in CT techniques, CT scans have become Assigning a clinical stage requires consideration of
the most frequent modality for doing initial clinical stag- the pathologic T stage (see Table 36-2), the clinical
ing of the chest. Chest involvement more frequently N stage (Table 36-3), the clinical M stage (Table 36-4),
occurs as peripheral pulmonary nodules in cases of low and the S stage (Table 36-5).6 The T stage is based on the
volume metastases and with the addition of mediastinal pathologic findings from the orchiectomy specimen. The
adenopathy in more advanced cases. It is important to clinical regional node stage (N) is based on imaging stud-
examine the lung fields using both soft tissue settings and ies of the abdomen and pelvis. The distant metastases
bone window settings. This allows parenchymal lesions stage (M) is assigned based on evidence from imaging
to be evaluated for the presence of calcification, which studies as well. The serum marker stage (S) is determined
leads to a finding of pulmonary granuloma rather than by the preorchiectomy marker determinations for the
metastasis. Figures 36-4 and 36-5 show CT scans of a initial clinical staging.6
patient with metastatic pulmonary lesions. Notice that The pT, N, M, and S data are used to assign the
the lesions visible in Figure 36-4 do not appear in Figure patient to a clinical stage group as shown in Table 36-6.
36-5, which indicates a lack of calcification. These clinical stage groups are used as a basis for treatment
The abdomen and retroperitoneum were originally recommendations and outcomes analysis.
evaluated by intravenous pyelograms (IVPs) to look for
evidence of displacement of the kidneys or ureters by
TOOLS OF TREATMENT
retroperitoneal masses. The CT scan made it possible
to much more accurately evaluate the abdominal viscera The various treatment modalities for testis cancer,
and retroperitoneum. There has been some debate including observation, are presented in the following
related to the size threshold of retroperitoneal lymph prior to specific treatment recommendation for the
nodes that is considered abnormal and likely to repre- clinical stage groups.
sent metastases. Many authors have picked a 1-cm
threshold in evaluating retroperitoneal lymph nodes.
Observation
Using this criterion, the sensitivity has been reported at
approximately 70%. The specificity has been approxi- In patients with a low risk for nodal or metastatic disease,
mately 94%.7 Liebovitch and his associates at Indiana observation may be appropriate. The criteria for observing
reported setting their size threshold at 3 mm for lymph patients who present with localized testicular cancer are
nodes that are in the areas most likely to have nodal metas- shown in Table 36-7. Primary tumor stage and cell types
tases and kept the 1-cm threshold for any other adenopa- present along with the cell type proportions are used
thy. This increased the sensitivity to 91% but decreased along with imaging study findings and marker levels to
the specificity to 52%.8 Using CT techniques, the sensi- determine eligibility for observation.
tivity will most likely not rise about 75% because approx- Table 36-8 presents the tests and examinations recom-
imately 25% of patients who have normal imaging studies mended for use in patients who will have observation as
and still undergo surgical staging will have microscopic or their primary treatment modality. The frequency recom-
small volume macroscopic retroperitoneal metastases.9 mended for tests and examinations decreases with time
Figure 36-6 shows typical CT findings in a patient with since the risk of recurrence diminishes with time.
low volume retroperitoneal lymphadenopathy, while
Figure 36-7 shows higher volume retroperitoneal disease.
Primary Retroperitoneal Lymph Node Dissection:
MRI scan and PET scan have been tried in evaluation
Surgical Staging and Treatment
of patients with testicular cancer.7,10 As of this time,
neither of these techniques offer significant advantages RPLND techniques have undergone significant evolu-
over CT scans in terms of initial staging of patients. tion over the last 40 years. In the 1960s the concept of
Chapter 36 Nonseminomatous Germ Cell Tests Tumors: Management and Prognosis 601

Figure 36-4 Chest x-ray of a patient who presented with hemoptysis from metastatic
testicular cancer.

“more is better” prevailed and the limits of RPLND were fluid emission secondary to the removal of the postgan-
extended. Figure 36-8 illustrates the limits of dissection glionic sympathetic nerve fibers that traversed the field of
that were established.11 The dissection was performed in dissection.
an en bloc style from the diaphragm to the level of the In the 1980s efforts were made to preserve emission in
bifurcation of the common iliac arteries and laterally patients first by modifying the templates of dissection based
from ureter to ureter. The ipsilateral gonadal vessels on the knowledge of the distribution of positive lymph
were removed completely. Although this dissection was nodes in patients with low volume metastatic disease.12,13
performed safely with low morbidity and mortality rates, Figures 36-9 and 36-10 show the limits of dissection gen-
virtually all patients lost their ability to have ejaculatory erally adopted for low-stage (stage I or stages IIA and IIB)
602 Part VI Testis

Figure 36-5 CT scan of chest of patient with metastases to the lungs (arrows) from a
primary testicular cancer.

disease with right- and left-sided primary tumors, respec- nerves are best identified by dissecting along the poste-
tively. The dissections within the limited templates were rior body wall from lateral to medial until the lumbar
still performed with the en bloc technique. This offered sympathetic chain is identified. The use of these prospec-
preservation of the contralateral sympathetic systems. The tive nerve-sparing techniques has resulted in preservation
success rates at preserving emission were greater on right- of emission in almost all patients.16
sided dissection than on the left-sided dissection. A laparoscopic approach to RPLND (LRPLND) for
Later in the 1980s and early 1990s a prospective low-stage disease was first performed in 1992 by two
nerve-sparing technique was developed and adopted European groups.17,18 These series reported favorable
using the modified templates. A better understanding of initial results for conversion to open procedures, 1/34
the anatomy of the lumbar sympathetic nerves allowed and 2/125, respectively.17,18 After the learning curve had
this technique to be applied very successfully in terms of been overcome, the mean operating times were 248 min-
preservation and maintenance of the efficacy of the dis- utes in Rossweiler’s series, 219 minutes for clinical stage I
section as measured by continued low tumor relapse patients in Jauetschek’s series for clinical stage I patients,
rates.14,15 Figure 36-11 shows a diagram of the lumbar and 226 minutes for clinical stage IIB patients. In both
sympathetics and the postganglionic branches as would series together there was only one retroperitoneal recur-
be seen in a right-sided modified template dissection. rence.
The postganglionic fibers are best identified as they Ogan et al.19 compared the cost of LRPLND versus
emerge under the medial edge of the vena cava in the open RPLND. Open RPLND was less costly at $7162
interaortocaval zone. Figure 36-12 shows the typical than LRPLND at $7804 for operative costs. LRPLND
arrangements of the left lumbar sympathetic fibers as showed a cost advantage for hospital stay costs. If an
they travel anteriorly and caudally onto the aorta. These LRPLND patient’s surgery took under 5 hours and his
Chapter 36 Nonseminomatous Germ Cell Tests Tumors: Management and Prognosis 603

Figure 36-6 Abdominal CT scan of a patient with a prominent small interaortocaval lymph
node consistent with metastatic testicular cancer.

hospital stay was less than 2.2 days, the overall cost of any residual disease apparent on physical examination or
LRPLND was less than open surgery. imaging studies.
With the increase in the number of surgeons per- Once gross nodal involvement occurs, retrograde lym-
forming laparoscopic surgery, more patients will have the phatic flow can occur with the subsequent spread of tumor
opportunity to be treated laparoscopically. It should be to nodes that would not ordinarily be involved. As a result
noted, however, that LRPLND is an advanced laparo- the limits of dissection are usually those of the full bilateral
scopic procedure that should only be performed by template (see Figure 36-8). Depending on the location of
advanced laparoscopists. nodal involvement noted on imaging studies, the nerve-
The AJCC has defined classifications for pathologic sparing technique may be used in zones where no obvious
nodal status that are valuable for planning the treatment nodal involvement has occurred. When a contralateral
for patients and assessing results of these treatments. zone can be dissected with a nerve-sparing technique, up
Table 36-9 shows these definitions. to 89% of the patients can have preservation of emission.20
Laparoscopic postchemotherapy RPLND (LPCR-
PLND) has been reported by Palese et al.21 Five of
Postchemotherapy RPLND
7 patients had successful completion of laparoscopic pro-
Generally, patients with bulky retroperitoneal disease cedures. Two required conversion to open surgery. There
(stage IIc) or disseminated disease (stages IIIA to IIIC) were 3 cases with major complications and 1 with minor
are treated initially with systemic chemotherapy. Surgical complications. The authors concluded that LPCRPLND
resection is usually used in those patients who do not should be attempted only in selected patients with small
achieve a complete response to chemotherapy to resect volume radical masses.
604 Part VI Testis

Figure 36-7 Abdominal CT scan of a patient with bulky retroperitoneal metastases (stage
IIC), which compresses the vena cava.

Table 36-3 Clinical Regional Node Stage (N)


NX Regional lymph nodes cannot be assessed

N0 No regional lymph node metastasis

N1 Metastasis with a lymph node mass 2 cm or less in


greatest dimension; or multiple lymph nodes, Table 36-4 Distant Metastases (M)
none more than 2 cm in greatest dimension MX Distant metastasis cannot be assessed
N2 Metastasis with a lymph node mass more than 2 cm M0 No distant metastasis
but not more than 5 cm in greatest dimension; or
multiple lymph nodes, any one mass greater than M1 Distant metastasis
2 cm but not more than 5 cm in greatest
dimension M1a Nonregional nodal or pulmonary metastasis

N3 Metastasis with a lymph node mass more than 5 cm M1b Distant metastasis other than to nonregional
in greatest dimension lymph nodes and lungs

These nodal stages are based on imaging studies. Evidence of metastases is based on imaging studies.
Chapter 36 Nonseminomatous Germ Cell Tests Tumors: Management and Prognosis 605

Table 36-5 Serum Marker Status (S) Table 36-7 Criteria for Primary Observation
SX Marker studies not available or not performed Primary tumor: pT1

S0 Marker study levels within normal limits AFP, B-hCG: normal or return to normal after or
orchiectomy
S1 LDH < 1.5 × N* AND
hCG (mIU/ml) < 5000 AND Embryonal carcinoma component ≤ 40%
AFP (ng/ml) < 1000
No teratomatous elements
S2 LDH 1.5 – 10 × N OR
hCG (mIU/ml) 5000–50,000 OR No choriocarcinoma
AFP (ng/ml) 1000-10,000
No nodal or metastatic masses by imaging studies
S3 LDH > 10 × N OR (N0, M0)
HCG (mIU/ml) > 50,000 OR
AFP (ng/ml) > 10,000

*N indicates the upper limit of normal for the LDH assay. Chemotherapy
Primary Short Course or Adjuvant Chemotherapy
for Low-Stage Disease (Stages I, IIA and IIB)
Primary chemotherapy has been a treatment option for
patients with clinical stages I, IIA, or IIIB. Since 1985,
two cycles of bleomycin, etoposide, and cisplatin (BEP)
have been the standard therapy in this setting.22-24
Table 36-6 Clinical Stage Groups Schefer et al.25 reported a trial of a single course of BEP
Stage 0 pTis N0 M0 S0 for high-risk stage I NSGCTT.
In the adjuvant setting, the Indiana group has reported
Stage 1 PT1–4 N0 M0 SX on 86 patients receiving 2 courses of BEP after RPLND
for pN1 disease or pN2 disease.22 Kondagunta and
Stage IA pT1 N0 M0 S0
Motzer26 published the Memorial Sloan-Kettering
Stage IB pT2 N0 M0 S0 Cancer Center experience using cisplatin and etoposide
pT3 N0 M0 S0 (EP) as adjuvant treatment for pN2 patients.
pT4 N0 M0 S0

Stage IS Any pT/Tx N0 M0 S1–3 Primary Chemotherapy for High-Stage Disease

Stage II Any pT/Tx N1–3 M0 SX


The addition of cisplatin to Velban and bleomycin (PVB)
revolutionized the treatment of metastatic testicular can-
Stage IIA Any pT/Tx N1 M0 S0 cer in 1974.27 The standard treatment of metastatic tes-
Any pT/Tx N1 M0 S1 ticular cancer changed to cisplatin, etoposide, and
bleomycin (BEP) in 1984 after a randomized trial showed
Stage IIB Any pT/Tx N2 M0 S0 that the BEP treatment was more effective and had a
Any pT/Tx N2 M0 S1
lower rate of toxicity.28
Stage IIC Any pT/Tx N3 M0 S0 The next area to be studied was trying to reduce the
Any pT/Tx N3 M0 S1 intensity of chemotherapy to reduce toxicity while main-
taining efficacy. In order to be able to do this effectively,
Stage III Any pT/Tx Any N M1 SX a system for stratification of the risk level of patients with
high-stage tumors was needed. Several attempts were
Stage IIIA Any pT/Tx Any N M1a S0
Any pT/Tx Any N M1a S1
made to define the risk categories. Starting in 1991 an
international group, the International Germ Cell Cancer
Stage IIIB Any pT/Tx N1–3 M0 S2 Cooperative Group (IGCCCG), was started. In 1997 this
Any pT/Tx Any N M1a S2 group published its validated definitions of good progno-
sis, intermediate prognosis, and poor prognosis groups.29
Stage IIIC Any pT/Tx N1–3 M0 S3 The definitions of the groups for NSGCTs are shown in
Any pT/Tx Any N M1a S3
Table 36-10. These groups are often equated to minimal
Any pT/Tx Any N M1b Any S risk, moderate risk, and high risk for the patients.
Treatment choices are frequently made on the basis of
606 Part VI Testis

Figure 36-8 Drawing of the margins of dissection for a full bilateral RPLND. The hatched
areas represent the right and left suprahilar zones, which are removed en bloc with the
interaortocaval zone and left periaortic zone, respectively.

minimal-to-moderate risk categories versus high-risk 1984 to 1989, vinblastine, ifosfamide, and cisplatin were
categories. used at Indiana for second-line therapy.31 Motzer et al.32
reported the use of 4 courses of paclitaxel, ifosfamide, and
cisplatin (TIP) for second-line chemotherapy.32 Hinton
Second-Line or Salvage Chemotherapy
et al.33 have reported a phase II Eastern Cooperative
For patients who did not achieve a complete response with Oncology Group (ECOG) trial of paclitaxel and gemc-
primary chemotherapy and still had positive markers, sal- itabine in refractory germ cell tumors.
vage or second-line chemotherapy was tried. EP was tried High-dose chemotherapy with antologous bone mar-
starting in 1978 at Indiana for salvage treatment.30 From row transplantation was tried at Indiana University
Chapter 36 Nonseminomatous Germ Cell Tests Tumors: Management and Prognosis 607

Figure 36-9 Modified template for an en bloc or prospective nerve-sparing RPLND for a
patient with a right-sided tumor.

between 1986 and 1989. Broun et al.34 reported the use platin, etoposide, and thiotepa (CET).37 The patients
of high-dose carboplatin and etoposide with bone mar- had rescue with antologous stem-cell transplantation.
row transplant rescue.34 A more recent report using the There are several additional trials still in progress.
same approach was made from the University of
Michigan by Ayash et al.35 The use of paclitaxel in sal-
TREATMENT BY STAGE
vage regimens along with cisplatin and/or ifosfamide has
been reported by Kollmannsberger et al.36 from This section will present treatment alternatives by clini-
Germany.36 Another new approach for salvage therapy cal stage. The tools of treatment and general follow-up
was reported by Rick et al.37 from Berlin. They used 3 routines have been presented in the previous section.
cycles of TIP followed by 1 cycle of high-dose carbo- When results are available from several series and are
608 Part VI Testis

Figure 36-10 Modified template for an en bloc or prospective nerve-spring RPLND for a
patient with a left-sided tumor.

similar, the percentages after various treatments or peri- approximately 70% of patients observed for clinical stage
ods of observation will be given on the treatment option IA disease will remain disease free. Approximately 30%
algorithms shown in Figures 36-13 to 36-15. will relapse over a 4-year period. Most relapses will occur
within the first year and nearly 90% within 2 years.
Patients on observation who relapse are treated based
Clinical Stage I
on their stage at relapse. Those who relapse with stage
For patients with clinical stage IA, there are three possible IIA disease or stage IIB disease are treated as shown in
choices: observation, primary RPLND, or short course Figure 36-14, while those with stage IIC disease or stage
primary chemotherapy (see Figure 36-13). The criteria for III disease are treated as shown in Figure 36-15.
observation are given in Table 36-7. The suggested follow- Stage IA patients may also be treated by RPLND
up testing for observation is shown in Table 36-8. Overall (modified template-nerve-sparing technique if fertility is
Chapter 36 Nonseminomatous Germ Cell Tests Tumors: Management and Prognosis 609

Figure 36-11 A view of the interaortocaval zone with the patient’s head to the right. Note
the right-sided postganglionic sympathetic nerve branches emerge from under the vena
cava obliquely in a caudal-anterior direction onto the anterior surface of the aorta. The
branches usually are located just cephalad to the lumber (suture around one).

Figure 36-12 A view of the right and left lumbar sympathetic trunks and the postganglionic
branches with the patient’s head to the right. Branches from each side join at the hypogastric
plexus around the origin of the inferior mesenteric artery. Branches from the hypogastric
plexus cross the area of the bifurcation of the aorta and travel caudally to the pelvic plexus.
610 Part VI Testis

Table 36-8 Primary Observation Schema


Year 1 Year 2 Years 3–4 Years = 5

Serum markers Monthly Bimonthly Biannually Annually

Chest x-ray Monthly Bimonthly Biannually Annually

CT scans (chest, abdomen, and pelvis) Quarterly Semiannually Annually —

Physical examination Bimonthly Every 4 months Biannually Annually

Table 36-9 Pathologic Nodal Stage (PN) an issue) or primary short course chemotherapy. RPLND
has been performed more in the United States, while
pNX Regional lymph nodes cannot be assessed
chemotherapy has been used more frequently in Europe.
pN0 No regional lymph node metastasis For clinical stage IB patients, RPLND has been the
more prevalent treatment and chemotherapy has been
pN1 Metastasis with a lymph node mass 2 cm or less in used less frequently in the U.S. In clinical stage IB,
greatest dimension and less than or equal to 5 patients who have T4 disease, a hemiscrotectomy is also
nodes positive, none more than 2 cm in greatest usually performed if it was not already completed as part
dimension of the radical orchiectomy.
pN2 Metastasis with a lymph node mass more than RPLNDs in clinical stages IA and IB patients yield neg-
2 cm but not more than 5 cm in greatest ative lymph nodes in approximately 70% of the patients,
dimension; or more than 5 nodes positive, none while the remaining 30% have positive lymph nodes (pN1
more than 5 cm; or evidence of extranodal or pN2) despite negative markers and imaging studies.
extension of tumor Patients with clinical stage is used to be treated rou-
tinely with RPLND. Davis et al.38 recommended the use
pN3 Metastasis with a lymph node mass more than of primary chemotherapy instead of RPLND because of
5 cm in greatest dimension
the high relapse rate after RPLND. In the last several
Nodal status is determined from RPLND specimens. years, the Indiana group has recommended primary
chemotherapy in these patients.39
Primary chemotherapy for stages IA, IB, or S has low
relapse and death rates (1% to 2% and <1%, respectively)
for an overall progression-free long-term survival rate
Table 36-10 IGCCCG NSGCT Definitions of about 97%.40 Most series involved 2 courses of BEP
Good prognosis Testis/retroperitoneal primary and and several have used 3 courses of BEP. In an attempt to
No nonpulmonary visceral metastases and reduce toxicity, a single course of BEP has been, and is
AFP < 1000 ng/ml and being, tried by several groups. A series by Corti et al.41 in
HCG < 5000 IU/1 and 1997 reported no relapses in 18 stage I NSGCTT
LDH < 1.5 × N* patients with 1 course of cisplatin-based chemotherapy
with a median follow-up of 46.9 months. Studies by the
Intermediate Testis/retroperitoneal primary and
prognosis No nonpulmonary visceral metastases
German Testicular Cancer Study Group are on-going.
and The overall survival rates for patients with stage I
AFP = 1000 and = 10,000 ng/ml and/or NSGCTT is between 97% and 99% for observation
HCG = 5000 and = 50,000 IU/1 and/or series or primary chemotherapy and 98% to 100% in
LDH = 1.5 × N and = 10 × N patients treated with primary RPLND. Long-term mor-
bidity, mortality, and treatment-induced infertility are
Poor prognosis Mediastinal primary very low. Good results are, of course, predicated on
or reliable patients and compulsive medical caregivers.
Nonpulmonary visceral metastases
or
AFP = 10,000 ng/ml and/or Clinical Stages IIA and IIB
HCG = 50,000 IU/1 and/or
LDH = 10 × N Clinical Stages IIA and IIB accounts for approximately
40% of NSGCTT patients at presentation. An algorithm
*N = upper limit of normal. for treatment of these patients is shown in Figure 36-14.
Treatment of clinical stages I NSGCIT (40%)

Stage IA Stage IB Stage IS


pT1 pT 2−4 pT 1−4

OR
OR OR

Primary RPLND
Primary short
with
Observation hemiscrotectomy
course
chemotherapy
for pT4 only

Partial
Complete
No relapse relapse Nodes (−) Nodes (+) remission or
remission
(70%) (30%) (70%) (30%) progression
(97%)
(2-3%)

OR

Adjuvant
PC-RPLND
Observation chemotherapy Observation
(see Figure 36−15
(see Table 36-11) (see Figure 36-14 (see Table 36-11)
PC-RPLND arm
RPLND arm)

Continued
observation IIA,B IIC,III
(see Table 36-8) (see figure 14) (see figure 15)

Figure 36-13 Schematic drawing of the treatment options and stratifications based on
tumor or marker parameters for clinical stage I NSGCTT.

Treatment of clinical stages II A and B (40%)

Chemotherapy OR
RPLND
(2−4 courses)

Partial remission,
Complete
no response, or Nodes (−)
remission Nodes (+)
progression markers (−)
(67%)
(33%)

PC-RPLND
Observation Observation Post-Op Post-Op
(see Figure 36-15
(see Table 36-11) (see Table 36-15) markers (+) markers (−)
PC-RPLND arm)

OR

Adjuvant Adjuvant
chemotherapy Observation chemotherapy
(3-4 courses) (2 courses)

Figure 36-14 Schematic drawing of treatment options and stratifications based on tumor
and marker parameters for patients with stages IIA and IIB NSGCTT.
612 Part VI Testis

Treatment of clinical stages II C and III (20%)

Primary
chemotherapy

CR PR
(70%) (30%)

Observation Markers (+)

Markers (−)

Relapse
Localized disease Diffuse disease
(10%)

Post-chemo, OR
RPLND
Surgical
resection Bone marrow
Salvage chemo
(RPLND +/− transplant (BMT)
thoracotomy)

Scar Teratoma Cancer


(44%) (44%) (12%)

Complete Incomplete
resection resection

Observations
(see Table 12)
OR

Salvage
Observation BMT
chemotherapy

PR CR

BMT Observation

Figure 36-15 Schematic drawing of treatment options and stratifications based on tumor
and marker parameters for patients with stage IIC/stage III NSGCTT.
Chapter 36 Nonseminomatous Germ Cell Tests Tumors: Management and Prognosis 613

There are two main treatment pathways or options: section techniques have evolved over this time period as
primary chemotherapy and primary RPLND. As in clin- presented in the section “Tools of Treatment.” In those
ical stage I, primary chemotherapy is favored more in patients with a desire to remain fertile, the modified
Europe, while primary RPLND is favored in the U.S. template, prospective nerve-sparing technique is the
Primary chemotherapy usually consists of 2 to 4 usual treatment. Sometimes the dissection is extended to
courses of cisplatin-based multiagent treatment. a bilateral template in the presence of medium volume
Horwich et al.42 reported 122 patients with stage IIA or nodal disease (2 to 5 cm).
stage IIB NSGCTT treated with 4 courses of cisplatin- Emission is preserved in nearly all patients in whom
based combination chemotherapy. Overall 97% of the the nerve-sparing technique is employed. Operative
patients were disease free with a median follow-up of 5.5 complication rates run in the range of 8% to 12%, the
years. In stage IIA patients, 17% required a PC-RPLND, reoperation rate is about 1% and the operative and post-
while 39% of stage IIB patients required this surgery. In operative mortality rate is approximately 0.25%.
a randomized trial by the German Testicular Cancer In those patients with pathologically negative nodes
Study Group, 87 patients received primary chemother- (pN0), observation is appropriate with a follow-up sched-
apy with 3 courses of BEP or carboplatin, etoposide and ule as shown in Table 36-11. The overall relapse rate in
bleomycin (CEB). Two-thirds achieved complete remis- pN0 patients is in the range of 7% to 12% with
sion (CR) with chemotherapy alone, while one-third retroperitoneal recurrences within the field of dissection
required PC-RPLND. With a 3-year follow-up, 11% being about 1%.
total in this arm relapsed.43 For patients who have pathologically positive lymph
The patients treated with primary chemotherapy for nodes (pN+), there are two options: observation or adju-
stage IIA disease or stage IIB disease that achieve a CR vant chemotherapy. Relapse in patients with pathologi-
can be followed using the same schedule as primary cally positive nodes is related to nodal tumor volume. In
RPLND follow-up as shown in Table 36-11. Those those patients with pN1 tumors, the relapse rate is in the
patients requiring PC-RPLND are treated as shown on range of 15% to 20%. For patients with pN2 tumors, the
the PC-RPLND arm of Figure 36-15 and follow as relapse rate runs between 30% and 50%, while it is 50%
shown in Table 36-12. to 90% in patients with pN3 tumors.
Primary RPLND has been the predominant treat- Most centers that treat testicular cancer tailor the rec-
ment of stages IIA and IIB NSGCTT in the U.S. for the ommendations for adjuvant chemotherapy based on the
last 4 decades. The templates for dissection and the dis- risk of recurrence, which is related to the pN stage as

Table 36-11 Follow-Up After Primary RPLND


Year 1 Year 2 Years 3–4 Years = 5

Serum markers Monthly Bimonthly Semiannually Annually

Chest x-ray Monthly Bimonthly Semiannually Annually

CT scans (chest, abdomen, and pelvis) None* None* None* None*

Physical examination Bimonthly Every 4 months Semiannually Annually

*CTs are needed for patients with teratoma in the nodes or with cancer in the nodes and normal serum markers using the same schedule as shown
in Table 36-8.

Table 36-12 Follow-Up After Postchemotherapy RPLND Pathology—Necrosis Only


Year 1 Year 2 Years 3–4 Years = 5

Serum markers Bimonthly Every 4 months Biannually Annually

Chest x-ray Bimonthly Every 4 months Biannually Annually

CT scans (chest, abdomen, and pelvis) None None None None

Physical examination Every 4 months Biannually Biannually Annually

Pathology—teratoma or cancer without serum marker elevation—as earlier and add CT scans as in Table 36-8.
614 Part VI Testis

shown earlier. When adjuvant therapy is used, it usually patient has a well-localized disease and all abnormal areas
consists of 2 courses of BEP or EP.26,44 Subsequent are respectable, a surgical approach is taken. This may
relapse rates are around 1%. Overall treatment mortality require procedures in addition to the PC-RPLND, such
rates are less than 1%. as a thoracotomy or cervical lymph node dissection.
If patients had pathologically positive lymph nodes Depending on the extent of disease, the procedures may
pN+ and positive postoperative markers, full course need to be performed in more than one sitting.
chemotherapy is given. The same applies for pN0 In these same circumstances, the patients with positive
patients who relapse after observation. markers after a PR from primary chemotherapy are
The overall cure rate for stages IIA and IIB patients treated with either salvage chemotherapy or high-dose
using the treatment scheme shown in Figure 36-14 is chemotherapy with autologous bone marrow transplan-
97% to 99%. tation. The choice of treatment approach is again usu-
ally determined based on the IGCCCG prognosis
classifications as discussed earlier.
Clinical Stage IIC and/or Stage III
Fortunately, the patients who present with stage
Based on the very high recurrence rate for patients with IIC/stage III account for only 20% of all patients with
stage IIC/stage III NSGCTT after RPLND, these NSGCTT. A high percentage of patients enjoy a durable
patients have been treated with primary chemotherapy CR with primary chemotherapy [70% CR minus 10% of
first since combination cisplatin-based treatment became these with relapse (7%) = 63%]. Approximately 75%
available in the mid-1970s to late 1970s. Figure 36-15 of patients who are treated with PC-RPLND after PR or
shows how these patients are usually treated. relapse after chemotherapy will have a long-term survival
Approximately 70% of patients with stage IIC/stage without or with salvage or high-dose chemotherapy and
III disease will achieve a CR after 3 or 4 courses of BEP autologous bone marrow transplantation. This yields a
or similar combination. Fortunately, 90% of these CR long-term survival or “cure” rate for stage IIC/stage III
patients will enjoy a durable response, while 10% will of approximately 90%, which is a dramatic improvement
relapse. About 30% of the patients with stage IIC/stage before the era of cisplatin-based chemotherapy.
III disease will only achieve a partial response (PR). This
group of patients and the patients who relapse after a
SUMMARY
primary chemotherapy CR are stratified for further treat-
ment by marker status. If the serum markers are negative, The treatment of NSGCTT has changed markedly over
the patients have a PC-RPLND without/with addi- the last 4 decades. The morbidity and mortality from
tional surgical procedures to clear the retroperitoneum treatment have decreased in this same period, while sur-
using the full bilateral template as shown in Figure 36-8 vival rates have increased for all stages of disease. There
and remove any other metastases in the chest or neck. are ongoing efforts to further reduce the amount and
The majority of these patients will only require the severity of treatment used for this disease. The preserva-
PC-RPLND. tion of fertility for these patients through modifications in
Patients who have a PC-RPLND for a PR after pri- surgical techniques and the intensity of chemotherapy has
mary chemotherapy have a 44% incidence of necrosis been a major area of progress. Careful reporting of results,
only, a 44% incidence of teratoma, and a 12% chance of complications, late recurrences, and the development of
active cancer in the tissue removed. Patients those have secondary malignancies will be important in guiding
necrosis only and/or teratoma can be observed as per future modifications of treatment schemes for this disease.
Table 36-12.
If active cancer is found in the tissue removed at
PC-RPLND, patients are stratified by complete or REFERENCES
incomplete surgical resections. For those patients in
whom complete resection was possible, observation 1. Patton JF, Hewitt CB, Mallis N: Diagnosis and treatment
as shown on Table 36-11 is used. If tumor resection of tumors of the testis. JAMA 1959; 171:2194.
was incomplete, patients are treated by salvage 2. Johnson DE: Epidemiology. In Johnson DE (ed):
chemotherapy (often VIP) or high-dose chemotherapy Testicular Tumors, 2nd edition, p 37. Flushing, NY,
Medical Examination Publishing, 1976.
with autologous bone marrow transplantation based
3. Weissbach L, Bussar-Maatz R, Mann K: The value of
on the good-to-moderate or poor prognosis status tumor markers in testicular seminomas. Results of a
as defined by the IGCCCG classification shown in prospective multicenter study. Eur Urol 1997;
Table 36-10. 32(1):16–22.
For patients who were treated with primary chemo- 4. Gomes PJ, Rustin GJ: Attempted preservation of one
therapy and had a PR with persistently positive markers, gonad in patients with bilateral germ cell tumours. Clin
treatment is determined by the imaging studies. If a Oncol (R Coll Radiol) 1996; 8(6):397–399.
Chapter 36 Nonseminomatous Germ Cell Tests Tumors: Management and Prognosis 615

5. Heidenreich A, Weissbach L, Holtl W, et al: Organ non-seminomatous testicular cancer. The Indiana
sparing surgery for malignant germ cell tumor of the University experience. Eur J Cancer 2000; 36(4):440–442.
testis. 2001; 166(6):2161–2165. 23. Amato R, Banks E, Ro J, Swanson DA: Postorchiectomy
6. AJCC. Cancer Staging Handbook, 6th edition. New York, adjuvant chemotherapy for patients with clinical stage I
Springer, 2002. non-seminomatous germ cell tumors of the testis
7. Cremerius U, Wildberger JE, Borchers H, et al: Does (NSGCTT) at high risk for relapse. Proc ASCO 18:157.
positron emission tomography using 18-fluoro-2- 24. Studer UE, Burkhard FC, Sonntag RW: Risk adapted
deoxyglucose improve clinical staging of testicular cancer? management with adjuvant chemotherapy in patients with
Results of a study in 50 patients. Urology 1999; high risk clinical stage I nonseminomatous germ cell
54(5):900–904. tumor. J Urol 163:1785–1787.
8. Leibovitch I, Foster RS, et al: Identification of clinical stage 25. Schefer H, Mattmann S, Borner M, et al: Single course
A nonseminomatous testis cancer patients at extremely low adjuvant bleomycin, etoposide, and cisplatin (BEP) for
risk for metastatic disease: a combined approach using high risk stage I non-seminomatous germ cell tumors
quantitive immunohistochemical, histopathologic, and (NSGCT). Proc ASCO 2000; 19:340a.
radiologic assessment. J Clin Oncol 1998; 16(1):261. 26. Kondagunta GV, Motzer RJ: Adjuvant chemotherapy for
9. Hermans BP, Sweeney CJ, Foster RS, et al: Risk of stage II nonseminomatous germ-cell tumors. Semin Urol
systematic metastases in clinical stage I nonseminoma Oncol 2002; 20(4):239–243.
germ cell testis tumor managed by retroperitoneal lymph 27. Einhorn LH, Donohue J: Cis-diamminedichloroplatinum,
node dissection. J Urol 2000; 163:1721–1724. vinblastine, and bleomycin combination chemotherapy in
10. Hogeboom WR, Hoekstra HJ, Mooyaart EL, et al: disseminated testicular cancer. Ann Intern Med 1977;
Magnetic resonance imaging of retroperitoneal lymph 87(3):293–298.
node metastases of non-seminomatous germ cell tumours 28. Williams SD, Birch R, Einhorn LH, et al: Treatment of
of the testis. Eur J Surg Oncol 1993; 19(5):429–437. disseminated germ-cell tumors with cisplatin, bleomycin,
11. Donohue JP: Surgery for testis cancer: a changing and either vinblastine or etoposide. N Engl J Med 1987;
perspective. Proc Inst Med Chic 1980; 33(2):42. 316(23):1435–1440.
12. Pizzocaro G, Salvioni R, Zononi F: Unilateral 29. The International Germ Cell Cancer Collaborative
lymphadenectomy in intraoperative stage I NSGCT. Group: International germ cell consensus classification: a
J Urol 1985; 134:485–489. prognostic factor-based staging system for metastatic
13. Richie JP: Modified retroperitoneal lymphadenectomy for germ cell cancers. J Clin Oncol 1997; 15:594–603.
patients with clinical stage I testicular cancer. Semin Urol 30. Williams SD, Einhorn LH, Greco FA, et al: VP-16-213
1988; 6:216–222. salvage therapy for refractory germinal neoplasms. Cancer
14. Jewett MA, Kong YS, Goldberg SD, et al: Retroperitoneal 1980; 46(10):2154–2158.
lymphadenectomy for testis tumor with nerve sparing for 31. Loehrer PJ Sr, Gonin R, Nichols CR, et al: Vinblastine
ejaculation. J Urol 1988; 139(6):1220–1224. plus ifosfamide plus cisplatin as initial salvage therapy in
15. Donohue JP, Foster RS, Rowland RG, et al: Nerve-sparing recurrent germ cell tumor. J Clin Oncol 1998;
retroperitoneal lymphadenectomy with preservation of 16:2500–2504.
ejaculation. J Urol 1990; 144(2 Pt 1): 287–291. 32. Motzer RJ, Sheinfeld J, Mazumdar M, et al: Paclitaxel,
16. Donohue JP, Thornhill JA, Foster RS, et al: ifosfamide, and cisplatin second-line therapy for patients
Retroperitoneal lymphadenectomy for clinical stage with relapsed testicular germ cell cancer. J Clin Oncol
A testis cancer (1965 to 1989): modifications of technique 2000; 18:2413–2418.
and impact on ejaculation. J Urol 1993; 149(2):237. 33. Hinton S, Catalano P, Einhorn LH, et al: Phase II study
17. Rassweiler JJ, Frede T, Lenz E, et al: Long-term of paclitaxel plus gemcitabine in refractory germ cell
experience with laparoscopic retroperitoneal lymph node tumors (E9897): a trial of the Eastern Cooperative
dissection in the management of low-stage testis cancer. Oncology Group. J Clin Oncol 2002; 20(7):1859–1863.
Eur Urol 2000; 37(3):251–260. 34. Broun ER, Nichols CR, Kneebone P, et al: Long-term
18. Janetschek G, Peschel R, Hobisch A, Bartsch G: outcome of patients with relapsed and refractory germ cell
Laparoscopic retroperitoneal lymph node dissection. tumors treated with high-dose chemotherapy and
J Endourol 2001; 15(4):449–453. autologous bone marrow rescue. Ann Intern Med 1992;
19. Ogan K, Lotan Y, Koeneman K, et al: Laparoscopic 117(2):124–128.
versus open retroperitoneal lymph node dissection: a cost 35. Ayash LJ, Clarke M, Silver SM, et al: Double dose-
analysis. J Urol 2002; 168(5):1945–1949. intensive chemotherapy with autologous stem cell support
20. Jacobsen KD, Ous S, Waehre H, et al: Ejaculation in for relapsed and refractory testicular cancer: the
testicular cancer patients after post-chemotherapy University of Michigan experience and literature review.
retroperitoneal lymph node dissection. Br J Cancer 1999; Bone Marrow Transplant 2001; 27(9):939–947.
80:249–255. 36. Kollmannsberger C, Mayer F, Kuczyk M, et al: Treatment
21. Palese MA, Su LM, Kavoussi LR: Laparoscopic of patients with metastatic germ cell tumors relapsing
retroperitoneal lymph node dissection after after high-dose chemotherapy. World J Urol 2001;
chemotherapy. Urology 2002; 60(1):130–134. 19(2):120–125.
22. Behnia M, Foster R, Einhorn LH, et al: Adjuvant 37. Rick O, Bokenmeyer C, Beyer J, et al: Salvage treatment
bleomycin, etoposide and cisplatin in pathological stage II with paclitaxel, ifosfamide, and cisplatin plus high-dose
616 Part VI Testis

carboplatin, etoposide, and thiotepa followed by 41. Corti OD, Foneron BA, Troncoso SL: Treatment of stage
autologous stem-cell rescue in patients with relapsed I nonseminomatous testicular cancer with one cycle of
or refractory germ cell cancer. J Clin Oncol 2001; adjuvant chemotherapy. Actas Urol Esp 1997; 21:961–963.
19:81–88. 42. Horwich A, Norman A, Fischer C, et al: Primary
38. Davis BE, Heu HW, Fair WR, Bosl GJ: The management chemotherapy for stage II nonseminomatous germ cell
of patients with nonseminomatous germ cell tumors of tumors of the testis. J Urol 1994; 151(1):72–77.
the testis with serologic disease only after orchiectomy. 43. Weissbach L, Bussar-Maatz R, Flechtner H, et al:
J Urol 1994; 152:111–113. RPLND or primary chemotherapy in clinical stage IIA/B
39. Saxman SB, Nichols CR, Foster RS, et al: The nonseminomatous germ cell tumors? Results of a
management of patients with clinical stage I prospective multicenter trial including quality of life
nonseminomatous testicular tumors and persistently assessment. Eur Urol 2000; 37(5):582–594.
elevated serologic markers. J Urol 1996; 155(2):593–594. 44. Williams SD, Stablein DM, Einhorn LH, et al:
40. Albers P, Perabo FGE, Melchior D, Siener R: Adjuvant Immediate adjuvant chemotherapy versus observation
chemotherapy in stage I and stage II testicular cancer. with treatment at relapse in pathological stage II testicular
World J Urol 2001; 19:76–81. cancer. N Engl J Med 1987; 317:1433–1438.
C H A P T E R

37Nongerm Cell Tumors of the Testis


Aaron J. Milbank, MD, Howard S. Levin, MD,
and Eric A. Klein, MD

Nongerm cell tumors of the testis comprise a heteroge- eral or bilateral testicular enlargement, rather than a dis-
neous group of benign and malignant neoplasms, which crete mass on physical examination or scrotal ultra-
may be primary or metastatic. As a group they account sonography, suggests a secondary rather than primary
for only 5% to 6% of all testicular neoplasms, the neoplasm. Peripheral lymphadenopathy is rare with pri-
remaining testicular tumors being of germ cell origin mary nongerm cell tumors and should prompt a search to
(Figure 37-1). The most commonly occurring primary exclude a primary tumor from another site, notably lym-
tumors of nongerm cell origin are classified as sex phoma. Gynecomastia, loss of libido or other signs of
cord/stromal tumors or tumors of specialized gonadal feminization may be present with some tumors (notably
stroma. These tumors may be associated with somatic or Leydig or Sertoli cell tumors) due to active secretion
constitutional chromosomal syndromes. Other less com- of estrogens, alterations in serum estrogen/testosterone
monly encountered primary tumors include mesenchy- ratios or peripheral aromatization of testosterone.3
mal tumors from nonspecialized stroma, such as blood Serum levels of human chorionic gonadotropin hCG and
vessels or smooth muscle, tumors arising from hematopoi- alpha fetoprotein (AFP) will be normal. All of these clinical
etic cells, and a multitude of other benign tumors, which features may also be present with primary germ cell
mimic malignant lesions. In addition, a number of malig- tumors, and histologic examination of the affected testicle
nancies from other sites may rarely present initially in the is the sine qua non for distinguishing these tumor types and
testis or as a late sign of more diffuse disease. Certain determining the need for further evaluation and treatment.
populations are at higher risk for the development of In adults with a palpably normal opposite testis,
germ cell and nongerm cell testis tumors; multiple cases inguinal orchiectomy without frozen section is consid-
of malignant lymphoma (ML) and malignant germ cell ered standard therapy. Occasionally a nongerm cell
tumors in patients with the acquired immune deficiency tumor or other benign process may be strongly suspected
syndrome and other forms of immunosuppression have based on presenting signs and symptoms and/or the
been reported.1,2 A useful practical classification of ultrasonographic appearance of the lesion. In these cases
nongerm cell testicular tumors is presented in Table 37-1. and in patients with a solitary testis or atrophic con-
Most primary tumors of nongerm cell origin are bio- tralateral testis, inguinal exploration and excisional
logically benign. The clinical presentation of these biopsy with the intent of sparing the uninvolved ipsilat-
tumors is similar to, and often indistinguishable from, eral testis should be considered. In such instances, frozen
those of germ cell origin. Common presenting symptoms section examination (FSE) of the excised tumor is neces-
include testicular pain and enlargement, which may be of sary to exclude the presence of malignancy. Elert et al.4
prolonged duration. Breast tenderness or enlargement recently retrospectively reviewed their experience with
may also be present. Physical examination typically con- 354 patients who underwent inguinal exploration, isola-
firms the presence of testicular swelling or intratesticu- tion of the testicle, and biopsy with FSE. With respect to
lar mass. Most primary nongerm cell tumors appear malignant versus benign, FSE correctly categorized all
as discrete hypoechoic lesions on ultrasonography, while lesions. Within the malignant category, 9% of the tumors
leukemic or lymphomatous infiltration demonstrate were incorrectly identified (nonseminoma versus semi-
enlargement with multiple focal or diffuse areas of noma or vice versa), a finding which did not affect the
decreased echogenicity. The presence of diffuse unilat- intraoperative management. In all such cases, the

617
618 Part VI Testis

Figure 37-1 Embryologic origin of testis tumors.

Table 37-1 Classification of Nongerm Cell Testicular Tumors


Tumors of Specialized Tumors of Generalized Hematologic
Stroma Stroma Tumors Miscellaneous Tumors

LCT Vascular tumors Lymphoma Carcinoid

Sertoli cell tumor Smooth and skeletal muscle Leukemia Epidermoid cyst
tumors

Granulosa cell tumor Plasmacytoma Adenocarcinoma of the rete testis

Mixed tumor Adenomatoid tumor Metastatic tumors

Incompletely differentiated Mesothelioma Ovarian surface epithelial—type


tumors tumors

PRIMARY TUMORS OF SEX CORD/STROMAL


clinical aspects and topography of the mass should be
ORIGIN
shared with the pathologist. In cases with equivocal find-
Leydig Cell Tumors
ings on frozen section, inguinal orchiectomy should be
Clinical Presentation
performed in patients with a normal contralateral testis.
Frozen section is also useful in identifying MLs so that Leydig cell tumors (LCTs) comprise approximately 1%
fresh tissue can be used for tumor phenotyping. A descrip- to 3% of testicular tumors. They occur in males of all
tion of individual tumor types with characteristic clini- ages with predominance in children older than 4 and
cal and pathologic features and recommendations for adults in the third to sixth decade. In prepubertal boys
management follows. they produce isosexual precocity with penile enlarge-
Chapter 37 Nongerm Cell Tumors of the Testis 619

ment, sexual hair growth, increased bone growth, deep- 10 cm in diameter (Figure 37-2). They may be bilateral.14
ened voice, and aggressive behavior as a consequence of The microscopic appearance is variable with a spectrum
increased testosterone production by the neoplasm. of cell shapes extending from polygonal to spindled.
Clinical signs and symptoms are usually manifest before Leydig cell nuclei are generally round with a small single
the discovery of the neoplasm, which may be impalpable nucleolus but binucleate, multinucleate, and giant cells
and difficult to detect with ultrasonography.5 In postpu- with giant nuclei may be present (Figure 37-3). Mitotic
bertal males, LCTs usually present with a unilateral pain- figures are generally sparse. Cell cytoplasm is usually
less testicular mass or incidentally as a mass discovered eosinophilic and sometimes ground glass in appearance
during physical examination. Only 30% have signs of but may be vacuolated depending on the amount of intra-
increased hormonal function, and in contrast to the pre- cytoplasmic lipid. Approximately 40% of LCTs contain
pubertal group affected adults exhibit feminization, with intracytoplasmic Reinke’s crystals. Lipochrome pig-
unilateral or bilateral gynecomastia, impotence, and hair ment may also be present in varying amounts. The tumor
loss.3 LCTs have been reported in cryptorchid testes cells may grow in sheets, cords, and tubular formations.
before and after orchidopexy, in the contralateral testis Capillaries are abundant between aggregates of epithelial
of a 40-year old, 7 years after orchiectomy for a mixed cells. The stroma may be loose or dense and hyaline. The
malignant germ cell tumor, in association with Klinefelter’s
syndrome and tuberous sclerosis, in the descended testicle
of a man with contralateral cryptorchidism, and in multi-
ple asymptomatic men presenting for evaluation of infer-
tility.6–11 There is some evidence from animal studies that
lack of regulation of steroid production/conversion may
be etiologically associated with LCTs. Overexpression of
aromatase has been shown to induce LCTs in transgenic
mice.12 Activating mutations of the gene encoding the
luteinizing hormone receptor have been described.13

Pathologic Characteristics
The gross appearance of LCTs is characteristically
mahogany or yellow-brown in color, rubbery, discrete, Figure 37-2 Benign LCT. Discrete nonencapsulated soft tan
nonencapsulated, and nonnecrotic. LCTs vary consider- 2.2 cm mass in bivalved testis in 29-year-old man with left
ably in size ranging from nonpalpable to more than testicular mass and bilateral gynecomastia.

Figure 37-3 LCT. Twenty-year-old with enlarged testis. The testicular parenchyma was
substantially replaced by a yellow-brown mass. The Leydig cells contain round uniform
nuclei and eosinophilic granular cytoplasm (H&E × 31.2).
620 Part VI Testis

ultrastructural appearance resembles that of other steroid


hormone-producing cells. LCTs stain positively for lipid.
In tumors of prepubertal males seminiferous tubules
adjacent to the neoplasm have shown response to local
androgen manifested as proliferative activity of germinal
epithelium with development to the spermatid stage.15
Seminiferous tubules in the contralateral testis and dis-
tant from the tumor in the ipsilateral testis typically
retain prepubertal morphology. Conversely, in postpu-
bertal men seminiferous tubules adjacent to feminizing
LCTs show reduced spermatogenesis and thickening of
the tunica propria.
Approximately 10% of LCTs are malignant and over
30 such cases have been reported. Malignant LCTs are
Figure 37-4 Malignant LCT. Sixty-three-year-old man noted
larger than benign LCTS have no clinical endocrine
enlargement and soreness of the testis for 4 to 5 weeks. The
hyperfunction and have a shorter clinical duration prior testis measured 6 × 7 × 7 cm and was almost totally
to orchiectomy.16 The absolute criterion for malignancy replaced by a bulging light tan to dark brown variegated
of an LCT is metastasis, but other morphologic features mass with multiple apparent septae. (Photograph courtesy of
that strongly suggest malignancy include large tumors, California Tumor Tissue Registry.)
marked cellular anaplasia, frequent or atypical mitoses,
infiltrating margin and necrosis16 (Figure 37-4). Invasion
of lymphatics and/or blood vessels are also suggestive of tumors not associated with metastasis. Metastases usually
malignancy (Figure 37-5). Cheville et al.17 retro- occur within months to years after the original diagnosis
spectively reviewed their experience with 30 LCTs, 23 but initial metastasis has been recorded as late as 17 years
without metastasis, and 7 with metastasis to identify after orchiectomy.18 In this latter case, the retroperi-
predictors of malignancy. They found that the follow- toneal metastasis exhibited sarcomatoid differentiation.
ing features were significantly associated with metastatic Initial metastasis is usually to retroperitoneal lymph nodes,
behavior: presence of cytologic atypia, necrosis, angi- but subsequent metastases involve remote lymph
olymphatic invasion, increased mitotic activity, atypical nodes and possibly viscera. Benign LCTs generally have
mitotic figures, infiltrative margins, extension beyond the diploid DNA content whereas malignant LCTs generally
testicular parenchyma, DNA aneuploidy, and increased have aneuploid DNA by flow cytometry.19,20
MIB-1 activity. Tumors associated with metastasis tended Ulbright et al.21 recently reported 19 cases of LCTs
to be larger (mean 4.7 cm versus 2.6 cm) and occur in with unusual features—adipose metaplasia, calcification
older patients (mean age 62 years versus 48 years) than (sometimes with ossification), and spindle cell growth.

Figure 37-5 Malignant LCT, same case as in Figure 37-4. Neoplastic Leydig cells are
present within a vein. The tumor thrombus shows central necrosis (H&E × 15.6).
Chapter 37 Nongerm Cell Tumors of the Testis 621

Follow-up was reported for 6 of 8 cases exhibiting spin-


dle cell growth. Two patients developed metastases and
both had malignant features apart from spindle cells.
The other 4 did not have malignant features and were
free of disease from 2.3 to 6 years after orchiectomy.
Recognizing that adipose tissue may be present in LCTs
may prevent the misdiagnosis of extratesticular growth or
LCT associated with congenital adrenal hyperplasia (see
later). Similarly, the recognition that spindle cells may be
identified with LCTs may limit misclassification of such
tumors as sarcomas.

Tumors in Males with Congenital Adrenal Figure 37-6 Probable congenital adrenal hyperplasia.
Hyperplasia Twenty-nine-year-old man had right testicular enlargement for
2 years. Left testis was removed 4 years earlier and
An important distinction must be made between LCTs demonstrated a similar mass. Bivalved testis demonstrates a
and the testicular masses in the salt-wasting form of con- dark mahogany colored bilobed 6.8 cm mass replacing
genital adrenal hyperplasia (CAH). In CAH males from testicular parenchyma.
the neonatal period to adulthood may develop bilateral
testicular or peritesticular masses, which are histologi- In order to avoid inappropriate orchiectomy in patients
cally similar to LCTs (Figures 37-6 and 37-7).22,23 One with CAH, and because of their response to corticos-
recent study identified testicular tumors ranging in size teroid therapy and the risk of adrenal insufficiency if
from 0.2 to 4 cm in 16 of 17 adolescents and adults with these patients are not treated with corticosteroids after
CAH (both salt-wasting and not salt-wasting).24 The orchiectomy, it is important to distinguish true LCT
cells of these masses do not contain Reinke’s crystals but from CAH with testicular masses.
do contain abundant lipochrome pigment that gives the
gross tumors a brown or black appearance. These masses Evaluation and Management
resemble those in patients with Nelson’s syndrome. The
fact that the masses often disappear after corticosteroid In children presenting with precocious puberty, bio-
therapy and are sensitive to ACTH stimulation suggests chemical evaluation is necessary to distinguish primary
they are of adrenocortical origin. Davis et al.25 described LCT from Leydig cell hyperplasia secondary to CAH.
one instance of a malignant LCT in a patient with CAH. The presence of a unilateral tumor with normal serum or

Figure 37-7 Probable congenital adrenal hyperplasia mimicking LCT. Same case as in
Figure 37-6. Uniform cells contain similar-appearing round nuclei and granular cytoplasm
containing fine brown pigment (H&E × 128).
622 Part VI Testis

urinary levels of DHEA, androstenedione, and 17- regimens), and only isolated reports of benefit from
hydroxyprogesterone strongly favors the diagnosis of pri- resection of low-volume or solitary metastatic deposits.
mary LCT, while bilateral testicular and/or peritesticular Complete responses to mitotane have been reported but
masses or those associated with abnormalities of steroid the patients ultimately relapsed and died of progressive
biosynthesis favor CAH. In either instance serum testos- disease.28
terone levels may be elevated. ACTH stimulation and
dexamethasone suppression tests are occasionally neces-
Sertoli Cell Tumor
sary to establish tumor origin. Gabrilove et al.3 reviewed
Clinical Presentation
37 cases of feminizing LCTs of the testis, 5 of which were
in prepubertal boys. In 20 patients where estrogens were A variety of Sertoli cell tumors (SCTs) have been
measured, 13 had high urinary titers. Serum estrogen described, which together comprise only 1% of testicular
levels may also be elevated with a concomitant reduction tumors. The principal cell types are classic Sertoli cell
in serum testosterone. Gonadotropin levels will vary with tumor (CSCT), sclerosing Sertoli cell tumor (SSCT),
associated levels of serum androgens and estrogens. large cell calcifying Sertoli cell tumor (LCCSCT), and
The treatment of LCT is surgical excision. In cases Sertoli cell adenoma (SCA) associated with androgen
where the diagnosis of LCT is not suspected preopera- insensitivity syndrome (AIS). All but the last of these
tively, inguinal orchiectomy is the procedure of choice. variants typically present with painless testicular enlarge-
In children and adults, in whom the diagnosis is sus- ment and occur in males of any age. SCA in AIS occurs
pected, a testis-sparing enucleation or excisional biopsy is in phenotypic females. Gynecomastia is present in about
acceptable provided that histologic confirmation of the 20% of cases of SCA and is a typical feature of malignant
lesion is obtained by frozen section. Following tumor SCTs. SCTs are to be distinguished from microscopic or
excision the endocrinologic manifestations of the tumor barely grossly visible masses of persistent immature
typically disappear in adults. In children with symptoms tubules that have been termed Pick’s adenomas or
of short duration the signs of masculinization generally androblastomas, which are often seen in cryptorchid
abate and puberty will proceed normally. However, in testes and have no malignant potential.
some children with long-standing symptoms the signs of
precocious puberty will persist. Pathologic Characteristics
The management of Leydig cell hyperplasia associ-
ated with CAH begins with the hormonal evaluation Classic sertoli cell tumor. On gross inspection CSCTs
described earlier. Some authors have advocated needle are generally circumscribed and yellow, tan or white, and
biopsies of such lesions to ascertain the presence or may be hemorrhagic (Figure 37-8). Microscopically, they
absence of Reinke’s crystals.23,26 If the crystals are not grow in tubules, cords, sheets, aggregates and occasional
identified, treatment with corticosteroid may be initiated retiform configurations separated by fibrovascular septae
and orchiectomy may be deferred. These lesions often (Figure 37-9). Nuclei are round to ovoid and may contain
diminish in size. Unfortunately, the probability of nucleoli. Tumor cell cytoplasm may be pale or
demonstrating Reinke’s crystals on needle biopsy is low. eosinophilic and may contain lipid vacuoles. The latter
Virtually all LCTs in children and 90% of LCTs in finding in a tumor with a diffuse growth pattern may be
adults are benign, have a good prognosis, and require no confused with the appearance of an LCT. The stroma of
therapy other than excision. Patients with malignant
tumors have a poor prognosis and usually die within
2 years after the discovery of metastatic disease. Patients
in whom a malignant tumor is suspected on histopathol-
ogy should undergo a metastatic evaluation, including
chest x-ray, abdominal and pelvic CT scan, and bio-
chemical evaluation in the hopes of identifying a usable
tumor marker. The optimum management of patients
with malignant LCT following excision of the primary
lesion is unknown. There are no available data to evalu-
ate the benefit of prophylactic retroperitoneal lym-
phadenectomy versus observation in patients with disease
clinically localized to the testis. In a review of the man-
agement of 32 patients with established metastatic LCT,
Bertram et al.27 reported no objective responses to radia- Figure 37-8 Sertoli cell tumor. The tumor is a discrete 8 mm,
tion therapy, minimal or no benefit to various com- nonencapsulated, nonnecrotic, firm mass in a 44-year-old
binations of chemotherapy (including platinum-based man.
Chapter 37 Nongerm Cell Tumors of the Testis 623

Figure 37-9 Sertoli cell tumor. The tumor contains solid uniform tubular structures with
round to oval nuclei and prominent nucleoli. The tubules are surrounded by basement
membrane. Elsewhere, similar-appearing cells lack lumens and grow in more solid
configurations (H&E × 62.2).

a CSCT may be scant or abundant and hyalinized. Most aggregates, and cords of epithelial cells. The cells were
CSCTs behave in a benign fashion, but up to 30% of medium sized with rounded vesicular to dark nuclei. The
CSCTs may be malignant as evidenced by the presence of epithelial elements proliferated in a dense hyaline stroma
local invasion and metastases. within which were entrapped nonneoplastic seminiferous
Young et al.29 reviewed 60 cases of CSCT. Age at pres- tubules lined by immature Sertoli cells. No patient showed
entation ranged from 15 to 80 years (mean 45 years) and evidence of malignant behavior, including the 80-year old
the size of the tumors ranged from 0.3 to 15 cm (mean man with histologic evidence of invasion. Several cases
3.6 cm). Long-term follow-up (>5 years) was available for have been reported subsequent to the report by Zukerberg
16 patients; 9 patients were alive with no evidence of dis- et al.31 bringing the total number of cases reported to 12.
ease, 4 were alive with recurrent disease and 3 were dead
with the cause of death attributed to recurrent SCT. The Large cell calcifying sertoli cell tumor. Proppe and
following pathologic features were correlated with a Scully,32 in 1980 characterized unusual and distinctive
malignant course: tumor diameter greater than 5 cm, tumors of the testis in 10 patients as LCCSCTs. Since
necrosis, moderate to severe nuclear atypia, vascular then, at least 35 additional LCCSCTs have been
invasion, and >5 mitoses per 10 high power fields. Only reported.33–35 Age at presentation ranges from 5 to 48
1 of 9 benign cases exhibited more than 1 of these fea- years with the majority less than age 20. As of 1995, only
tures; 5 of 7 of the malignant cases exhibited 3 or more. 2 reported cases were deemed malignant.36 There have
been subsequent reports of malignant cases.37 The tumor
Sclerosing sertoli cell tumor. SSCT is a recently has been associated with a variety of somatic syndromes,
described entity. Zukerberg et al.30 described 10 patients including Carney’s syndrome (cardiac myxomas, spotty
aged 18 to 80 years (median 30) with SSCT. The tumors skin pigmentation, endocrine abnormalities, and schwan-
generally presented as a painless mass, with one arising in nomas), Peutz-Jeghers syndrome (gastrointestinal poly-
a cryptorchid testis and another in a testis that had posis and mucocutaneous pigmentation), adrenal cortical
undergone orchiopexy. All tumors were unilateral and hyperplasia, primary pigmented adrenal cortical disease,
ranged from 0.4 to 4.0 cm in diameter. Eight of 10 LCTs of the testis, acromegaly, pituitary gigantism,
tumors were l.5 cm or less. The tumors were well demar- Cushing’s syndrome, acidophilic adenoma of the pitu-
cated, hard, and yellow-white to tan. Microscopically 9 of itary gland, gynecomastia, isosexual precocity, and
10 tumors were discrete. The 10th tumor invaded the rete AIS.33,34,38 A recent review of Carney’s syndrome identi-
testis, epididymis, and blood vessels. The tumors con- fied 26 reported cases of testicular tumors in patients
tained simple and anastomosing tubules, large cellular with Carney’s syndrome, most of proven LCCSCT
624 Part VI Testis

pathology.39 The neoplasms may involve the entire testis


but in general are 4 cm or less in maximum dimension.
They are frequently multifocal, bilateral in approxi-
mately 40%, and are well circumscribed. On cross-sec-
tion the masses are yellow-tan sometimes with calcific
foci. Microscopically, the tumor grows both within sem-
iniferous tubules and within the interstitium. Within
tubules the neoplastic cells are large with abundant pink
cytoplasm and expand the tubules. Some tubules have a
markedly thickened tunica propria, which extends into
tubular lumens as eosinophilic spherules. The interstitial
tumor is composed of cords, nests, and trabeculae. The
tumor cells range from 12 to 25 μm in diameter and have
ground glass or finely granular cytoplasm. Transitions
between intratubular and interstitial growth may be seen. Figure 37-10 Sertoli cell adenoma of right testis in patient
Calcific rounded nodules, plaques, and masses may be with AIS. The patient was a 17-year-old virilized phenotypic
female. In the right orchiectomy specimen is a firm 1.6-cm
present as intratubular and extratubular aggregates.
nonencapsulated tan nodule. The white firm areas at the
Of the first 35 reported cases of LCCSCT, metastasis bottom of the photographs of the testes are smooth muscle
was observed in 2 cases.35 However, in 1997, Kratzer masses at the medial portion of the testes and are of
et al.35 reported 6 malignant and 6 benign LCCSCTs. probable müllerian duct origin.
Malignant tumors were more likely to be unilateral (all 8
cases to date), solitary, occurred in older patients (mean
age 39 versus 17 for benign cases), and occurred in the patients with complete AIS will develop malignant germ
absence of a genetic syndrome. Pathologic features that cell tumors by age 50.40
were more frequently identified in the malignant cases
included the following: size >4 cm, extratesticular growth, Evaluation and Management
necrosis, high-grade cytologic atypia, vascular space inva-
sion, and more than 3 mitoses per 10 high power fields. Most SCTs are not suspected preoperatively and are
All malignant cases exhibited at least 2 of these features; diagnosed only at the time of orchiectomy performed for
all benign cases exhibited none of these features. a testicular mass. In the context of a known associated
syndrome, the presence of the LCCSCT variant may be
Sertoli cell adenoma associated with androgen insen- suspected preoperatively by the presence of testicular
sitivity syndrome. AIS is caused by defective or absent calcifications on scrotal ultrasound, but this finding is not
cellular androgen receptors. As a result, patients with specific to LCCSCT. When suspected, a testis-sparing
complete AIS are phenotypic females with a shallow surgical approach as described for LCT may be consid-
vagina and absent Wolffian duct derivatives. The patients ered remembering that rare LCCSCTs may be malignant
have 46 XY karyotypes, and bilateral intraabdominal and that some may be multifocal. Histologically, benign
testes that frequently contain nodular masses comprised tumors require no further therapy, although long-term
of multiple tubules lined almost exclusively by Sertoli follow-up may be indicated, as the occurrence of
cells (Figure 37-10). Hamartomas contain small tubules metastatic disease has been reported as long as 15 years
lined by immature Sertoli cells and may have hyperplas- after initial diagnosis. For metastatic lesions, involvement
tic Leydig cells and ovarian type stroma. Young and of the retroperitoneal nodes is frequent and there are sev-
Scully40 and Rutgers and Scully41 distinguish between eral case reports of long-term complete remission fol-
hamartomas and pure SCA, which is comprised exclu- lowing retroperitoneal lymphadenectomy. Experience
sively of tubules lined by Sertoli cells (Figure 37-11). with chemotherapy for malignant SCTs is limited and
Although the SCA may be a monomorphic manifestation the response rate is undefined. There are reported cases
of a hamartoma, some SCAs achieve sizes up to 25 cm in of complete responses to multiagent chemotherapy.43
diameter. SCAs and hamartomas are completely benign.
Rarely, other types of sex cord-stromal tumors have been Granulosa Cell Tumors
reported in AIS, including a few malignant tumors.42 Clinical Presentation
While the SCAs and hamartomas are benign, it is well to
remember that these masses are occurring in cryptorchid Two forms of granulosa cell tumors have been described
testes, and malignant germ cell tumors, principally in males, adult and juvenile. Nineteen cases of adult
malignant intratubular germ cell neoplasia and semino- granulosa cell tumor (AGCT) of the testis have been
mas may develop concomitantly. Approximately 30% of reported in the literature, although additional sex cord-
Chapter 37 Nongerm Cell Tumors of the Testis 625

Figure 37-11 Sertoli cell adenoma. Same patient as in Figure 37-10. The SCA is on the right
and is comprised of discrete uniform tubules lined by Sertoli cells. The larger tubules on the left
are of the nonadenomatous testis and are separated by prominent Leydig cells (H&E × 31.2).

stromal tumors with granulosa cell differentiation have


been reported.44–46 The majority have presented as
scrotal masses with testicular enlargement ranging from
2 months to 15 years in the described cases. The patient
with a mass of 15 years duration had delayed testicular
descent and 2 other patients had been cryptorchid. Five
tumors have had associated estrogenic clinical manifes-
tations (gynecomastia). Four of these patients had
lymph node or visceral metastases and 2 patients died of
their disease. One patient developed metastases 121
months after diagnosis and died of disease 13 months
later.
Juvenile granulosa cell tumors (JGCT) rarely arise in
infants over 6 months old and in all probability arise in
utero. A total of 48 cases have been reported.47 Fifty per-
cent of cases were diagnosed in newborns and 90% by 6
months of age (mean 1 month, range 0 to 11 months). Figure 37-12 Juvenile granulosa cell tumor. One-month-old
They have been associated with 45XO/46XY and boy with left testicular mass. The testis measured 3 × 2.5 ×
45XO/46XY iso(Yq) karyotypes and numerous other 1.5 cm. The surface was smooth and red-tan. On cut
forms of mosaicism.48 The tumors are generally found in sections, there was a variegated red to yellow lobulated
descended testes although they have been discovered parenchyma with cystic areas up to 5 mm in diameter.
in undescended and torsed testes. The tumor is not (Photograph and case courtesy of California Tumor Tissue
associated with isosexual precocity. The testis harbor- Registry.)
ing this tumor is usually enlarged, solid, and/or cystic
(Figure 37-12). and either solid or cystic. Two patients with metastases
had hemorrhagic or friable and necrotic foci in their
tumors. Their microscopic appearance has resembled
Pathologic Characteristics that of ovarian granulosa cell tumor with solid, cystic,
The reported AGCTs ranged in size from 4 microscopic insular, gyriform, and trabecular patterns. Individual
lobules to 13 cm and were well circumscribed.44 They granulosa cells have been described as fairly uniform with
have been described as brownish, white, yellow, and pink scanty cytoplasm, indistinct cell borders, and longitudi-
626 Part VI Testis

nal nuclear grooves in elongate cells. Jimenez-Quintero AFP are normally elevated in the neonate relative to adult
et al.44 recorded up to 26 mitotic figures in 50 high power values, so serum AFP levels are of no value in the diagnosis
microscopic fields. Tumor cells have been described as of this tumor.
positive for vimentin and negative for keratin or epithe-
lial membrane antigen.
Evaluation and Management
JGCT are grossly multicystic and contain follicular
structures of varying size lined by one to multiple layers The diagnosis of AGCT is not usually established until
of cells and contain pale eosinophilic or basophilic intra- the time of orchiectomy and surgical excision may be cur-
luminal material (Figures 37-12 and 37-13). The stroma ative. There is insufficient experience with malignant
is fibrous and may contain groups of cells that resemble forms of this tumor to comment on therapy. JGCT can be
theca cells and granulosa cells, which do not resemble the suspected on the basis of patient age and karyotype. They
cells of AGCT. Lawrence et al.48 found only a few are invariably benign and are cured by excision. Although
grooved cells in a minority of tumors. In the JGCTs the follow-up in many cases is still limited, none of the 48
granulosa cells have round to oval nuclei and pale to reported cases have been associated with recurrence.47
eosinophilic cytoplasm. Tumor granulosa cells stain pos-
itively for vimentin and some cells stain positively for ker-
MIXED SEX CORD/GONADAL STROMAL TUMORS
atin and S100 protein with immunoperoxidase stains.49
AND INCOMPLETELY DIFFERENTIATED TUMORS
The cells that resemble theca cells have been demonstrated
to stain positively for muscle-specific actin, vimentin, and Tumors of mixed histology occasionally occur and may
focally for desmin with immunoperoxidase stains. Mitoses consist of some areas with recognizable elements mixed
may be numerous. Tumor cells may be present in relation- with incompletely differentiated cells resembling ovarian
ship to seminiferous tubules and have even been described stroma (Figure 37-14). A recently described transgenic
within a seminiferous tubule. The differential diagnosis of mouse model suggests that at least some of these neo-
JGCT of the testis consists of those tumors developing in plasms may arise from tumors, that begin as SCTs.50 Like
the neonatal period. Probably most congenital testicular pure forms, tumors of mixed histology and incompletely
tumors and those discovered in the first 4 months of life are differentiated tumors present as an isolated testicular mass
JGCTs rather than yolk sac tumors. Yolk sac tumors may be usually without endocrinologic signs or symptoms and
solid and/or cystic and may have macrocystic or microcys- are diagnosed at the time of orchiectomy (Figure 37-15).
tic areas. However, definitive areas diagnostic for yolk sac Most are biologically benign but some metastatic tumors
tumor, including Schiller-Duval bodies and immunoperox- have been described.51,52 Treatment is by inguinal
idase staining for AFP, are absent in JGCT. Levels of serum orchiectomy. Retroperitoneal lymph node dissection may

Figure 37-13 Juvenile granulosa cell tumor. Same patient as in Figure 37-2. Multiple small
cysts and areas of solid polyhedral to rounded cells comprise the tumor. Elsewhere, larger
follicles were present (H&E × 31.2).
Chapter 37 Nongerm Cell Tumors of the Testis 627

Figure 37-14 Incompletely differentiated gonadal stromal tumor. Testicular mass in 46-year-
old. Interlacing fascicles of basophilic spindled cells with large uniform nuclei resemble cells
of ovarian stroma (H&E × 62.2).

Figure 37-16 Epidermoid cyst. Eighteen-year-old with 3-


month history of nontender mass of left testis. The testis
contains a well-circumscribed 2-cm cyst with a 1-mm thick
Figure 37-15 Incompletely differentiated gonadal stromal
gray wall filled with yellow-white friable cheesy material.
tumor; 2.2-cm tan nonencapsulated, subcapsular tan firm
mass. Same patient as in Figure 37-14.

have value in some cases with metastasis.53 Good slightly more commonly on the right side and rarely are
responses to platinum-based chemotherapy have been much larger than 2 to 3 cm. Grossly they appear as a usu-
described for metastatic disease.54 ally solitary cyst containing laminated keratinous mate-
rial (Figure 37-16). Histologically they are composed of
a squamous lining and filled with keratin.55 Treatment
Miscellaneous Tumors has historically been by orchiectomy, but there are sev-
Epidermoid Cysts eral reports in the literature describing a characteristic
Epidermoid cysts are benign lesions, which typically appearance of this lesion on ultrasound, which may allow
present as a painless testicular mass and are frequently diagnosis preoperatively.56 The ultrasonographic
noted incidentally on physical examination. They occur appearance of a markedly heterogeneous intratesticular
628 Part VI Testis

mass with or without alternating hypoechoic and hyper- Pathologic characteristics. RTCs are poorly circum-
echoic layers surrounded by a hypoechoic or echogenic scribed gray nodules at the hilus of the testis. Most
rim and absence of blood flow on color Doppler sonog- tumors are single, but multiple masses have been
raphy suggest the diagnosis of a testicular epidermoid reported. Reported tumors have ranged in size from 1 to
cyst. Although these criteria are not diagnostic, they may 15 cm. Histologically, RTCs are adenocarcinomas. The
strengthen the indication for excisional biopsy. In such predominant growth pattern is papillary with solid, spin-
cases, inguinal exploration and excisional biopsy seem dled, and cystic areas less common (Figures 37-18 and
reasonable, especially in children or patients with a soli- 37-19). Tumor cells are columnar to cuboidal with aci-
tary testicle or bilateral lesions.57,58 Epidermoid cysts dophilic to amphophilic cytoplasm. Nuclei are enlarged,
have rarely been reported in a patient with Klinefelter’s pleomorphic and round to oval with coarsely granular
syndrome, and we have seen another such patient (Figure chromatin and sometimes prominent nucleoli. Mitoses
37-17).59 may be frequent. The tumors stain negative for mucin,
occasionally positive for CEA, and positive for vimentin,
epithelial membrane antigen, and keratin with
Rete Testis Carcinoma
immunoperoxidase stains. Immunoperoxidase stains are
Clinical presentation. Orozco and Murphy60 reported negative for AFP, hCG, and PSA. The diagnosis of RTC
one case and reviewed 43 cases of rete testis carcinoma should be made in cases where the tumor is present in the
(RTC) that have been recorded in the literature through hilus of the testis, where there is a transition from histo-
1992. The patients have ranged in age from 17 to 91 years logically normal rete testis to RTC, where primary tes-
of age with a mean of 50 years. The clinical onset may or ticular tumors of germ cell and nongerm cell origin and
may not be associated with pain. Testicular enlargement mesothelioma can be excluded, and where extratesticular
averaged about 2 years in duration but had been present origin can be reasonably excluded on a histologic and
for 5 years in four instances. Serum tumor markers were clinical basis. The histologic distinction between an RTC
not elevated. All patients were treated by orchiectomy and and adenoma or hyperplasia of the rete testis should not
some were also treated with radiotherapy, chemotherapy, be difficult.
and retroperitoneal lymphadenectomy. Of the reviewed
patients, 33% died of RTC, 75% within a year of diagno- Evaluation and management. RTCs are usually found
sis. Twenty patients were alive at the time of the report, incidentally by palpation or at the time of orchiectomy
80% free of disease at a maximum 2-year follow-up. for a testicular mass. They may be suspected on preoper-
ative examination or ultrasound by a hilar location

Figure 37-17 Epidermoid cyst. Sixteen-year-old with cystic testicular mass. The cyst is lined
by flattened squamous epithelium. Laminated keratin is present in the left upper corner.
Mostly flattened seminiferous tubules lined by Sertoli cells are present beneath the cyst.
Leydig cells are prominent. The patient was subsequently found to have Klinefelter’s
syndrome (H&E × 31.2).
Chapter 37 Nongerm Cell Tumors of the Testis 629

Figure 37-18 RTC. Forty-seven-year-old with recurrent hydrocele and firm nontender mass
at the superior pole of the right testis. Orchiectomy specimen contained numerous irregular
firm 1- to 4-cm nodules with involvement of epididymis. Papillary and solid neoplasm
involves the rete testis, surrounding and projecting into rete testis lumens (H&E × 31.2).

Figure 37-19 RTC. Same case as in Figure 37-18. Glandular spaces are lined by epithelial
cells with round to oval, pleomorphic, enlarged nuclei with prominent nucleoli. Glands are
infiltrating connective tissue (H&E × 62.2).

or involvement of the epididymis. Initial therapy is


Malignant Mesothelioma of the Tunica Vaginalis
orchiectomy followed by a staging evaluation for distant
disease.61 One long-term complete remission has been Clinical presentation. Malignant mesothelioma (MM)
reported following retroperitoneal lymphadenectomy for is a rare lesion. The literature consists of 81 cases with
micrometastatic disease. Chemotherapy for disseminated the largest series reported by Jones et al. in 1995.62,63
disease has generally been unsuccessful.61 The age range is 7 to 80 years with a mean of 53.5 years.
630 Part VI Testis

Two-thirds of patients were over 50 years at the time of Pathologic characteristics. Seventy-five percent of the
diagnosis. Most patients presented with a benign appear- tumors described by Jones et al.62 were epithelial and
ing hydrocele with or without an inguinal or scrotal mass. 25% were biphasic. Epithelial tumors grew in a tubular
Of those patients who were asked about asbestos expo- and/or papillary growth pattern (Figure 37-21). Cellular
sure, 41% had some degree of occupational exposure. anaplasia was variable with some tumors comprised of
The large majority of patients had a hydrocele sac stud- bland cells without mitotic activity growing in a papillary
ded with papillary excrescences from several millimeters pattern on fibrous stalks with others containing large
to several centimeters (Figure 37-20). In some cases, a eosinophilic cells with hyperchromatic irregular nuclei,
mass involved the spermatic cord, other paratesticular prominent nucleoli, and frequent mitoses growing in an
structures, or rarely, the testis proper. MM that extended infiltrating tubular pattern. Tumor stroma was usually
beyond the confines of the hydrocele invaded local struc- dense. Psammoma bodies were occasionally present. The
tures, including spermatic cord, epididymis, testis, and majority of tumors, even the best differentiated, showed
penile, scrotal and lower abdominal wall skin. Jones’ some degree of stromal invasion. MMs with a biphasic
series includes follow-up on 52 patients (6 months to 15 pattern had a sarcomatous component with spindled cells
years, mean 2.8 years). Twenty-five patients developed of variable differentiation, sometimes containing numer-
distant metastases. The principle sites of metastasis were ous mitotic figures.
retroperitoneal and inguinal lymph nodes, other lymph MM must be distinguished from a number of benign
nodes and lung. Retroperitoneal and inguinal lymph and malignant lesions. Mesothelial cell hyperplasia,
node involvement was present at the time of diagnosis in although prominent in a hernia sac, is rarely present in a
9 and 4 cases, respectively. Of the 52 patients with fol- hydrocele and does not contain the fibrous stalks charac-
low-up data, 44% died of disease, 17% were alive with teristic of the papillary MM. Adenomatoid tumor is a
disease, and 38% had no evidence of disease. The latter benign tumor derived from mesothelial cells but has a
figure should be considered with caution since late recur- characteristic gland-like architecture and cytology that
rence of well-differentiated epithelial MM has been distinguishes it from MM. It is far more difficult to dis-
observed. Forty-six percent of patients who presented tinguish MM from metastatic adenocarcinoma, RTC,
with tumor confined to the hydrocele sac were free of and serous borderline tumors of müllerian origin (ovar-
disease at 2 years compared with 5.3% of patients who ian surface epithelial type) that resemble ovarian tumors
presented with local invasion of the spermatic cord, skin, of borderline malignancy, which may involve the testis or
testis, or with distant metastasis. In a review of 74 cases paratesticular tissue.65 Borderline serous tumors gener-
of MM of the tunica vaginalis, Plas et al.64 found a ally have broad papillae with stratified epithelial cells
median survival time of 23 months. having a more columnar appearance, some of which are

Figure 37-20 Papillary mesothelioma. Fifty-three-year-old man with recurrent left hydrocele.
The tunica vaginalis is studded with yellow-tan excrescences from 0.2 to 1.0 cm in
maximum dimension.
Chapter 37 Nongerm Cell Tumors of the Testis 631

Figure 37-21 Papillary mesothelioma. Same case as in Figure 37-20. The tumor is
multifocal and comprised of a complex papillary growth lined by a single layer of mesothelial
cells with oval, prominently nucleolated nuclei growing on fibrous stalks. The patient is free
of MM 5 years after orchiectomy (H&E × 3 1.2).

ciliated. Immunohistochemical stains may be useful in tation with the patient. Orchiectomy is associated with a
distinguishing between these two tumors since serous lower local recurrence rate compared with hydrocelec-
tumors are frequently positive for Leu-M1, B72.3, CEA, tomy (11% versus 36%, respectively) although no differ-
and CA125, whereas MMs are negative for these anti- ence in overall survival has been demonstrated.64 A
gens.65 MMs are usually positive for CK 5/6 and calretinin. metastatic evaluation is appropriate but there is little
RTC may be associated with a hydrocele and focally may experience in the literature to address the issues of sur-
have a histologic resemblance to MM, but if the criteria veillance, adjuvant chemotherapy, or prophylactic
of diagnosis enumerated earlier are adhered to, there retroperitoneal or inguinal lymphadenectomy.
should be no problem distinguishing the two entities.
Electron microscopy may also help distinguish the enti-
Metastatic Tumors
ties since mesothelial cells have long thin, bushy
microvilli, and epithelial cells of rete testis origin do not. Symptomatic metastatic nonhematologic tumors of the
As in the pleura and peritoneum, there may rarely be a testis occur only rarely. Price and Mostofi66 identified
problem distinguishing MM from metastatic adenocarci- only 38 metastatic carcinomas involving the testis suit-
noma. The same immunohistochemical stains used to able for study at a time when the AFIP had 1600 primary
distinguish müllerian papillary serous tumors are useful testicular tumors. Four of the patients had bilateral
in distinguishing metastatic adenocarcinomas from MM. tumors. Only 6 tumors were clinically symptomatic, gen-
Adenocarcinomas, that may be of gastrointestinal origin erally due to testicular enlargement. The majority of
should stain positively for Leu-Ml, B72.3, and CEA and tumors in their report were discovered at autopsy. Of the
may contain intracytoplasmic mucin. 38 tumors, 14 were from the lung and 12 were from the
prostate gland. Tiltman67 found metastases in 6 of 248
Evaluation and management. MM is usually not sus- autopsies in males with metastatic carcinoma. Testicular
pected preoperatively; 97.3% are encountered inciden- metastases occurred in 2 of 12 cases of prostate carci-
tally during hydrocelectomy.64 When the initial noma, 2 of 9 cases of malignant melanoma, 1 of 4 cases of
presentation is a scrotal mass, inguinal orchiectomy malignant pleural mesothelioma, and 1 of 89 cases of car-
should be performed with en bloc excision of involved cinoma of the lung. Haupt et al.68 reviewed the literature
adjacent structures. If the tumor is incidentally discov- through 1982 and found the most common tumors
ered during a transscrotal procedure, frozen section metastasizing to the testes were (in descending order)
should be performed and inguinal orchiectomy com- carcinomas of the prostate and lung, malignant
pleted at the same sitting or soon thereafter after consul- melanoma, and carcinomas of the kidney, stomach, and
632 Part VI Testis

pancreas. These findings emphasize the relative rarity of


metastases to the testis and the fact that in most patients
testicular involvement will be discovered incidentally
despite a known history of cancer. Only 24 cases of tes-
ticular enlargement as the primary manifestation of a
tumor metastasis have been reported. These included
primary tumors of the prostate, kidney, and GI tract,
some of which were initially thought to represent pri-
mary testicular tumors even after histologic examination.
Grossly metastatic tumors are generally multinodular
(Figure 37-22). Microscopically, the tumor grows in the
interstitium and may be within endothelial-lined spaces.
The morphology of the tumor may be characteristic of
the primary tumor (Figure 37-23). Treatment for tumors
metastatic to the testis is directed at the underlying Figure 37-22 Metastatic undifferentiated carcinoma, small
malignancy. cell type, of lung. Seventy-six-year-old man with left testicular
mass and previous history of undifferentiated small cell
Carcinoid Tumors carcinoma of lung. Metastatic confluent nodules of neoplasm
are present in the lower left portion of the testis.
Carcinoid tumor (CT) involving the testis and testicular
adnexa represents a special problem inasmuch as both reported through 1992 were metastatic. The largest of
primary and metastatic tumors may have similar mor- the metastatic CTs on which data were recorded was 2.5
phology. The nests of tumor grow in an insular pattern cm, and 3 patients had symptoms of the carcinoid syn-
with groups of uniform centrally nucleated cells having drome. Most patients with metastatic CTs died within a
fine nuclear chromatin and granular eosinophilic cyto- year although one patient survived for 12 years. Factors
plasm (Figure 37-24). The majority of CTs stain posi- that favor a metastatic origin for a CT are bilaterality,
tively with argentaffin and argyrophil stains and for involvement of peritesticular structures, absence of a ter-
chromogranin, neuron-specific enolase, and other atomatous component, and the presence of carcinoid
polypeptides with immunoperoxidase stains. Zavala- syndrome symptomatology. Most primary testicular car-
Pompa et al.69 indicated that 9 of 62 testicular CTs cinoids are cured with inguinal orchiectomy. However,

Figure 37-23 Metastatic prostatic adenocarcinoma. Fifty-eight-year-old man with bilateral


orchiectomy for prostate cancer. One testis contained a firm tan irregular mass. Solid
aggregates of tumor and neoplastic glands expand the interstitium adjacent to a
seminiferous tubule (H&E × 31.2).
Chapter 37 Nongerm Cell Tumors of the Testis 633

Figure 37-24 Carcinoid tumor. Forty-two-year-old male with enlarged right testis and
epididymis containing a 3.5-cm yellow to red, firm to fibrous mass. The tumor is comprised
of solid nests and cysts growing in an insular pattern. Tumor cells contain uniform, centrally
nucleated cells with fine nuclear chromatin and granular eosinophilic cytoplasm (H&E × 3
1.2). (Case courtesy of California Tumor Tissue Registry.)

all patients should have a metastatic evaluation and malignant testicular tumors and, in the case of malako-
retroperitoneal lymphadenectomy should be considered plakia and granulomatous orchitis, from ML.
in those tumors associated with teratoma.

Hematologic Tumors
Other Miscellaneous Tumors
Malignant Lymphoma
A variety of other uncommon and usually benign lesions Clinical presentation. While MLs comprise only a
may mimic more aggressive testicular tumors. Simple small percentage of testicular tumors, they account for
cysts have been described in 17 patients of all ages and more than 50% of testicular tumors in men over age 65.
ranging in size from 1 to several centimeters in diameter About 80% of testicular MLs occur in men over age 50.
but are probably more common.70 Histologically, they The neoplasm generally presents as a unilateral, painless,
are lined by flat or cuboidal epithelium and have a benign
appearance. Cysts may be suspected preoperatively by a
smooth-walled, anechoic appearance on ultrasound.
Adenomatoid tumors are benign mesenchymal prolifera-
tions that usually arise from the epididymis but may also
arise from the tunica albuginea and may infiltrate testic-
ular parenchyma. Two reports of true intratesticular ade-
nomatoid tumors have been published.71,72 These tumors
present as painless enlargement and are firm or hard to
palpation (Figures 37-25 and 37-26). Fibromas and
fibrous pseudotumors of the testicular tunics presenting
as hard painful or incidentally discovered masses have
also been reported. Granulomatous orchitis and malako-
plakia are benign inflammatory conditions usually diag-
nosed following orchiectomy. They have characteristic
histologic appearances, which are pathognomonic. All of Figure 37-25 Adenomatoid tumor of epididymis. The
these lesions are cured by orchiectomy or simple excision epididymal specimen consists of a discrete white uniform
and are important mostly to be differentiated from rubbery 0.9-cm nodule in a 44-year-old man.
634 Part VI Testis

Figure 37-26 Adenomatoid tumor of epididymis. Same patient as in Figure 37-25. Gland-
like structures lined by flattened cells infiltrate collagenous tissue. Numerous intraluminal and
intracytoplasmic vacuoles of varying size are present, creating a spider-web appearance in
some areas (H&E × 31.2).

intrascrotal mass. About 5% of men present with bilat- in the working formulation, 10 immunoblastic, 6 small
eral synchronous involvement, and approximately 20% noncleaved, and 6 were NOS. Of those tumors that were
with a unilateral presentation will develop lymphomatous immunophenotype 33 were of B-cell lineage, one was of
involvement of the opposite testis. Bilateral disease may T lineage, and 5 were of indeterminate lineage.
occur in the absence of any other systemic disease.73 Irrespective of cell type, the pattern of infiltration is sim-
Generally, the diagnosis of ML is first made at the time ilar. Tumor cells proliferate in the interstitium separating
of orchiectomy. Only a small percentage of patients have seminiferous tubules, infiltrating the tunica propria and
a history of antecedent lymphoma. Of 127 men with ML blood vessel walls, eventually obliterating many tubules
of the testis, Gowing74 reported only 8 with antecedent and vessels (Figure 37-27). Sections may contain no rec-
ML and 13 with active lymphoma elsewhere at the time ognizable testicular parenchyma. Tumor cells may extend
of orchiectomy. into the rete testis, epididymis, tunica albuginea, or
peritesticular soft tissue.
Pathologic characteristics. The neoplasm mainly
involves the testis but often extends into the epididymis Evaluation and management. The prognosis of men
or spermatic cord. The testis is enlarged, sometimes mas- with ML of the testis has been poor. Gowing74 indicated
sively, and on cut section is partially or extensively 62% of their patients died of disseminated ML. Only 12
replaced by an ill-defined tan-grey mass. The tumor of 124 patients (10%) survived 5 years after orchiectomy.
merges imperceptibly with testicular parenchyma. In a more recent study, Ferry et al.75 found 20 of 55
Tumor consistency is more rubbery than hard, and patients (36%) to be free of disease a median of 49
necrosis may be present. The large majority of testicular months after orchiectomy, 6 (11%) were alive with dis-
lymphomas are diffuse non-Hodgkin’s type. Gowing74 ease and 29 (53%) died of ML. With careful staging of
indicated that 41% of the British Testicular Tumor Panel ML it is apparent that prognosis is related to the stage of
cases were poorly differentiated lymphocytic lymphomas the disease at the time of orchiectomy. Turner et al.76
and 59% were large cell lymphomas (undifferentiated reported 60% disease-free survival in stage I ML con-
“stem cell reticulum cell type”). They had no cases of trasted with a 17% disease-free survival for stages II, III,
Hodgkin’s disease among their 127 cases. Virtually all and IV. Turner et al.’s cases were classified according to
types of ML other than Hodgkin’s disease occur in the the Rappaport system and the working classification of
testis. Ferry et al.75 in 1994 reported 64 ML, which pre- non-Hodgkin’s lymphomas. In the latter classification,
sented primarily in the testis. Of these, 53 were diffuse the large cell noncleaved, large cell cleaved, and diffuse
large cell lymphoma of which 27 were of noncleaved type mixed lymphomas were designated intermediate grade
Chapter 37 Nongerm Cell Tumors of the Testis 635

Figure 37-27 ML, diffuse large cell type. A 225-g 9.5 cm in maximum dimension testis was
subtotally replaced by a dark tan, focally hemorrhagic, focally necrotic, ill-defined mass. An
extensive cellular infiltrate greatly expands the interstitium and compresses seminiferous
tubules. The individual cells contain large, variably shaped, prominently nucleolated nuclei
with sparse cytoplasm (H&E × 62.2).

lymphomas, and immunoblastic lymphoma, Burkitt’s was 4.8 years, but the survival curves showed no clear evi-
lymphoma and diffuse undifferentiated lymphomas were dence of a substantial portion of cured patients. There
designated as high-grade lymphomas. Eight of 17 men was a 52% relapse rate at a median follow-up of 7.6 years.
with intermediate grade ML were alive and well with an Relapses occurred in the CNS in 15% of the patients,
average follow-up of 24 months. There were no survivors occasionally as late as 10 years postorchiectomy. The
among 6 men in the high-grade group whose average authors identified a continuous risk of contralateral tes-
survival was 13 months. Thus, grade appears to be ticular recurrence in those patients not receiving scrotal
another prognostic variable. Stage I disease, unilateral radiotherapy. On multivariate analysis, lack of B-
right-sided ML, and microscopic sclerosis were also asso- symptoms, use of anthracyclines, and scrotal radiotherapy
ciated with an improved prognosis in Ferry et al.’s were associated with longer survival.
study.75 Evidence is accumulating that primary testicular lym-
In 2001, Lagrange et al.77 reviewed their experience phoma in children has a very different natural his-
with 84 cases of primary testicular lymphoma. The tory.79–81 Most lymphomas involving the testicle are
median age at presentation was 67 (range 17 to 85). secondary lesions in patients with diffuse extrascrotal
Forty-two patients presented with stage I disease, 19 with lymphoma. Only 10 cases of primary testicular lym-
stage II, and 23 with stage III/IV. Using the REAL clas- phoma in children have been reported. All have shared
sification, 75% of the patients exhibited diffuse large B- similar histology (follicular large cell lymphoma) and,
cell histology. Treatment was generally multimodal and despite the aggressive histologic appearance, a good
involved orchiectomy and chemotherapy ± radiation. A prognosis. Most patients have been treated with orchiec-
complete response was obtained in 72.6% of patients tomy and multiagent chemotherapy. The 10 reported
(100%, 68%, and 33% for stages I, II, and III/IV, respec- cases include children ranging in age from 3 to 11 years.
tively). However, median survival was 32 months (52, 32, None of the tumors has demonstrated the presence of
and 12 months for stages I, II, and III/IV, respectively). bcl-2 protein. The size of the tumors ranged from 2 to 4
Zucca et al.,78 in 2003, examined their experience with cm. All 10 patients are reported to be free of disease with
373 patients with primary testicular diffuse large B-cell follow-up ranging from 7 to 59 months.
lymphoma. The median age at diagnosis was 66 years. It is clear that some cases of ML originate in the testis.
Anthracycline-based chemotherapy was used in 68%, Although rare survivals have been reported following
prophylactic intrathecal chemotherapy in 18%, and pro- orchiectomy alone, this is certainly insufficient therapy since
phylactic scrotal radiotherapy in 36%. Median survival the large majority of men ultimately develop extratesticular
636 Part VI Testis

ML. Extratesticular involvement tends to occur in sev- acute leukemia (AL) and 22% of 76 males with chronic
eral areas, including Waldeyer’s ring and the central leukemia (CL) had testicular involvement. All varieties of
nervous system. Turner et al.’s76 series also suggests that AL were represented. It is recognized that children with
MLs at these sites may have been present at the time of acute lymphoblastic leukemia (ALL) in bone marrow
orchiectomy. Ferry et al.’s75 data indicate that MLs that remission may have recurrence first demonstrated in the
relapsed in the testis tended to be extranodal, and that testis. In Givler’s study, 8 children with ALL developed
ML presenting in the testis tended to have lymphoma in testicular masses while receiving chemotherapy. Five of
extranodal sites, notably bone, CNS, skin, orbit, the children were in hematologic remission and the tes-
paranasal sinuses, stomach, nose, thyroid, and larynx.75 ticular masses were either the sole evidence of leukemia
Treatment should be based on stage and histologic type or associated with other extramedullary leukemic masses.
of lymphoma. Hematologic relapse followed in all cases. In 5 of 8 cases,
testicular involvement was bilateral. In cases of ALL fol-
lowing completion of chemotherapy, biopsy of the testes
Plasmacytoma
to rule out occult leukemic involvement is part of some
Plasma cell neoplasms involving the testis are rare and current protocols. Neither palpation nor radiographic
have the same implications as ML. The tumors are bilat- studies, such as ultrasound or MRI, are sufficiently sensi-
eral and sequential in about 20% of men. Tumors gener- tive to the presence of leukemic infiltrates to obviate the
ally manifest as painless enlargement and are part of a need for biopsy. Buchanan et al.87 reported that one-third
systemic process sometimes identified prior to orchiec- of patients with overt testicular recurrence of ALL
tomy. Tumors may be associated with hydroceles and, on treated with an intense protocol exhibited prolonged sec-
at least two occasions, the diagnosis was made on the basis ond remissions with the potential for cure. Only a small
of cytologic analysis of hydrocele fluid.82 All patients with percentage of patients will develop testicular recurrence
sufficient follow-up reviewed by Levin and Mostofi83 and following a negative biopsy. The interstitial pattern of
all 7 in the literature until that time succumbed to sys- testicular involvement in ALL is similar to that of ML.
temic disease. The mean age at the time of diagnosis was Occasionally ALL involvement of the testis produces a
55 years with only 8 patients under 50 and the youngest massive enlargement (Figure 37-29).
aged 26. There have been 42 reported cases of testicular
plasmacytoma, only 10 of which had no documented sys-
Tumors of Generalized Stroma
temic myeloma.82,84,85 The gross and microscopic appear-
ances of plasmacytoma are similar to those of ML, the Tumors of generalized stroma (blood vessels, smooth
only difference being the cell type, which is a neoplastic muscle, and other supporting stroma), also known as
plasma cell of variable morphology (Figure 37-28). mesenchymal tumors, rarely occur in the testis.
Petersen70 reported a total of 26 such tumors described
in the literature, most of which (62%) were malignant.
Leukemia
The histologies of the benign tumors included 5 heman-
Leukemic involvement of the testis is common. In an giomas, 3 hemangioendotheliomas, 1 leiomyoma, and 1
autopsy study, Givler86 found 63% of 140 males with

Figure 37-29 Acute lymphocytic leukemia. Massive


Figure 37-28 Plasmacytoma. Massive enlargement and replacement of right testis by a cream-colored, focally
complete replacement of testicular parenchyma by a hemorrhagic, ill-defined mass in a 10-year-old with an 8-year
lobulated gray-yellow 11-cm mass in a 42-year-old man. history of treated ALL.
Chapter 37 Nongerm Cell Tumors of the Testis 637

myxoid neurofibroma. The malignancies included 14 lial origin are paratesticular, but at least 7 principally
rhabdomyosarcomas, 1 osteosarcoma, and 1 leiomyosar- involved testicular parenchyma. Other areas of involve-
coma. The microscopic appearance of these tumors is ment included the tunica vaginalis and the testicular-
similar to those that occur at more typical sites. Most epididymal groove at the upper pole of the testis. The
of these patients presented with testicular enlargement majority of cases have been of serous borderline type, but
and underwent inguinal orchiectomy with the diagnosis serous papillary carcinoma, mucinous cystadenoma, and
of a mesenchymal tumor made only after surgical exci- cystadenocarcinoma, endometrioid adenoacanthoma,
sion of the testis. Benign tumors found in this manner do clear cell carcinoma, and Brenner tumors have been
not require further evaluation or treatment. reported.92,93 These tumors may be derived from müller-
Rhabdomyosarcomas of the testis proper occur less fre- ian duct remnants, the tunica vaginalis, or both. Most
quently than in paratesticular locations but should be borderline serous tumors behave in a benign fashion if
similarly treated with retroperitoneal lymphadenectomy, completely excised, but the clear cell adenocarcinoma
chemotherapy, and radiation, depending on stage. and two cases of papillary serous carcinoma behaved in a
malignant fashion, one causing metastasis and death and
another having extensive abdominal recurrence.93 A third
Testis Tumors in Acquired Immunodeficiency
patient without demonstrable recurrence has persistent
Syndrome
elevation of serum CA125.93 It is important to recognize
Patients infected with HIV or with full-blown AIDS the müllerian nature of these neoplasms and to accurately
appear to be at a 20- to 50-times higher risk of develop- distinguish between borderline and frankly malignant
ing primary or secondary testis tumors.88 Testis tumors variants.
are the third most common HIV-associated/AIDS-asso- The differential diagnosis of müllerian surface epithe-
ciated malignancy.88 The first 2 cases of germ cell tumors lial tumors includes germ cell tumors, rete testis adenocar-
of the testis in AIDS patients were reported in 1985.89 cinoma, mesothelioma of the tunica vaginalis, and
Wilson et al.89 reported 5 of 3015 HIV positive men pre- metastatic adenocarcinoma. Germ cell tumors, because
senting with testicular tumors over a 5-year period, an they are the most frequent tumor in this region, must be
incidence of 0.2%, more than 50 times the incidence of considered in the differential diagnosis, but only teratomas
testis tumors in the general population. There have been with mucinous glandular differentiation or transitional cell
additional cases of testis tumors described in the AIDS foci might remotely be considered in the differential diag-
population in the urologic literature, including semi- nosis of mucinous or Brenner variants of müllerian
noma and mixed nonseminomatous germ cell tumors, tumors. As described earlier, rete testis adenocarcinomas
Kaposi’s sarcoma, MLs, and plasmacytomas.90 Some of have a combination of diagnostic features that must be rec-
these cases presented with testicular swelling as the initial ognized for a correct diagnosis. Paratesticular and tunica
and primary manifestation of HIV infection,91 and some vaginalis MM may closely resemble paratesticular and
of these patients presented with symptoms of acute pro- tunica vaginalis borderline serous müllerian tumors. MMs
statitis or epididymo-orchitis and were initially treated are about three times more frequent. Müllerian tumors
with antibiotics before returning with complaints of pro- stain positively for keratin and frequently for CEA, B72.3,
gressive testicular enlargement. Reported cases were ulti- BER-EP4, EMA, Leu-M1, S-100 protein, and PLAP,
mately managed by inguinal orchiectomy and additional whereas MMs stain positively for cytokeratins, including
therapy based on tumor histology and status of the patient’s CK 5/6 and calretinin.65 Although borderline serous
immune system. These reports highlight the need for a tumors and epithelial MMs may have papillary configura-
high index of suspicion for testicular tumors in men at tions, borderline serous tumors generally have stratified
risk for HIV who present with testicular enlargement. nuclei and may be ciliated, whereas MMs are lined by a
single cell layer. Metastatic carcinomas to the testis, tunica
vaginalis and paratesticular tissues are rare but should be
Testicular and Paratesticular Tumors of Ovarian
considered in the differential diagnosis.
Surface Epithelial Type
Prior to 1995, 17 cases had been reported of testicular
and paratesticular tumors of ovarian surface epithelial REFERENCES
type, when Jones et al. reported an additional 5 cases of
paratesticular serous carcinoma.92,93 These occurred in 1. Buzelin F, Karam G, Moreau A, Wetzel O, Gaillard F:
an age range of 11 to 68 years with a median of 47 years Testicular tumors and the acquired immunodeficiency
in Young et al.’s series of testicular and paratesticular syndrome. Eur Urol 1994; 26:71–76.
tumors and in a range of 16 to 42 years with a mean of 31 2. Mueller N: Overview of the epidemiology of malignancy
years in Jones et al.’s series of paratesticular serous carci- in immune deficiency. J Acquir Immune Defic Syndr
noma. The majority of tumors of ovarian surface epithe- 1999; 21(S1):S5–S10.
638 Part VI Testis

3. Gabrilove JL, Nicolis GL, Mitty HA, Sohval AR: 19. Palazzo JP, Petersen RO, Young RH, Scully RE:
Feminizing interstitial cell tumor of the testis: personal Deoxyribonucleic acid flow cytometry of testicular Leydig
observations and a review of the literature. Cancer 1975; cell tumors. J Urol 1994; 152:415–417.
35:1184–1202. 20. McCluggage WG, Shanks JH, Arthur K, Banerjee SS:
4. Elert A, Olbert P, Hegele A, et al: Accuracy of frozen Cellular proliferation and nuclear ploidy assessments
section examination of testicular tumors of uncertain augment established prognostic factors in predicting
origin. Eur Urol 2002; 41(3):290–293. malignancy in testicular Leydig cell tumours.
5. Richter-Unruh A, Wessels HT, Menken U, et al: Male Histopathology 1998; 33(4):361–368.
LH-independent sexual precocity in a 3.5-year-old boy 21. Ulbright TM, Srigley JR, Hatzianastassiou DK, Young
caused by a somatic activating mutation of the LH RH: Leydig cell tumors of the testis with unusual features:
receptor in a Leydig cell tumor. J Clin Endocrinol Metab adipose differentiation, calcification with ossification, and
2002; 87(3):1052–1056. spindle-shaped tumor cells. Am J Surg Pathol 2002;
6. Dieckmann K, Loy V: Metachronous germ cell and 26(11):1424–1433.
Leydig cell tumors of the testis: Do testicular germ cell 22. Kirkland RT, Kirkland JL, Keenan BS, et al: Bilateral
tumors and Leydig cell tumors share common etiologic testicular tumors in congenital adrenal hyperplasia. J Clin
factors? Cancer 1993; 72:1305–1307. Endocrinol Metab 1977; 44:369–378.
7. Poster RB, Katz DS: Leydig cell tumor of the testis in 23. Rich MA, Keating MA, Levin HS, Kay R: Tumors of the
Klinefelter syndrome: MR detection. J Comput Assis adrenogenital syndrome: an aggressive conservative
Tomogr 1993; 17:480–481. approach. J Urol 1998; 160(5):1838–1841.
8. Calleja RK, Rice A, Bullock KN: Unilateral Leydig cell 24. Stikkelbroeck NM, Otten BJ, Pasic A, et al: High
tumour associated with a contralateral undescended testis. prevalence of testicular adrenal rest tumors, impaired
BJU Int 1999; 83(1):152. spermatogenesis, and Leydig cell failure in adolescent and
9. Hekimgil M, Altay B, Yakut BD, et al: Leydig cell tumor adult males with congenital adrenal hyperplasia. J Clin
of the testis: comparison of histopathological and Endocrinol Metab 2001; 86(12):5721–5728.
immunohistochemical features of three azoospermic 25. Davis JM, Woodroof J, Sadasivan R, Stephens R: Case
cases and one malignant case. Pathol Int 2001; report: congenital adrenal hyperplasia and malignant
51(10):792–796. Leydig cell tumor. Am J Med Sci 1995; 309:63–65.
10. Mostafid H, Nawrocki J, Fletcher MS, Vaughan NJ, 26. Levin HS: Tumors of the testis in intersex syndromes.
Melcher DH: Leydig cell tumour of the testis: a rare Urol Clin North Am 2000; 27(3):543–551.
cause of male infertility. Br J Urol 1998; 81(4):651. 27. Bertram KA, Bratloff B, Hodges GF, Davidson H:
11. Soria JC, Durdux C, Chretien Y, et al: Malignant Leydig Treatment of malignant Leydig cell tumor. Cancer 1991;
cell tumor of the testis associated with Klinefelter’s 68:2324–2329.
syndrome. Anticancer Res 1999; 19(5C):4491–4494. 28. van der Hem KG, Boven E, van Hennik MB, Pinedo
12. Fowler KA, Gill K, Kirma N, Dillehay DL, Tekmal RR: HM: Malignant Leydig cell tumor of the testis in
Overexpression of aromatase leads to development of complete remission on o,p′-dichlorodiphenyl-
testicular Leydig cell tumors: an in vivo model for dichloroethane. J Urol 1992; 148(4):1256–1259.
hormone-mediated testicular cancer. Am J Pathol 2000; 29. Young RH, Koelliker DD, Scully RE: Sertoli cell tumors
156(1):347–353. of the testis, not otherwise specified: a clinicopathologic
13. Liu G, Duranteau L, Carel JC, et al: Leydig-cell tumors analysis of 60 cases. Am J Surg Pathol 1998;
caused by an activating mutation of the gene encoding the 22(6):709–721.
luteinizing hormone receptor. N Engl J Med 1999; 30. Zukerberg LR, Young RH, Scully RE: Sclerosing Sertoli
341(23):1731–1736. cell tumor of the testis: a report of 10 cases. Am J Surg
14. Masoudi JF, Van Arsdalen K, Rovner ES: Organ-sparing Pathol 1991; 15:829–834.
surgery for bilateral Leydig cell tumor of the testis. 31. Gravas S, Papadimitriou K, Kyriakidis A: Sclerosing Sertoli
Urology 1999; 54(4):744. cell tumor of the testis—a case report and review of the
15. Gittes RF, Smith G, Conn CA, Smith F: Local androgenic literature. Scand J Urol Nephrol 1999; 33(3):197–199.
effect of interstitial cell tumor of the testis. J Urol 1970; 32. Proppe KH, Scully RE: Large-cell calcifying Sertoli cell
104:774–777. tumor of the testis. Am J Clin Pathol 1980; 74:607–619.
16. Kim F, Young RH, Scully RE: Leydig cell tumors of the 33. Proppe KH, Dickerson GHL: Large-cell calcifying
testis. A clinicopathologic analysis of 40 cases and review Sertoli cell tumor of the testis. Hum Pathol 1982;
of the literature. Am J Surg Pathol 1985; 9:177. 13:1109–1114.
17. Cheville JC, Sebo TJ, Lager DJ, Bostwick DG, Farrow 34. Tetu B, Ro JY, Ayala AG: Large cell calcifying Sertoli cell
GM: Leydig cell tumor of the testis: a clinicopathologic, tumor of the testis: a clinicopathologic,
DNA content, and MIB-1 comparison of immunohistochemical and ultrastructural study of two
nonmetastasizing and metastasizing tumors. Am J Surg cases. Am J Clin Pathol 1991; 96:717–722.
Pathol 1998; 22:1361–1367. 35. Kratzer SS, Ulbright TM, Talerman A, et al: Large cell
18. Gulbahce HE, Lindeland AT, Engel W, Lillemoe TJ: calcifying Sertoli cell tumor of the testis: contrasting
Metastatic Leydig cell tumor with sarcomatoid features of six malignant and six benign tumors and a
differentiation. Arch Pathol Lab Med 1999; review of the literature. Am J Surg Pathol 1997;
123(11):1104–1107. 21(11):1271–1280.
Chapter 37 Nongerm Cell Tumors of the Testis 639

36. Nogales FF, Andujar M, Zuluaga A, Garcia-Puche JL: 54. Stewart DA, Stewart DJ, Mai KT: Active chemotherapy
Malignant large cell calcifying Sertoli cell tumor of the for metastatic stromal cell tumor of the testis. Urology
testis. J Urol 1995; 153:1935–1937. l993; 42:732–734.
37. Cano-Valdez AM, Chanona-Vilchis J, Dominguez- 55. Price EB Jr: Epidermoid cysts of the testis: a clinical and
Malagon H: Large cell calcifying Sertoli cell tumor of the pathologic analysis of 69 cases from the testicular tumor
testis: a clinicopathological, immunohistochemical, and registry. J Urol 1969; 102:708–731.
ultrastructural study of two cases. Ultrastruct Pathol 1999; 56. Cho JH, Chang JC, Park BH, Lee JG, Son CH:
23(4):259–265. Sonographic and MR imaging findings of testicular
38. Dryer L, Jacyk WK, du Plessis DJ: Bilateral large-cell epidermoid cysts. AJR Am J Roentgenol 2002;
calcifying Sertoli cell tumor of the testes with 178(3):743–748.
Peutz–Jeghers syndrome: a case report. Pediatr Dermatol 57. Ross JH, Kay R, Elder J: Testis sparing surgery for
1994; 11:335–337. pediatric epidermoid cysts of the testis. J Urol l993;
39. Washecka R, Dresner MI, Honda SA: Testicular tumors 149:353–356.
in Carney’s complex. J Urol 2002; 167(3):1299–1302. 58. Davi RC, Braslis KG, Perez JL, Soloway MS: Bilateral
40. Young RH, Scully RE (eds): Testicular Tumors, pp epidermoid cysts of the testis. Eur Urol 1996;
140–150. Chicago, ASCP Press, 1990. 29(1):122–124.
41. Rutgers JL, Scully RE: Pathology of the testis in intersex 59. Baniel J, Perez JM, Foster RS: Benign testicular tumor
syndromes. Semin Diagn Pathol 1987; 4:275–291. associated with Klinefelter’s syndrome. J Urol 1994;
42. Wysocka B, Serkies K, Debniak J, Jassem J, Limon J: 151:157–158.
Sertoli cell tumor in androgen insensitivity syndrome—a 60. Orozco RE, Murphy WM: Carcinoma of the rete testis:
case report. Gynecol Oncol 1999; 75(3):480–483. case report and review of the literature. J Urol l993;
43. Athanassiou AE, Barbounis V, Dimitriadis M, Pectasidis 150:974–977.
D, Bafaloukos D: Successful chemotherapy for 61. Stein JP, Freeman JA, Esrig D, Chandrasoma PT, Skinner
disseminated testicular Sertoli cell tumour. Br J Urol DG: papillary adenocarcinoma of the rete testis: a case
1988; 61(5):456–457. report and review of the literature. Urology 1994;
44. Jimenez-Quintero LP, Ro JY, Zavala-Pompa A, et al: 44:588–594.
Granulosa cell tumor of the adult testis: a 62. Jones MA, Young RH, Scully RE: Malignant
clinicopathologic study of seven cases and a review of the mesothelioma of the tunica vaginalis. A clinicopathologic
literature. Hum Pathol 1993; 24:1120–1126. analysis of 11 cases with review of the literature. Am J
45. Al-Bozom IA, El-Faqih SR, Hassan SH, El-Tiraifi AE, Surg Pathol 1995; 19:815–825.
Talic RF: Granulosa cell tumor of the adult type: a case 63. Iczkowski KA, Katz G, Zander DS, Clapp WL:
report and review of the literature of a very rare testicular Malignant mesothelioma of tunica vaginalis testis: a fatal
tumor. Arch Pathol Lab Med 2000; 124(10):1525–1528. case with liver metastasis. J Urol 2002; 167(2 Pt
46. Morgan DR, Brame KG: Granulosa cell tumour of the 1):645–646.
testis displaying immunoreactivity for inhibin. BJU Int 64. Plas E, Riedl CR, Pfluger H: Malignant mesothelioma of
1999; 83(6):731–732. the tunica vaginalis testis: review of the literature and
47. Nieto N, Torres-Valdivieso MJ, Aguado P, et al: Juvenile assessment of prognostic parameters. Cancer 1998;
granulosa cell tumor of the testis: case report and review 83(12):2437–2446.
of literature. Tumori 2002; 88(1):72–74. 65. De Nictolis M, Tommasoni S, Fabris G, Prat,
48. Lawrence WD, Young RH, Scully RE: Juvenile granulosa J: Intratesticular serous cystadenoma of borderline
cell tumor of the infantile testis. Am J Surg Pathol 1985; malignancy. A pathological, histochemical and DNA
9:87–94. content study of a case with long-term follow-up.
49. Tanaka Y, Sasaki Y, Tachibana K, et al: Testicular juvenile Virchows Archiv A Pathol Anat 1993; 423:221–225.
granulosa cell tumor in an infant with X/XY mosaicism 66. Price EB Jr, Mostofi FK: Secondary carcinoma of the
clinically diagnosed as true hermaphroditism. Am J Surg testis. Cancer 1957; 10:592–595.
Pathol 1994; 18:316–322. 67. Tiltman AJ: Metastatic tumors of the testis.
50. Paquis-Flucklinger V, Rassoulzadegan M, Michiels J-F: Histopathology 1979; 3:31–37.
Experimental Sertoli cell tumors in the mouse and their 68. Haupt HM, Mann RB, Trump DL, Abeloff MD:
progression into a mixed germ cell-sex cord proliferation. Metastatic carcinoma involving the testis. Clinical and
Am J Pathol 1994; 144:454–459. pathologic distinction from primary testicular neoplasms.
51. Lawrence WD, Young RH, Scully RE: Sex cord-stromal Cancer 1984; 54:709–714.
tumors. In Talerman A, Roth LM (eds): Pathology of the 69. Zavala-Pompa A, Ro JY, el-Naggar A, et al: Primary
Testis and its Adnexa, Vol 7, Chap 4, Contemporary Issues carcinoid tumor of testis. Immunohistochemical,
in Surgical Pathology, pp 67–92. New York, Churchill ultrastructural and DNA flow cytometric study of three
Livingstone, 1986. cases with a review of the literature. Cancer 1993;
52. Dieckmann K-P, Loy V: Response of metastasized sex 72:1726–1732.
cord gonadal stromal tumor of the testis to cisplatin-based 70. Peterson RO (ed): Urologic Pathology, 2nd edition,
chemotherapy. J Urol 1994; 151:1024–1026. p 451. Philadelphia, JB Lippincott, 1992.
53. Gohji K, Higuchi A, Fujii A, Kizaki T: Malignant gonadal 71. Horstman WG, Sands JP, Hooper DG: Adenomatoid
stromal tumor. Urology 1994; 43:244–247. tumor of testicle. Urology 1992; 40(4):359–361.
640 Part VI Testis

72. Samad AA, Pereiro B, Badiola A, Gallego C, Zungri E: 82. Suzuki K, Shioji Y, Morita T, Tokue A: Primary testicular
Adenomatoid tumor of intratesticular localization. Eur plasmacytoma with hydrocele of the testis. Int J Urol
Urol 1996; 30(1):127–128. 2001; 8(3):139–140.
73. Hurley LJ, Burke CR, Shetty SK, et al: Bilateral primary 83. Levin HS, Mostofi FK: Symptomatic plasmacytoma of the
non-Hodgkin’s lymphoma of the testis. Urology 1996; testis. Cancer 1970; 25:1193–1203.
47(4):596–598. 84. Reddi VR, Anne GP, Rani AV, et al: Primary
74. Gowing NFC: Malignant lymphoma of the testis. In Pugh plasmacytoma of testis. Report of a case. Indian J Cancer
RCB (ed): Pathology of the Testis, pp 334–355. Oxford, 1998; 35(4):152–155.
Blackwell Scientific Publications, 1976. 85. Fischer C, Terpe HJ, Weidner W, Schulz A: Primary
75. Ferry JA, Harris NL, Young RH, et al: Malignant plasmacytoma of the testis. Case report and review of the
lymphoma of the testis, epididymis and spermatic cord. literature. Urol Int 1996; 56(4):263–265.
A clinicopathologic study of 69 cases with 86. Givler RL: Testicular involvement in leukemia and
immunophenotypic analysis. Am J Surg Pathol 1994; lymphoma. Cancer 1969; 23:1290–1295.
18:376–390. 87. Buchanan GR, Boyett JM, Pollock BH, et al: Improved
76. Turner RR, Colby TV, MacKintosh FR: Testicular treatment results in boys with overt testicular relapse
lymphomas, a clinicopathologic study of 35 cases. Cancer during or shortly after initial therapy for acute
1981; 48:2095–2102. lymphoblastic leukemia. A Pediatric Oncology Group
77. Lagrange JL, Ramaioli A, Theodore CH, et al: Radiation Study. Cancer 1991; 68:48–55.
Therapy Group and the Genito-Urinary Group of the 88. Leibovitch I, Baniel J, Rowland RG, et al: Malignant
French Federation of Cancer Centres. Non-Hodgkin’s testicular neoplasms in immunosuppressed patients.
lymphoma of the testis: a retrospective study of 84 J Urol 1996; 155(6):1938–1942.
patients treated in the French anticancer centres. Ann 89. Wilson WT, Frenkel E, Vuitch F, Sagalowsky AI:
Oncol 2001; 12(9):1313–1319. Testicular tumors in men with human immunodeficiency
78. Zucca E, Conconi A, Mughal TI, et al: Patterns of virus. J Urol 1992; 147:1038–1040.
outcome and prognostic factors in primary large-cell 90. Ramadan A, Naab T, Frederick W, Green W: Testicular
lymphoma of the testis in a survey by the international plasmacytoma in a patient with the acquired
extranodal lymphoma study group. J Clin Oncol 2003; immunodeficiency syndrome. Tumori 2000;
21(1):20–27. 86(6):480–482.
79. Pakzad K, MacLennan GT, Elder JS, et al: Follicular 91. Munver R, Donehower RC, Kronz JD, Polascik TJ: HIV
large cell lymphoma localized to the testis in children. infection presenting as an unusually large pure yolk sac
J Urol 2002; 168(1):225–228. tumor of the testis. J Urol 2000; 164(5):1653–1654.
80. Finn LS, Viswanatha DS, Belasco JB, et al: Primary 92. Young RH, Scully RE: Testicular and paratesticular
follicular lymphoma of the testis in childhood. Cancer tumors and tumor-like lesions of ovarian common
1999; 85(7):1626–1635. epithelial and Mullerian types. Am J Clin Pathol 1986;
81. Pileri SA, Sabattini E, Rosito P, et al: Primary follicular 86:146–152.
lymphoma of the testis in childhood: an entity with 93. Jones MA, Young RH, Srigley JR, Scully RE:
peculiar clinical and molecular characteristics. J Clin Paratesticular serous papillary carcinoma. A report of six
Pathol 2002; 55(9):684–688. cases. Am J Surg Pathol 1995; 19:1359–1365.
C H A P T E R

38 Radical Orchiectomy and


Retroperitoneal Lymph Node Dissection
Richard S. Foster, MD, Ashraf Mosharafa, MD,
and Richard Bihrle, MD

inguinal orchiectomy includes an inguinal approach to


RADICAL ORCHIECTOMY
high ligation of the spermatic cord at the internal ring
General Considerations
and the subsequent removal of the testis. It is typically
Approximately 95% of primary intratesticular tumors are performed as an outpatient procedure.
of germ cell origin. The other 5% of tumors consist of a The technique is relatively straightforward; an
variety of benign and malignant lesions. These include inguinal incision is made parallel to the inguinal ligament
Leydig cell and Sertoli cell tumors, adrenal cortical rests, and the dissection is carried through the subcutaneous
and other benign lesions.1 Similarly, secondary tumors of tissues to the aponeurosis of the external oblique. The
the testis may occur rarely and mainly are hematopoietic external ring is identified, and the aponeurosis of the exter-
in origin. Hence, patients presenting with a tumor that is nal oblique is split from the external ring to the internal
intratesticular are usually found histologically to have a ring. Cremasteric fibers are divided and the ilioinguinal
germ cell tumor. nerve is identified and dissected from the cord structures.
Most patients present with a palpable intratesticular Next, the cord is dissected from the inguinal floor and
mass. Typically, the mass is firm and definitely different circumferential control is attained at the internal ring. If
from the consistency of normal testicular tissue. If such a the decision has been made to perform inguinal orchiec-
lesion is discovered, immediate determination of serum tomy, the cord is divided into 2 segments with a large
alpha fetoprotein and beta-HCG is necessary and the clamp, after which the segments are ligated with perma-
elevation of either of these markers confirms the diagno- nent sutures and/or suture ligatures. Sometimes it is nec-
sis of a germ cell tumor. If the tumor is palpable, testicu- essary to dissect the peritoneum away from the medial
lar ultrasound is not necessary and the patient may be aspect of the cord prior to performing ligation. Typically,
taken immediately to radical inguinal orchiectomy. after ligating the cord the cord stump retracts into the
Alternatively, if there is doubt as to the diagnosis, transs- abdomen through the internal ring. This facilitates
crotal ultrasound is useful and the finding of an intrates- removal of the cord stump at subsequent retroperitoneal
ticular abnormality on testicular ultrasound in a patient lymph node dissection (RPLND). After ligation of the
in the appropriate age range for germ cell tumors may cord the dissection is carried distally with subsequent
confirm the diagnosis. Magnetic resonance imaging has mobilization of the testis through the upper part of the
been studied in the diagnosis of testicular tumors but is scrotum and division of the gubernacular fibers. If
rarely needed clinically.2 the testis tumor is so large that it cannot be easily deliv-
Historically, radical inguinal orchiectomy meant ered through the opening in the scrotum, the incision is
removal of the testis and cord but sometimes also extended on to the scrotum in order to remove the testis
included an extension of the incision and palpation of the intact. After hemostasis is obtained the aponeurosis
retroperitoneum.3 With modern imaging and staging of the external oblique and Scarpa’s fascia are subse-
techniques this extension of the incision to palpate the quently closed. Typically, the skin is closed using a
retroperitoneum is not necessary. Currently, radical running subcuticular suture.

641
642 Part VI Testis

If the diagnosis is unclear at the time of exploration of lymphatic metastasis. From a urologic point of view,
and a frozen section is necessary, the same steps are per- this is perhaps the most unique aspect of the surgical
formed with the exception of early division of the cord. treatment of germ cell tumors.
Typically, if a frozen section is to be obtained, a tourni- Hence, since surgery may be curative in many patients
quet is applied to the cord at the internal ring after which with metastatic germ cell tumor, urologic oncologists
the testis is mobilized up into the incision. Drapes are necessarily need to understand and be proficient in tech-
placed over the incision and the frozen section is done niques of removal of lymph nodes in the retroperi-
away from the incision itself so as to not spill tumor into toneum. RPLND is essentially two different operations;
the incision. Subsequently, if a frozen section diagnosis one operation for low-stage disease in patients who have
confirms a germ cell tumor, the radical orchiectomy is not received chemotherapy and a completely different
completed. operation for those patients who require an RPLND for a
The reasoning behind an inguinal approach to a germ residual mass after chemotherapy. These two techniques
cell tumor of the testis is to avoid spilling tumor into the are discussed separately.
wound. Typically, germ cell tumor implants and if tumor
is spilled into the inguinal area, potentially another area
RETROPERITONEAL LYMPH NODE
of lymphatic drainage (inguinal nodes) is contaminated.
DISSECTION FOR LOW-STAGE DISEASE
Historically, if a scrotal approach to a testis tumor was
carried out, a subsequent recommendation was made to Low-stage seminoma is not managed surgically and the
perform hemi-scrotectomy so as to avoid contamination discussion here relates only to low-stage nonseminoma.
of the inguinal lymphatics. With the advent of systemic Clinical stage I disease is defined as no evidence of metas-
chemotherapy this procedure now is rarely necessary tasis on radiologic imaging. Typically, this involves CT
although the inguinal approach to radical orchiectomy scanning of the abdomen and chest. Some prefer only a
continues to be the standard of care and should be appro- chest x-ray as opposed to a CT of the chest. Additionally,
priate management for any patient presenting with a serum alpha fetoprotein and beta-hCG should have nor-
germ cell tumor of the testis.4 malized after radical orchiectomy or alternatively should
Approximately 1% to 2% of all patients who present be decaying based on normal half-lives of approximately
with a testicular germ cell tumor will develop a subse- 5 days for alpha fetoprotein and 11⁄2 days for beta-hCG.9
quent tumor on the contralateral side.5 Historically, rad- If the patient has no evidence of metastasis on radiologic
ical inguinal orchiectomy was performed for the second imaging but these markers are not normalizing according
tumor and the patient was placed on testosterone supple- to half-life, the patient should be treated with
mentation so as to maintain libido and sexuality. It is now chemotherapy because of the high probability of having
clear that small secondary tumors at the polar aspects of occult systemic disease.10,11
the testis can be managed by partial orchiectomy with Several alternatives for management exist in clinical
good long-term results. Most series of partial orchiec- stage I that in the short term yield roughly the equivalent
tomy have recommended postoperative radiation therapy chance for survival. These alternatives include RPLND
to the testis, which prevents local recurrence and eradi- (with nerve sparing), surveillance with chemotherapy at
cates carcinoma in situ in the remaining testicular tissue.6 relapse, or primary chemotherapy. Pro and con argu-
The benefit of this approach is that over the long term ments exist for each of these approaches depending on
these patients are able to sustain production of androgens long-term side effects, psychologic issues, access to
from the remaining Leydig cells and therefore do not health care, etc. As opposed to surveillance, the benefits
need supplementation. However, the postoperative radi- of RPLND include the immediate determination of
ation therapy effectively eliminates fertility, and therefore pathologic stage, the avoidance of chemotherapy (since
the benefit of partial orchiectomy is to maintain adequate many of these patients are cured with surgery alone), and
androgen serum levels. the elimination of the necessity of using CT scans of the
abdomen in follow-up. Conversely, the follow-up period
after RPLND is only 2 years, whereas patients managed
RETROPERITONEAL LYMPH NODE DISSECTION
on surveillance have to be followed for a longer period of
The germ cell tumors of the testis are not only chemo- time.12 Similarly, the main benefit of surveillance is that
sensitive but also “surgery sensitive.” Even after lym- patients who have no metastasis receive no treatment and
phatic metastasis has occurred, the surgical removal of only patients documented to have metastatic disease are
involved lymph nodes may be curative from 50% to 75% subject to any sort of therapy.
of the time, dependent on the volume of metastatic dis- Approximately 30% of clinical stage I patients are sub-
ease.7,8 Most other cancers (breast, colon, lung, etc.) are sequently found to have metastatic disease.13 Generally,
not surgically curable if lymphatic metastasis occurs; metastatic tumor is found in retroperitoneal lymphatics.
germ cell testis tumors are surgically curable in the face Rarely, the retroperitoneum is normal and metastasis is
Chapter 38 Radical Orchiectomy and Retroperitioneal Lymph Node Dissection 643

hematogenous only. Hence, the high probability of


retroperitoneal metastasis is the rationale for performing
RPLND. Risk factors identified in the orchiectomy spec-
imen may increase the probability of metastasis to 50%,
but prediction of metastasis at a higher level than 50% is
not possible.14 Therefore, clinical stage I patients will
have a 50% to 70% chance of having normal lymph
nodes removed.
Since some patients who undergo RPLND for clinical
stage I disease have no metastasis, it is important and per-
tinent to limit the morbidity of the procedure. Currently,
RPLND for low-stage disease has an acceptably very low
morbidity, which consists of about a 1% chance of devel-
oping a subsequent small bowel obstruction and a 3% to
5% chance of an abdominal incisional hernia.15 The
operative procedure is approximately 2 to 2 hours in
length. Transfusions are not required and the average
hospitalization is about 3.3 days. Return to full physical
activity occurs in three to 6 weeks depending on age of
the patient and body habitus.

TECHNIQUE FOR LOW-STAGE DISEASE


In low-stage nonseminoma mapping studies have shown
that metastatic spread is unilateral.16 Hence, for a patient
presenting with a right-sided primary, the involved lym-
phatics are usually interaortocaval, precaval, and right
paracaval (Figure 38-1). Similarly, for a left-sided pri-
mary, the involved lymphatics are typically left paraaortic
and preaortic (Figure 38-2). Though historically full
bilateral RPLND was performed (Figure 38-3), these
mapping studies illuminated the fact that a unilateral Figure 38-1 The template of dissection for a patient
template could be used. This was important since a uni- presenting for a right-sided primary is displayed.
lateral template dissection preserved contralateral sym-
pathetics important for emission and ejaculation. These discovery of higher-volume metastasis than was predicted
so-called modified templates preserved ejaculation at the by preoperative scans, a full bilateral dissection may be
30% to 90% level, depending on individual technique.7,8 required. Currently, CT scans are high resolution and
In order to further minimize morbidity, it was neces- the intraoperative discovery of unsuspected high-volume
sary to improve the technique to maintain emission and metastasis is rare.
ejaculation at a higher level. These modifications were After palpation and inspection an incision is made in
termed nerve sparing since nerve-sparing RPLND the posterior peritoneum from the cecum to the area
involves the prospective dissection of efferent sympathetic of the ligament of Treitz. The root of the small bowel
fibers followed by removal of lymphatics based on a uni- and the right colon are reflected to the patient’s right and
lateral template (Figure 38-4).17,18 Thus, nerve-sparing held in place with self-retaining retractors. The template
RPLND preserves sympathetic nerves bilaterally of dissection includes the interaortocaval, precaval, and
whereas a template dissection preserves only contralat- right paracaval lymph nodes. The so-called “split and roll”
eral nerves. The prospective dissection and preservation maneuver is employed. This is essentially a vascular iso-
of efferent sympathetic fibers maintains emission and lation technique whereby lymphatic tissue is split at the
ejaculation at the 99% level. 12 o’clock position over vessels and rolled laterally away
from those vessels. RPLND is a vascular procedure.
First, the split maneuver is performed over the left renal
Right Nerve-Sparing RPLND
vein and tissue is rolled inferiorly. The anterior aspect of
A midline incision is made and a self-retaining retractor the aorta is identified posterior to the left renal vein and
is used. A general palpation and inspection of the the split maneuver is carried out on the 12 o’clock posi-
abdomen is performed and in the rare circumstance of tion of the aorta from the crossing of the left renal vein
644 Part VI Testis

Figure 38-2 The template of dissection for a left-sided Figure 38-3 Full bilateral RPLND includes the removal of
primary is displayed. right paracaval, interaortocaval, left peri-aortic, and interiliac
nodal packages.
distally to the origin of the inferior mesenteric artery.
Tissue is rolled medially into the interaortocaval zone in tocaval area and then pass distally into the interiliac area
order to determine whether or not a lower pole right- at the bifurcation of the aorta. Attention is then turned
sided precaval renal artery is present. Attention is then back to the aorta at which point the split maneuver is
turned to the vena cava. The split maneuver is performed continued along the distal aorta and right common iliac
at the 12 o’clock position on the vena cava (Figure 38-5). artery. The nerves have been previously dissected and
The origin of the right gonadal vein is identified and therefore are avoided in the process of this split maneu-
divided between silk ties. It is subsequently dissected ver. Tissue is rolled medially into the interaortocaval area
to the internal ring at which point the cord stump from and the right-sided lumbar arteries are divided between
the radical orchiectomy is identified, mobilized from the ties. The renal artery is typically seen passing over the
internal ring, and the specimen is removed as right crus of the diaphragm and lymphatic tissue, is dissected
gonadal vein. The roll maneuver is then performed on away from it. Finally, the right ureter is dissected laterally
the vena cava after which all lumbar veins passing poste- away from lymphatic tissue after which the right para-
riorly from the cava to the posterior body wall are iden- caval and interaortocaval nodal packages are dissected
tified and divided between silk ties (Figure 38-6). from the posterior body wall (Figure 38-7). Clips are
Typically, the efferent sympathetic fibers from the right used as necessary to secure lymphatics or to gain hemo-
sympathetic chain pass cephalad to these lumbar veins. stasis from the lumbars as they penetrate the posterior
Care must be taken to not injure the nerves in the course body wall. Cautery is also helpful in this regard. After
of dividing the veins. The nerves are then dissected away irrigation the posterior peritoneum is closed. Closure of
from lymphatic tissue and placed in vessel loops. the abdominal incision is performed using a looped
Typically, the right-sided nerves coalesce in the interaor- absorbable suture.
Chapter 38 Radical Orchiectomy and Retroperitioneal Lymph Node Dissection 645

Figure 38-5 Viewed from the patient’s left side, the split
maneuver on the superior portion of the aorta and the vena
cava as performed for a right modified nerve-sparing RPLND
is displayed.

Technique for Left-Sided Nerve-Sparing RPLND


For a left-sided dissection, after palpation and inspection
has been performed, an incision is made in the posterior
peritoneum lateral to the left colon. The left colon is
mobilized medially and held in place with self-retaining
retractors. The first step in a left-sided dissection is to
identify the efferent sympathetic fibers. Typically, these
can be seen coursing anterior to the left common iliac
artery and can be dissected away from lymphatic tissue
Figure 38-4 The anatomy of retroperitoneal sympathetic and placed in vessel loops (Figure 38-8). If these fibers
nerves in relationship to the vascular structures is shown. are not easily seen at this level an alternative approach is
to mobilize the left ureter laterally and roll lymphatic tis-
sue off the psoas muscle to expose the sympathetic chain.
The efferent fibers can then be seen passing from the
sympathetic chain to the interiliac area. After the nerve

Figure 38-6 Mobilization of the vena cava is performed by rolling lymphatic tissue away
from the vena cava, identifying the lumbar veins and subsequently dividing them. The three
lumbar veins passing to the posterior body wall are shown in the figure prior to division.
646 Part VI Testis

Figure 38-7 In a completed right modified nerve-sparing Figure 38-9 The lymphatic tissue has been rolled away from
RPLND all lymphatic tissue is removed in the interaortocaval the left side of the aorta. Two lumbar arteries passing
and right para-caval packages. Shown in the figure are the posteriorly are seen. A silk tie has been passed around one in
mobilized vena cava, the sympathetic chain, and the efferent preparation for subsequent ligation distally and division of the
sympathetic fibers in vessel loops. lumbar artery.

fibers are identified they are dissected slightly proximally. Next, the split maneuver is performed on the aorta from
In a left-sided dissection, the lymphatics intermingle the crossing of the left renal vein distally to the bifurca-
with the efferent fibers and the dissection is technically tion of the left common iliac artery. The bifurcation of
slightly more difficult because of this. The split maneu- the left common iliac represents the lower boundary
ver is performed over the left renal vein and tissue is of the dissection. Tissue is rolled into the left paraaor-
rolled inferiorly. The origin of the left gonadal vein is tic area and the left-sided lumbar arteries are identi-
identified, dissected, and divided between silk ties. The fied, dissected, and divided between silk ties (Figure
left gonadal vein is then dissected to the internal ring at 38-9). Care must be taken to determine whether or not
which point the cord stump is mobilized from the inter- lower pole renal arteries exist and these should be pre-
nal ring and the left gonadal vein specimen is removed. served if possible. The ureter is dissected laterally away

Figure 38-8 In this figure the anatomy of left-sided sympathetic efferent fibers is shown.
Note the confluence of the fibers anterior to the left common iliac artery. Right-sided fibers
join the left-sided fibers in the inter-iliac area.
Chapter 38 Radical Orchiectomy and Retroperitioneal Lymph Node Dissection 647

from the lymphatic tissue and this represents the lateral


POSTCHEMOTHERAPY RETROPERITONEAL
boundary of the dissection. Finally, lymphatic tissue is
LYMPH NODE DISSECTION
dissected off the posterior body wall as one package
Indications
taking care to avoid the sympathetic chain and the
efferent sympathetic fibers passing into the interiliac Patients who present with high volume distant metastatic
area. At the renal hilum lymphatic tissue is dissected germ cell cancer are typically managed with systemic
away from the renal artery and clips are applied at the chemotherapy. After the administration of systemic
crus of the diaphragm. The left colon is then placed chemotherapy, if serum alpha-fetoprotein and beta-hCG
back in the anatomic position and the abdomen have normalized and no evidence of metastatic tumor
is closed as noted earlier with a running, looped remains on radiographic imaging, patients are observed.
absorbable suture. The probability of relapse in this clinical situation is low
and hence observation is reasonable. Some centers advo-
cate postchemotherapy RPLND in the absence of per-
POSTOPERATIVE MANAGEMENT
sistent radiographic tumor if the transverse diameter of
Historically, nasogastric tubes were used because of the disease in the retroperitoneum before chemotherapy was
suspicion of the postoperative paralytic ileus. Currently, >3 cm.19 However, the practice at Indiana University is to
no postoperative nasogastric decompression is necessary. observe patients with a complete clinical remission.20
Patients are given liquids the day after the procedure and If patients normalize serum markers and have persist-
subsequently are advanced to full diet. The average hos- ent tumor on radiographic imaging in the retroperi-
pitalization for this procedure is around 3 days and return toneum, postchemotherapy RPLND is advised (Figure
to full physical activity is three to 6 weeks. Patients are 38-10). Histologically, remaining tumor consists of ter-
followed postoperatively with chest x-rays, physical atoma in 40% to 60% of cases, fibrosis and necrosis in
examination, and determination of serum alpha-fetopro- approximately 40% of cases, and persistent germ cell
tein and beta-hCG. This monitoring typically is per- cancer (or nongerm cell cancer arising in a teratoma) in
formed for 2 years. The frequency of these examinations 5% to 10% of cases. The surgical removal of teratoma or
is contingent on the pathologic stage. CT scans of the cancer is therapeutic and hence the rationale for RPLND
abdomen are not necessary postoperatively. in these clinical situations is solid. However, the surgical

Figure 38-10 CT scan performed after chemotherapy in a patient with normal alpha
fetoprotein and beta-hCG. Pathologically the resected tumor proved to be teratoma.
648 Part VI Testis

removal of necrosis confers no survival advantage on the there is commonly a fibrotic and desmoplastic reaction in
patient and it would be desirable to select such patients the retroperitoneum that makes tumor and lymphatics
preoperatively so as to avoid postchemotherapy quite adherent to surrounding structures, such as the
RPLND. It is very difficult to clinically select for patients aorta, the vena cava, and the renal arteries and veins.
who have only necrosis and therefore RPLND is advised Dissection along the great vessels should be in an extra
for any persistent mass. Exceptions to this rule include adventitial plane so as to not weaken these vessels, which
those patients with pure seminoma managed with sys- can lead to significant vascular problems. Surgeons who
temic chemotherapy and some highly selected patients undertake postchemotherapy RPLND should be well
with no teratoma in the orchiectomy specimen who versed in techniques of vascular control and repair, as this
experience a dramatic response to systemic chemother- essentially is a vascular procedure.
apy. However, controversy exists in the management of The “subtraction” technique was initially described by
the postchemotherapy mass in pure seminoma patients. Donohue. This technique employs the prospective dis-
Some advocate resection of the mass if it is >3 cm in section of the vessels away from tumor followed by resec-
transverse diameter; at Indiana University these patients tion of tumor and lymphatics from the posterior body
are observed because of the high probability of the mass wall (Figure 38-11). Hence, after the incision is made,
containing fibrosis only.21,22 the retroperitoneum is exposed by incising in the poste-
Rarely, postchemotherapy RPLND is recommended rior peritoneum from the foramen of Winslow distally
in patients who have not experienced a normalization of around the cecum up to the area of the inferior mesen-
serum AFP or HCG. These are very highly selected teric vein. The inferior mesenteric vein is divided and the
patients who have failed all systemic chemotherapy but right colon and root of the small bowel is dissected off
have a persistent retroperitoneal mass. Since beta-HCG the retroperitoneum and retracted onto the patient’s
and/or alpha fetoprotein are elevated, germ cell cancer chest with a self-retaining retractor. Typically, tumor
remains within the mass. Postchemotherapy RPLND in does not invade the mesentery but sometimes may invade
this situation is termed “desperation RPLND” but is the duodenum, which requires resection of duodenum
capable of curing such patients around 30% of the time.23 and subsequent repair. Fortunately, invasion of the duo-
It is truly remarkable that in this situation of chemo denum is relatively rare and typically the bowel is easily
refractory metastatic disease surgery remains a therapeu- dissected onto the patient’s chest. The split maneuver is
tic option. Testis cancer is a unique and interesting then performed on the left renal vein and tissue is rolled
disease. inferiorly. The anterior aspect of the aorta is identified
and the split maneuver is performed on the aorta at the
12 o’clock position from the crossing of the left renal
TECHNIQUE OF POSTCHEMOTHERAPY RPLND
vein distally to the origin in the inferior mesenteric
Generally, postchemotherapy RPLND is a full bilateral artery. For full bilateral dissection the inferior mesen-
dissection, which includes the resection of tumor and teric artery is typically dissected and divided between silk
lymphatics from the crus of the diaphragm to the bifur-
cation of the common iliac arteries, from ureter to ureter.
The reasoning behind performing a full bilateral dissec-
tion is that mapping studies showed that the higher vol-
ume of metastasis, the greater the likelihood of bilateral
retroperitoneal disease. It is clear, however, that highly
selected patients do not require full bilateral RPLND
and that the likelihood of bilateral disease in some
patients is very low. Typically, these are patients who have
been administered so-called good risk chemotherapy and
initially presented with relatively low volume metastatic
disease to the retroperitoneum. However, generally, full
bilateral RPLND is recommended.
The type incision is contingent on the position and
size of the tumor. Depending on the individual patient,
a midline, chevron, supra 11th extra pleural approach,
or thoracoabdominal approach may be used. The same Figure 38-11 The subtraction concept involves the
techniques employed in low-stage disease are used in mobilization of great vessels away from retroperitoneal tumor
postchemotherapy RPLND. These techniques include and lymphatics with a subsequent resection of tumor and
the split and roll technique and in highly selected patients, lymphatics from the posterior body wall. In this figure the
nerve sparing. However, in postchemotherapy RPLND aorta is being mobilized from tumor and lymphatics.
Chapter 38 Radical Orchiectomy and Retroperitioneal Lymph Node Dissection 649

ties. This allows the left mesocolon to be retracted later-


ally. Because testis cancer is typically a disease of young
men, the vascularity of the left colon is maintained
through collaterals and colon ischemic has not occurred
in these postchemotherapy patients. The dissection is
then carried distally along the aorta and along both com-
mon iliac arteries. The split maneuver is continued and
subsequently tissue is rolled medially and laterally off the
aorta and the common iliac arteries. Lumbar arteries are
subsequently identified and divided. Typically, three lum-
bar arteries exist on each side of the aorta and their posi-
tion is fairly predictable. Superiorly, the left gonadal vein
is divided from the left renal vein, and the left renal vein
and left renal artery are dissected from lymphatics and
tumorous tissue. Attention is then turned to the vena
cava. A similar split maneuver at the 12 o’clock position
is performed on the vena cava. The origin of the right
Figure 38-12 Postchemotherapy, full bilateral RPLND with
gonadal vein is divided and subsequently tissue, tumor, bilateral nerve sparing. Such patients maintain emission and
and lymphatics are rolled medially and laterally from the ejaculation and if recovery from chemotherapy is complete
vena cava. The lumbar veins are identified, dissected, and may maintain fertility.
divided between ties. The position and size of the lumbar
veins is not as predictable as arterial lumbar anatomy.
Lymphatics and tumor are dissected from the right renal Nerve sparing is possible in some patients who
artery as it passes over the lower portion of the crus of undergo postchemotherapy RPLND.25 Indeed, some of
the diaphragm. Similarly, the ureters are dissected later- these patients may recover fertility after recovery from
ally away from the lymphatics and tumorous tissue. The the effects of chemotherapy. Again, the decision to per-
gonadal vein is removed along with the cord stump, form nerve sparing is dependent on the clinical situation
depending on the laterality of the primary. Finally, as the and intraoperative findings (Figure 38-12).
vessels and ureters have been dissected away from the The complications of postchemotherapy RPLND
tumor and lymphatics, these lymphatic packages are dis- are generally more significant and frequent compared
sected from the posterior body wall. Typically, in full to primary RPLND.26 The major source of posto-
bilateral RPLND there will be four packages: the right perative morbidity relates to the lungs, as many of
paracaval, interaortocaval, left para-aortic, and interiliac. these patients have received bleomycin as a part of the
The bowel is then placed back in anatomic position and chemotherapeutic regimen. Typically, an effort is made
secured in anatomic position with running absorbable to restrict fluids perioperatively so that if bleomycin-
sutures. The bowel is run to make sure there is no evi- induced ARDS occurs the patient is not volume over-
dence of any retractor injury, after which closure of the loaded. Other complications of postchemotherapy
abdomen is performed. RPLND include ileus, lymphatic or chylous ascites, and
other complications associated with a major surgical
procedure. Nasogastric tubes are used variably and the
SPECIAL CONSIDERATIONS
hospitalization is typically more lengthy compared to
Preoperatively, postchemotherapy RPLND patients are primary RPLND.
given informed consent indicating that intraoperative
decisions may be necessary. The decision to resect adja-
SUMMARY
cent organs, such as bowel or kidneys, requires intra-
operative judgment.24 A decision to resect an adjacent Germ cell tumors of the testis are not only chemo sensi-
organ depends on the amount of adherence to the tumor tive but also surgery sensitive. For optimal care of a
and also the clinical situation. For instance, the thresh- patient with germ cell cancer chemotherapeutic therapies
old for resecting an adjacent organ is much lower in and surgical techniques are complimentary. Surgeons
desperation RPLND as opposed to straightforward who perform these procedures should be well versed in
RPLND performed for a patient with low volume tumor techniques of vascular mobilization and control. Finally,
and normal markers. However, a partial removal of though some of these procedures may be time consum-
tumor is not therapeutic and surgeons performing ing and arduous providing, excellent long-term outcomes
RPLND should have the mind set to remove all palpable are possible after complete surgical removal of metastatic
and visual tumor. tumor.
650 Part VI Testis

REFERENCES 14. Read G, Stenning SP, Cullen MH, et al: Medical research
council prospective study of surveillance for stage I
1. Ulbright TM, Amin MB, Young RH: Tumors of the testicular teratoma. J Clin Oncol 1992; 10:1762.
Testis, Adnexa, Spermatic Cord, and Scrotum. 15. Baniel J, Foster RS, Rowland RG, et al: Complications of
Washington, Armed Forces Institute of Pathology, 1999. primary retroperitoneal lymph node dissection. J Urol
2. Thurnher S, Hricak H, Carroll PR, et al: Imaging the 1994; 152:424.
testis: comparison between MR staging and us. Radiology 16. Donohue JP, Zachary J, Maynard B. Distribution of nodal
1988; 167:633. metastases in nonseminomatous testis cancer. J Urol 1982;
3. Hinman F: The operative treatment of tumors of the 128:315.
testicle with the report of thirty cases treated by 17. Jeweh MA, Kong YS, Goldberg SD, et al: Retroperitoneal
orchiectomy. JAMA 1914; 63:2009. lymphadenectomy for testis tumor with nerve sparing for
4. Our scrotal violation paper. ejaculation. J Urol 1988; 139:1220.
5. Kristianslund S, Fossa SD, Kjellevold K: Bilateral 18. Donohue JP, Foster RS, Rowland RG, et al: Nerve-
malignant testicular germ cell cancer. Br J Urol 1986; sparing retroperitoneal lymphadenectomy with
58:60. preservation of ejaculation. J Urol 1990; 144:287.
6. Heidenreich A, Weissbach L, Holtl W, et al: Organ 19. Toner GC, Panicek DM, Heelan RT, et al: Adjunctive
sparing surgery for malignant germ cell tumor of the surgery after chemotherapy for nonseminomatous germ
testis. J Urol 2001; 166:2161. cell tumors: recommendations for patient selection. J Clin
7. Richie JP: Clinical stage I testicular cancer: the role of Oncol 1990; 8:1683.
modified retroperitoneal lymphadenectomy. J Urol 1990; 20. Debono DJ, Heilman DK, Einhorn LH, et al: Decision
144:1160. analysis for avoiding post chemotherapy surgery in
8. Donohue JP, Thornhill JA, Foster RS, et al: patients with disseminated nonseminomatous germ cell
Retroperitoneal lymphadenectomy for clinical stage A tumors. J Clin Oncol 1997; 15:1455.
testis cancer (1965 to 1989): modifications of technique 21. Motzer R, Bosl G, Heelan R, et al: Residual mass: an
and impact on ejaculation. J Urol 1993; 149:237. indication for further therapy in patients with advanced
9. Nichols CR, Timmerman R, Foster RS, et al: Neoplasms seminoma following systemic chemotherapy. J Clin Oncol
of the testis in cancer medicine. Baltimore, Williams & 1987; 5:1064.
Wilkins, 1997 22. Schutlz SM, Einhorn LH, Conces DJ, et al: Management
10. Davis BE, Herr HW, Fair WR, et al: The management of of post chemotherapy residual mass in patients with
the patients with nonseminomatous germ cell tumors advanced seminoma: Indiana University experience. J Clin
of the testis with serologic disease only after orchiectomy. Oncol 1989; 7:1497.
J Urol 1994; 152:111. 23. Donohue JP, Leibovitch I, Foster RS, et al: Integration of
11. Saxman SB, Nichols CR, Foster RS, et al: The surgery and systemic therapy: results and principles of
management of patients with clinical stage I integration. Semin Urol Oncol 1998; 16:65.
nonseminomatous testicular tumors and persistently 24. Our nephrectomy paper.
elevated serologic markers. J Urol 1996; 155:587. 25. Wahle, GR, Foster RS, Bihrle R, et al: Nerve-sparing
12. Sharir S, Foster RS, Donohue JP, et al: What is the retroperitoneal lymphadenectomy after primary
appropriate follow-up after treatment? Semin Urol Oncol chemotherapy for metastatic testicular carcinoma. J Urol
1996; 14:45. 1994; 152:428.
13. Roeleveld TA, Horenblas S, Meinhardt W, et al: 26. Baniel J, Foster RS, Rowland RG, et al: Complications of
Surveillance can be the standard of cave for stage I postchemotherapy retroperitoneal lymph node dissection.
nonseminomatous testicular tumors and even high risk J Urol 1994; 152:424.
patients. J Urol 2001; 166:2166.
C H A P T E R

39 Retroperitoneal Tumors: Diagnosis,


Staging, Surgery, Management, and
Prognosis
S. Bruce Malkowicz, MD, and Victor Ferlise, MD

Retroperitoneal tumors are uncommon lesions that are Less than one-half of all retroperitoneal tumors are
important to the urologist because they occur in our sur- retroperitoneal sarcomas. Generally, 15% to 20% of
gical domain and often involve urologic organs. These retroperitoneal tumors are benign (e.g., lipoma). The
lesions often present as very large lesions, which have remainders comprise lymphomas or primary urologic
been relatively indolent. Because of their rarity (0.5% to tumors.2 Approximately 500 to 1000 new cases of
1.0% of adult malignancies) it is important to be aware of retroperitoneal sarcoma are diagnosed each year.
the systematized approach to the treatment of these Incidence figures on specific genitourinary sarcomas are
tumors. Beyond the attempt to attain complete surgical difficult to establish due to the rarity of such lesions.2,3
resection, it is difficult to outline the optimal treatment Retroperitoneal sarcomas arise most commonly in the
scheme for these lesions since there are few large cohorts 5th and 6th decade of life, but age incidence may span
of patients treated in a similar fashion. Additionally, from the 2nd to 8th decades.3,4
many clinical reports have a combination of extremity, as There is a slight male predominance but no distinct
well as retroperitoneal lesions mixed together in the ethnic or racial distribution. While any histologic pat-
results. tern may be seen at any age, rhabdomyosarcoma gener-
The clinical assessment of these lesions and the surgi- ally clusters in younger patients, even excluding the
cal approach to therapy is fairly well outlined and pediatric population, and malignant fibrous histiocy-
remains the foundation of treatment. Radiation therapy toma is usually seen in older age groups. Generally, these
and chemotherapy for these lesions continue to evolve, lesions are not associated with other conditions and a
although their overall efficacy in retroperitoneal lesions pattern of familial transmission has not been demon-
is unclear. strated. However, rare patients with neurofibromatosis
Primary sarcomas of the genitourinary organs are may develop malignant schwannomas at an anatomic
even rarer than primary retroperitoneal sarcomas. When site.5
radical extirpation is possible, it is usually the appropriate There is little known of the etiology of retroperitoneal
option, although in some instances, such as bladder sarcomas. Radiation injury, prior trauma, and environ-
lesions, other choices do exist. The evaluation of adju- mental exposure to agents, such as dioxin and asbestos,
vant and salvage therapies for these lesions is limited. But have been implicated.6,7 Radiation may predispose
overall frequently with these lesions it is required to reor- patients to the development of sarcomas. Approximately
ganize and appropriately treat these lesions. 0.1% of patients treated with radiation therapy who sur-
vive >5 years may develop a sarcoma at that site.8 To
qualify as a postradiation sarcoma a lesion must meet spe-
PRIMARY RETROPERITONEAL SARCOMA
cific established criteria.6 In these cases, the sarcoma has
Incidence and Etiology
to develop within the radiated field and prior documen-
Retroperitoneal sarcomas are rare tumors that account tation stating that the area was normal must be estab-
for only 0.1% to 0.2% of all malignant tumors and lished. Additionally, histologic confirmation of the
approximately 10% to 20% of all soft tissue sarcomas.1 diagnosis is necessary, and a latency of at least 3 years is

651
652 Part VI Testis

required. There appears to be no difference in the inci- gene and can significantly affect the expression of that
dence of postradiation-induced disease between those gene at the messenger RNA level. The nucleic acid bind-
patients treated with orthovoltage and megavoltage. The ing domain of the chimeric transcription factor confers
most common postradiation tumor is the malignant target specificity within the tissue genome, while the tran-
fibrous histiocytomas (MFHs) followed by osteosarcoma scription factor portion of this novel protein determines
and fibrosarcoma. Most of these postradiation-induced the transactivation potential and expression level of the
lesions appear to be of high grade and generally have target gene.29–31
poorer survival.8 Additionally, the development of extra abnormal chro-
Earlier epidemiologic studies reported that exposure mosomes (ring chromosomes and giant rods) involving
to herbicides, such as dioxin and wood preservatives, may chromosome 12 can result in the amplification of certain
contribute to the development of retroperitoneal sarco- gene products, such as MDM2 and SAS. MDM2 can be
mas.9 More recent data are mixed in its findings and may involved in p53 inactivation that can contribute to car-
be affected by the particular herbicide used and the cinogenesis32,33 Additionally, alterations in cell cycle reg-
degree of dioxin contamination.10 In studies of Vietnam ulating elements, such as CDK4, have been
era soldiers exposed to Agent Orange, no significant demonstrated.34 Novel these insights into sarcoma
association was found between the development of sarco- pathology may provide approaches for future therapeutic
mas and exposure in case control studies.11 In some strategies directed at these lesions.
industrial studies, an association was noted between
dioxin exposure and the development of sarcomas if the
Benign Lesions
exposure was prolonged (>1 year) and the period signifi-
Lipomas
cant (over 20 years).12 No distinct viral or immunologic
etiologies have been proposed for the development of Lipomas consist almost entirely of mature fat and are
retroperitoneal sarcomas. uncommonly found in the retroperitoneum (Table 39-1).
They are probably the most common soft tissue tumor in
man. Most of these lesions occur superficially but they
PATHOLOGY
may occur in other areas, such as the retroperitoneum.
Retroperitoneal sarcomas arise primarily from soft tis- Deep lipomas within the retroperitoneum are usually not
sues of fibrous and adipose origin, as well as muscle, as well circumscribed as their superficial counterparts
nerve, and lymphatic tissues. These tissues are derived and can conform to irregular spaces in this body
from primitive mesenchyme from the mesoderm with space.35,36 The adipocytes are normal or slightly larger in
some contribution from neuroectoderm.13 Their loca- appearance and have a well-developed vascular network.
tion allows for a rather long indolent preclinical course at There is very little in the way of nuclear irregularity.
that time the tumor can grow to significant proportions. Differing levels of fibrous connective tissue can be found
This growth may result in local areas of necrosis or liq- in these lesions. The rim of the lipocyte is reactive for S-
uefaction as the tumor outstrips its vascular supply. 100 protein. While the majority of these masses are idio-
In classic reviews of these lesions the common tissue pathic in nature, they can occasionally be a manifestation
distribution in descending order is liposarcoma, of steroid lipomatosis.
leiomyosarcoma, and fibrosarcoma followed by other Pelvic lipomatosis while not a distinct tumor per se was
histologies.4,14–26 MFH figures much more prominently first described in 1959 as an overgrowth of fat in the
in contemporary series; however, owing to intensive perivesical and perirectal area. It is a hyperplastic rather
pathologic interest in defining this disorder, many
tumors previously described as variants of fibrosarcoma
or liposarcoma have been reclassified as MFH.27,28 Table 39-1 Benign Lesions of the Retroperitoneum
Therefore, fibrosarcoma has been replaced in frequency Lipoma
order by this condition.
Although a well-developed understanding of the funda- Pelvic lipomatosis
mental pathology of these lesions has not yet emerged,
Myelolipoma
evaluation of the cytogenetic alterations in many sarcomas
is beginning to suggest some molecular themes. Several of Leiomyoma
these tumors display specific chromosomal translocations.
Although the translocations appear unique for specific Ganglioneuroma
tumors [t(12;16)(q13;p11) in myxoid liposarcoma] nearly
Hemangiopericytoma
all of these translocations result in the production of novel,
tumor-specific, chimeric, transcription factors. These fac- Schwannoma
tors interact with the upstream regulatory component of a
Chapter 39 Retroperitoneal Tumors 653

neoplastic entity which can create a space occupying rate of metastasis depends on the degree of tumor differ-
lesion. Approximately two-thirds of patients are African- entiation, with nearly 90% of poorly differentiated
American and women are rarely affected.37 The growth is tumors metastasizing.46–48
diffused rather than nodular and often it is difficult to dis- Significant advances in cytogenetics has allowed the
tinguish it from normal adipose tissue. The condition may reclassification of these lesions on a molecular basis
be associated with cystitis glandularis.38 The general clini- (Table 39-2). Well-differentiated and dedifferentiated
cal course is slowly progressive and may result in the need
for urinary diversion.39 Occasionally fat necrosis in these
lesions can be mistaken for sarcomatous degeneration.40
Table 39-2 Malignant Lesions of the Retroperitoneum
and Histologic Subclassifications
Myelolipoma Liposarcoma

This is a tumor-like growth of mature fat and bone mar- Myxoid liposarcoma
row elements. Although it usually occurs in the adrenal
gland, it can be seen as an isolated pelvic lesion.41,42 It is Well differentiated
distinct from extra-medullary hematopoietic tumors Lipoma-like
which are usually multiple and generally associated with
mild proliferative diseases and skeletal disorders. These Inflammatory
generally occur in patients older than 40 years of age and
are rarely >5 cm in size.43 They are usually found as inci- Sclerosing
dental imaging findings. The adrenal gland can create Differentiated
inferior renal displacement due to radiolucent mass.
Pathologically, it may display the features of a lipoma or Round cell
have a darker appearance if myeloid elements predomi-
nate. Adrenal myelolipoma development may be second- Pleomorphic
ary to prolonged stress and excessive stimulation with Leiomyosarcoma
adrenocorticotropic hormones.
Malignant fibrous histiocytoma (MFH)
Leiomyoma Storiform-pleomorphic
These, generally rare lesions, are seen in a distribution Myxoid MFH
that represents smooth muscle tissue in the body. They
are overwhelmingly found in the female genital tract but MFH-giant cell type
have also been reported in the urinary bladder. There are
occasional reports of these tumors in the retroperi- Inflammatory MFH
toneum, where they can grow asymptomatically to a con- Fibrosarcoma
siderable size. Leiomyomas stain positive for desmin,
which separates them from their malignant counterpart. Rhabdomyosarcoma
Extension of these lesions from the uterus into the vas-
cular system can create tumor thrombi not unlike those Embryonal
seen with renal lesions.44 Botryoid

Alveolar
Malignant Lesions
Liposarcoma Pleomorphic
Liposarcomas are among the most common of primary Spindle cell
retroperitoneal tumors that are often distinguished by
their large dimensions and range of subtypes. These Malignant hemangiopericytoma
lesions are found with their peak incidence between ages
40 and 60.45 They account for 10% to 15% of sarcomas Malignant peripheral nerve sheath tumors (malignant
and approximately 20% of these lesions arise in the schwannoma)
retroperitoneum. One unfortunate clinical feature of Synovial sarcoma
these lesions is their great tendency to recur often within
the first 6 months after surgery. The principal tissue type Angio sarcoma
is usually recapitulated at the time of recurrence. The
654 Part VI Testis

lesions are a continuum of lesions based on the genetic lesions (Figure 39-2). These lesions have as their com-
abnormality of giant and ring chromosomes usually mon pathology fusion proteins as mentioned previously.
involving chromosome 12. Gene amplification, particu- Higher-grade lesions tend to demonstrate a higher num-
larly MDM2 drives their pathology.49,50 Myxoid and ber of p53 mutations.52 The round cell subtype is also
round cell, cell lesions (poorly differentiated myxoid) are referred to as a lipoblastic variant and is distinguished by
another continuum, which have fusion transcripts caused sheets of round cells with lipoblastic differentiation.
by translocations in chromosomes 16 and 12 as their These lesions are considered the most aggressive part of
principle pathologic feature. Pleomorphic liposarcomas the spectrum of myxoid lesions.
are rare and poorly understood.51 Well-differentiated liposarcomas mostly resemble
Unlike benign lipomas, liposarcomas may bear little lipomas and are usually designated as low grade. They
resemblance to classic fat filled structures. The gross are a common sarcoma of later life. Subvariants include
lesion is usually described as having a “fish flesh” appear- the lipoma like, inflammatory, sclerosing, and differenti-
ance, and while generally encapsulated, it can often ated. The first 3 variants are often confused with benign
display invasive characteristics. Besides the retroperi- processes, such as scarring or inflammation, while the
toneum, these lesions arise in the deep soft tissues of the differentiated subtype is often noted in long-standing
proximal extremities. They generally present as very retroperitoneal lesions and considered higher grade.
large lesions when originating from the retroperi- Generally, these lesions are now considered as a group
toneum. with dedifferentiated lesions since they share genetic
Myxoid liposarcoma is the most common liposarcoma similarities.
usually occurring in the lower extremity. It accounts for Pleomorphic liposarcoma comprises 10% to 15% of
50% of sarcomas and represents a large proportion of liposarcomas and are defined as a high-grade malignant
retroperitoneal lesions. Its peak presentation is in the variant with very bizarre nuclei and huge lipoblasts.
fifth decade. They display a background of stellate mes-
enchymal cells, and a prominent capillary pattern often
Leiomyosarcoma
described as a “chicken wire” (Figure 39-1). The distinct
cell is the lipoblast that is similar to the fetal adipocyte. This tumor accounts for <10% of all soft tissue sarcomas,
These cells are noted by a lipid vacuole that scallops the yet it is a significant percentage of retroperitoneal sarco-
nucleus. This creates the lipoma-like appearance of these mas since almost half of them appear in this region. They

Figure 39-1 Myxoid liposarcoma displaying myxoid vascular pattern and minimal lipoblasts
(H&E × 250).
Chapter 39 Retroperitoneal Tumors 655

Figure 39-2 Myxoid liposarcoma displaying lipoblasts and myxoid matrix (250 ×).

have a 2:1 female to male presentation and generally vessels usually present with lower extremity edema, while
occur in middle age (mean age 60). The molecular those of the inferior vena cava can display findings consis-
pathology of these lesions is not understood. They are tent with Budd-Chiari syndrome.55 Resection of these
usually categorized by site of origin (soft tissue, vascular, lesions is recommended when anatomically feasible.
or superficial) although many of the soft tissue lesions are Survival, however, is usually <2 years and many inferior
felt to originate from smaller blood vessels. On imaging vena caval tumors are unresectable because of the intra-
examinations, however, they appear to have moderate to hepatic location of many of these lesions.
low vascularization.
Grossly, low-grade leiomyosarcoma can be difficult to
Malignant Fibrous Histiocytoma
distinguish from leiomyomas, but higher-grade lesions
display a more infiltrative flesh-like appearance.53 Low- MFH was originally described in 1963 and has come to
grade lesions are often distinguished from leiomyomas by be the predominant histologic diagnosis for contem-
their chromatin pattern and the number of mitoses (>5 porarily reported soft tissue sarcomas.56 It was defined as
mitoses per high power field) present in the specimen. a sarcoma of primary histocytic origin, yet it is now felt
Again, a continuum exists in the pathologic progression of that it is a lesion derived from fibroblast differentiation.
these smooth muscle lesions; therefore, the cut point Many pleomorphic variants of fibrosarcoma, liposar-
between benign, moderate and high grade may sometimes coma, and rhabdomyosarcoma have been reclassified
be arbitrary. There is little evidence of sarcomatous degen- into this category. With the thorough evaluation of a
eration from benign leiomyomas. The characteristic tumor specimen, distinct regions of leiomyosarcoma,
pathologic finding of a leiomyosarcoma is malignant spin- liposarcoma, and other soft tissue sarcomas may be iden-
dle cells with “cigar”-shaped nuclei. The muscle fascicles tified. In such cases, the tumor may be defined by those
interweave. These tumors immunohistochemically stain findings and the mention of associated MFH pattern.57
for smooth muscle myosin, vimentin, actin, and less often These lesions are most often seen on the extremities and
for desmin. They stain negative for S-100. Ultrastructural are less common in the retroperitoneum. Their molecu-
features include bundles of thin cyto-filaments that can lar pathology is unclear. An analysis of these lesions in
help distinguish leiomyosarcomas from other lesions. the retroperitoneum suggests that many of these could
The rare retroperitoneal variants of these tumors are be reclassified as a dedifferentiated liposarcoma.58
leiomyosarcomas, which originate from the great vessels.54 Several subtypes of MFH that may coexist in a particular
These occur predominantly in women. Tumors of the iliac lesion: storiform-pleomorphic, myxoid, giant cell, and
656 Part VI Testis

inflammatory. The angiomatoid variant is seen almost with matrix of reticular cells. Often an exact diagnosis is
exclusively in the extremities and trunk.59 not made until a recurrence is biopsied.
Storiform-pleomorphic variant has a distinct but not
pathognomonic pinwheel pattern caused by the collagen
Fibrosarcoma
pattern of curling fascicles of cells (Figure 39-3). It is the
most common variant, usually comprising 40% to 60% Fibrosarcomas are malignant tumors of fibroblast origin.
of most series. While this pattern may predominate, it They have a range of gradation and grossly display a clas-
may not be present throughout the lesion in question. sic “fish flesh” pattern with hemorrhage and necrosis.
Areas of collagen or foamy cells may also be encountered. Low-grade lesions can display a herringbone—spindle-
The nuclei tend to be irregular and large with discernible shape histologic pattern. On retrospective review, these
mitosis. The constellation of tissue histology and nuclear lesions are often reclassified as MFH or as a desmoid
irregularities in addition to the inflammatory compo- lesion (aggressive fibromatosis). Therefore, these lesions
nents differentiate this lesion from benign entities. represent a smaller proportion of primary retroperitoneal
Myxoid MFH make 25% of this overall tumor type. It is tumors than previously reported. There is a greater inci-
often difficult to distinguish it from liposarcoma, yet the dence of blood bone metastases with this tumor to the
myxoid MFH has little neutral fat but is rich in lung and bones.60
mucopolysaccharide residing in intracytoplasmic vacuoles.
MFH giant cell type is distinguished by large benign
Rhabdomyosarcoma
appearing osteoclasts. Multinucleated giant cells and the
stoma comprise the malignant components of this lesion. Rhabdomyosarcoma comprises a minor percentage of
If cartilage or osteoid is detected, these lesions are classi- reported retroperitoneal sarcomas, yet it is a significant
fied as a soft tissue osteosarcoma, which usually has a lesion in pediatric oncology, as well as pediatric tumors
poorer prognosis than MFH. associated with the genitourinary system. This topic is dealt
Inflammatory malignant fibrous histiosarcoma is rare with extensively in Chapter 49. These lesions are generally
and often difficult to distinguish from benign processes, classified as embryonal, botryoid, alveolar, or pleomorphic,
such as nodular fascitis, when the presentation is truncal with spindle cell more recently described as a subtype of
or extremity. A neutrophilic infiltrate is generally noted the embryonal form.61,62 The pathology of these lesions is

Figure 39-3 Storiform pattern of malignant fibrous histiocytoma (250 ×).


Chapter 39 Retroperitoneal Tumors 657

associated with chromosomal abnormalities and the fusion there is usually a lag time of 5 months from initial symp-
transcripts associated with these translocations. toms to diagnosis. The principle clinical finding is abdom-
Embryonal rhabdomyosarcoma comprises more than inal mass and abdominal pain (60% to 80%).1 Many
half of all rhabdomyosarcomas and has a large array of patients also experience nausea and vomiting and weight
anatomic locations, including the genitourinary tract loss (20% to 30%). Neurologic findings are noted in 30%
and retroperitoneum. It can range from very well- of patients.66 Lower extremity edema is seen in 17% to
differentiated lesions to poorly differentiated lesions and 20% of patients, while urinary symptoms are surprisingly
express the multiple stages of muscle development. rare, seen in 3% to 5% of patients in most series.
Botryoid lesions are an anatomic variant of the embry- Physical examination will generally demonstrate a
onal type and are generally seen in hollow viscera. The protuberant abdomen that may be accompanied by an
spindle cell subtype is noted for its favorable clinical appearance of extremity wasting. Peripheral or inguinal
behavior. adenopathy may be present although lymphadenopathy
Alveolar rhabdomyosarcoma accounts for approxi- is not usually associated with these tumors (<5%).65
mately 20% of rhabdomyosarcomas and tend to occur Lower extremity edema and or increased abdominal wall
more frequently in the extremities. The tumor is venous markings suggest the presence of vena caval com-
arranged in aggregates of round or oval cells that create pression or obstruction.
irregular spaces reminiscent of alveoli. These tissues may Diagnostic imaging is imperative to delineate the
be surrounded by fibrous septae, and areas of necrosis are anatomic limits of the lesion and assess the integrity and
not uncommon. function of adjacent organs. This is most appropriately
Pleomorphic rhabdomyosarcoma comprise 5% of adult demonstrated with cross-sectional imaging68,69 (Figure
rhabdomyosarcomas and generally occur in older patients. 39-4). Important issues to address with this modality are
It generally occurs in the extremities and is similar to bilateral renal function, the presence or absence of vis-
MFH in appearance. The absence of cross-striations ceral metastases, and lymphadenopathy. Attention should
makes diagnosis difficult. With all rhabdomyosarcomas, also be given to the axial skeleton. Tumor involvement of
immunostaining for desmin, myoglobin, and muscle- neural foramina suggest unresectability of the lesion.
specific actin is useful. Germ cell tumors, which should be suspected in young
men and diagnosed through a thorough physical exami-
nation and testicular ultrasound, are usually distin-
Malignant Hemangiopericytoma
guished on computed tomography (CT) scan from other
These rare lesions originate from pericytes, which are primary retroperitoneal lesions. This is also generally
unique cells that arborize about small vessels and capillar- true for lymphomas. The CT scan, however, is not
ies. Their contractile properties allow them to control
microvascular flow and permeability. Less than 200 of
these lesions have been described, yet 25% of them have
occurred in the retroperitoneum or pelvis. They are usu-
ally well circumscribed, and if possible complete surgical
excision is warranted. Hemangiopericytomas display a rich
vascular pattern and stain positive for factor VIIIa. They
are negative for desmin and actin. The clinical behavior of
these lesions is difficult to predict, but metastases may
develop in 20% to 50% of cases. Those lesions with >4
mitoses per 10 HPF have a worse prognosis.64,65

DIAGNOSIS
Because of their slow growth and anatomic location,
retroperitoneal tumors (usually sarcomas) tend to grow to
a large size before they are detected. In a recent series of
MFHs, the average size at diagnosis for extremity lesions
was 5 cm, while retroperitoneal lesions were 16.5 cm in
size,27 thus such lesions can often be enormous by usual
pathologic standards. While the age at presentation is dis-
tributed across a large spectrum, the majority of patients Figure 39-4 CT image demonstrates massive right-sided
are diagnosed with retroperitoneal tumors in the sixth liposarcoma with suggestion of internal fibrous bands
decade of life. Since the progression of symptoms is slow, displacing the liver and right-sided retroperitoneal structures.
658 Part VI Testis

particularly diagnostic for the multiple tumor histologies by few mitoses, acellularity, minimal alterations of the
of primary retroperitoneal tumors besides a liposarcoma nucleus, few or absence of nucleoli. Scoring often takes
with high fat content. into consideration other factors, such as tumor size and
Earlier literature attests to the value of intravenous patient age. Grade 2 lesions display small to moderate
urography, upper and lower gastrointestinal (GI) series, size nucleoli, moderate nuclear pleomorphism, irregular
and angiography in defining displacement and delineat- chromatin distribution, and a moderate amount mitosis.
ing function. Advances in cross-sectional imaging have Areas of necrosis may be noted in the microscopic field.
largely supplanted this multistudy approach, and the Grade 3 lesions are characterized by numerous mitoses,
application of magnetic resonance arteriography allows gross chromatin clumping, moderate or considerable
for the delineation of vascular involvement by these necrosis, and variation in nuclear morphology. The dis-
uncommon lesions. From a practical standpoint, CT and tinction between T1 and T2 lesions is made at 5 cm.
magnetic resonance imaging (MRI) are complimentary Clinical staging beyond physical examination should
rather than redundant and should be employed as also include a bone scan and a dedicated chest CT, as well
needed.70 In some cases, imaging will not provide a rea- as an abdominal/retroperitoneal CT scan with attention to
sonable diagnosis; in that case, a biopsy prior to definitive the liver. Bilateral renal function must be determined pre-
resection may need to be performed. Classic references operatively. In experienced hands, cross-sectional scanning
suggest that percutaneous needle biopsies of these alone may provide enough local staging information. Since
retroperitoneal masses are generally unsatisfactory even these lesions are rare, however, any question regarding the
when aided with imaging techniques, such as ultrasound extent of contiguous organ involvement should be
or CT.21 More recent sources indicate that diagnosis may answered by the appropriate confirmatory imaging test
be a function of experience with these lesions and that (e.g., upper GI series). Laboratory tests may provide some
percutaneous biopsy or even needle aspiration may be diagnostic discrimination, yet generally add little to a stag-
quite satisfactory.71,72 Due to the uncommon nature of ing evaluation. Detailed evaluation of sarcomas can be
these lesions at most centers, it would seem appropriate found in the NCCN sarcoma practice guidelines.75
to obtain a correct tissue diagnosis with a reasonable tis-
sue sample when the diagnosis is in question.
SURGERY
In specific cases, an intraoperative biopsy may be indi-
cated to diagnose the lesion or differentiate it from a lym- Extirpative surgery is the principle and most effective
phoma or germ cell tumor. In those instances, care must form of therapy for primary retroperitoneal tumors.
be taken to obtain the sample from a solid part of the Preoperative planing to assess the extent of the disease is
lesion and employ meticulous technique to avoid any essential since successful surgery is defined by complete
tumor spillage. This is accomplished through appropriate excision of the mass with adequate margins of normal tis-
draping of the operative field, excellent hemostasis, and sue. A complete resection rate between 38% and 78%
meticulous closure with covering of the wound.21 (average 55%) is reported in many large series14–26,75,76
Laparoscopic sampling, while possible, would probably (Table 39-4). In several large series of retroperitoneal
not be feasible in most of these patients. Tumor specimens sarcomas, univariate and multivariate analyses of inde-
should be sent for frozen section and touch preparations pendent treatment variables have been performed. In
prepared to establish a diagnosis. In the appropriate over 280 patients, a complete resection was the single
patient, the serum tumor markers (beta-HCG and alpha most important positive predictive feature.77
fetoprotein) for germ cell tumors should be obtained pre- Intermediate or high tumor grade is a strong negative
operatively. There are no specific serum tumor markers feature.20,78–80 In most reported series, intermediate and
for primary retroperitoneal sarcomas, yet vanillylmandelic high-grade lesions comprise 55% to 70% of the tumors
acid (VMA) may be elevated in the rare case of gan- described. Tumor histology, tumor size, or patient age
glioneuroma or extra adrenal pheochromocytoma. were not significant factors in survival in multivariable
analysis.81,82 Therefore, carefully planned and skillfully
executed surgical therapy is critical for any chance at
STAGING
long-term success.83,84
The primary determinant of staging for primary
retroperitoneal tumors is the tumor grade. The grading
Operative Technique
is based on a G1 to G3 or G4 scale, and stage grouping
is done through the TNM system. Because of the impact The classic approach for surgery on these lesions is
of tumor grade this is often referred to as a GTMN sys- described through long midline incisions or chevron
tem (Table 39-3).72 incisions. The tumors are usually right-sided or left-
Tumor grading is determined by several factors, sided; however, few actually rising from the midline. A
including atypical mitosis, cytoplasmic and nuclear pleo- full flank position, such as that employed for renal stone
morphism, and necrosis.73,74 Grade 1 lesions are denoted surgery, is discouraged since it can limit exposure to the
Chapter 39 Retroperitoneal Tumors 659

full retroperitoneum or abdomen.1,21 Since these lesions the lateral borders of the great vessels extending dorsally
are not compartmentalized by the retroperitoneum, they between the spine and psoas and quadratus muscles
can cross many anatomic boundaries. It is important, (Figure 39-5). In this manner one can determine if the
therefore, to determine the potential for resectability, tumor has spread along a spinal nerve route and into the
beyond that presupposed through imaging. This is first spinal foramina or even further into the spinal cord. If
accomplished by developing a subadventitial plane along this is the case, the tumor is generally considered unre-

Table 39-3 American Joint Commission on Cancer


GTNM Classification and Stage Grouping of Soft
Tissue Sarcomas
Tumor grade

G1 Well differentiated

G2 Moderately differentiated

G3 Poorly differentiated

G4 Undifferentiated

Primary tumor

T1 Tumor ≤ 5 cm in greatest diameter

T1a Superficial tumor

T1b Deep tumor

T2 Tumor > 5 cm in greatest diameter

T2a Superficial tumor

T2b Deep tumor

Regional lymph node involvement

N0 No known metastasis

N1 Verified metastases to lymph nodes

Distant metastasis

M0 No known distant metastasis

M1 Known distant metastasis

Stage grouping

Stage 1A Low grade, small (G1-2, T1a or b, N0, M0)

Stage 1B Low grade, large, superficial (G1-2, T2a, N0, M0)

Stage IIA Low grade, large, deep (G1-2, T2b, N0, M0)

Stage IIB High grade, small (G3-4, T1b.N0, M0)

Stage IIC High grade, large, superficial G3-4, T2a, N0, M0

Stage III High grade, large, deep G3-4, T2b, N0, M0

Stage IV Nodal or distant metastases Any G, any T, N1, M0 or any G, any T, any N, M1

Continued
660 Part VI Testis

Table 39-3—cont’d
DEFINITIONS
Clinical Pathologic Primary Tumor (T)
Notes
■ ■ TX Primary tumor cannot be assessed 1. Superficial tumor is located
exclusively above the superficial fascia
■ ■ T0 No evidence of primary tumor without invasion of the fascia; deep
tumor is located either exclusively
■ ■ T1 Tumor 5 cm or less in greatest dimension beneath the superficial fascia, superficial
to the fascia with invasion of or through
■ ■ T1a superficial tumor(1) the fascia, or both superficial yet beneath
the fascia. Retroperitoneal, mediastinal,
■ ■ T1b deep tumor and pelvic sarcomas are classified as deep
tumors.
■ ■ T2 Tumor more than 5 cm in greatest dimension 2. Ewing’s sarcoma is classified as G4.

■ ■ T2a superficial tumor(1)

■ ■ T2b deep tumor

Regional lymph nodes (N)

■ ■ NX Regional lymph nodes cannot be assessed

■ ■ N0 No regional lymph node metastasis

■ ■ N1 Regional lymph node metastasis

Distant metastasis (M)

■ ■ MX Distant metastasis cannot be assessed

■ ■ M0 No distant metastasis

■ ■ M1 Distant metastasis

Biopsy of metastatic site performed ■ Y ■ N

Source of pathologic metastatic specimen __________________

Stage Grouping

■ ■ I T1a N0 M0 G1-2 G1 Low

T1b N0 M0 G1-2 G1 Low

T2a N0 M0 G1-2 G1 Low

T2b N0 M0 G1-2 G1 Low

■ ■ II T1a N0 M0 G3-4 G2-3 High

T1b N0 M0 G3-4 G2-3 High

T2a N0 M0 G3-4 G2-3 High

■ ■ III T2b N0 M0 G3-4 G2-3 High

■ ■ IV Any T N1 M0 Any G Any G High or low

Any T N0 M1 Any G Any G High or low


Chapter 39 Retroperitoneal Tumors 661

Table 39-3—cont’d
Histologic Grade (G) Notes

■ GX Grade cannot be assessed Additional Descriptors

Lymphatic vessel invasion (L)


■ G1 Well differentiated
LX Lymphatic vessel invasion cannot be assessed
■ G2 Moderately differentiated L0 No lymphatic vessel invasion
L1 Lymphatic vessel invasion
■ G3 Poorly differentiated
Venous Invasion (V)
■ G4 Poorly differentiated or
undifferentiated (four-tiered VX Venous invasion cannot be assessed
systems only)(2) V0 No venous invasion
V1 Microscopic venous invasion
V2 Macroscopic venous invasion
Residual Tumor (R)

■ RX Presence of residual tumor cannot be


assessed

■ R0 No residual tumor

■ R1 Microscopic residual tumor

■ R2 Macroscopic residual tumor

Table 39-4 Pooled Data on Complete Versus Partial Resection

No. of Resection (No. %) Mean Survival (Months) Complete Resection


Series Patients Complete Partial Complete Partial 5–Yr Survival (%)

Dalton et al.19 116 63/54 25/21 72 13 54

Jacques et al.20 86 43/50 34/39.5 65 28 74

McGrath et al.18 47 18/38 18/38 120 24 70

Glenn et al.22 50 37/74 8/16 40 — 38

Karakouis et al.24 68 27/40 7/10 84 48 64

Kilkeny et al.26 63 49/78 10/16 41 9 48

Zornig et al.23 51 30/59 21/41 60 — 35

TOTAL 481 267/55.5 123/25.6 70.3 24.4 54.7

sectable. It has also been suggested that the lateral inci- adequate negative margins. The most frequently resected
sion of the peritoneum into the deep body wall is impor- organs are listed in decreasing order in (Table 39-5). A
tant to explore on a level posterior to the spinous recent review reported that 20% of patients undergoing
processes (see Figure 39-5). If the operator’s fingers can surgery for a retroperitoneal sarcoma required nephrec-
be bi-manually palpated, this sarcoma is considered tomy. The majority of them (72%) underwent this dur-
resectable. ing their initial resection. The usual reason for
It is imperative in the preoperative period that both nephrectomy was total encasement of the organ, fol-
the patient and the surgeon realize the potential need for lowed by dense adherence to the kidney, and less often
en bloc resection of affected organs. This can include direct invasion of the renal unit.85 While a Dacron graft
vascular structures, as well as the kidney, portion of the can be employed to replace a portion of resected aorta,
diaphragm, liver, stomach, gall bladder, spleen, pancreas, there is debate among surgeons with regard to the need
and gut. In review of several series, up to 68% of opera- for venous reconstruction. Many feel that the venous col-
tions required resection of an adjacent organ to insure laterals that develop secondary to vena caval compression
662 Part VI Testis

Determination of resectability one to dissect those areas that are amenable to dissection,
Operative maneuvers and which in turn may free up a previously more con-
strained area of the operative field.86 Morbidity and mor-
tality rates for contemporary surgical series are
1 acceptable. An operative mortality of 2% to 7% with
2 morbidity rates of 6% to 25% is the norm. Hemorrhage,
IVC A
o
intra-abdominal abscess, and enterocutaneous fistula are
the most commonly described complications.
2
Another surgical technique is the modified thoracoab-
dominal approach, which has been developed and popu-
larized by Skinner. Those familiar with the technique feel
that it allows maximum exposure to the posterior
Psoas major muscle
Quadratus lumbarum muscle
retroperitoneum, which is often a difficult area to assess
Vertebra Spinal cord through a midline incision. Additionally, it provides
access and control to the ipsilateral great vessels above
Figure 39-5 Transabdominal approach to determine the diaphragm. Whether a left- or right-sided procedure
resectability of a primary retroperitoneal sarcoma. Pertinent
is performed, excellent exposure can be developed in the
maneuvers consist of intraabdominal assessment of visceral
contralateral retroperitoneal region without difficulty. A
and vascular contents and retroperitoneal assessment of
attachment to musculoskeletal and spinous structures. technique described here is a summary of Skinner’s
approach.87
With the thoracoabdominal approach, patient posi-
tioning is critical. The patient is not placed in a “pure
Table 39-5 Organs Sacrificed During Complete
flank” position, similar to that used in stone surgery.
Resection of Sarcoma
Rather, a modified flank position is employed. The con-
Organ Frequency of Resection (%) tralateral leg is flexed 90 degrees, and the hip is flexed
approximately 30 degrees. The ipsilateral shoulder and
Kidney 32–46
chest is placed 20 degrees off the horizontal with the arm
Colon 25 brought across the chest and placed in an adjustable
armrest. The pelvis is at most rotated 10 degrees off the
Adrenal gland 18 horizontal, and this position is maintained with a role
sheet. The table is fully hyperextended with the break
Pancreas 15
located above the iliac crest. The patient is secured with
Spleen 10 wide adhesive tape, and the ipsilateral leg is supported on
a pillow.
A mid-axillary incision is carried in the mid-axillary
line from the 8th, 9th, or 10th rib. The height of this
are adequate to allow for appropriate drainage from oth- incision is based on the size of the primary lesion. It
erwise vascularly compromised organs. When bowel via- extends over the rib and costochondral junction into the
bility is questionable, it should be resected if possible. epigastrium and then proceeds inferiorly as a midline
Although many visceral organs can be resected, unre- incision into the pelvis. The rib is resected subpe-
sectability is usually denoted by sarcomatosis, nerve root riosteally and the costochondral junction is divided. The
involvement, pelvic sidewall involvement, malignant rectus muscle is divided and retracted laterally. In most
ascites, or the presence of distant metastasis.1,21 instances, the peritoneum will be opened, and the bowel
When defining the operative field it is often necessary contents mobilized superiorly. This mobilization is based
to release multiple intraabdominal adhesions. This must on the superior mesenteric artery pedicle. It is important
be performed meticulously since an enterotomy and sub- that this artery be identified initially, and care is taken not
sequent fistula formation can cause major morbidity. to traumatize it. In many instances, the inferior mesen-
Tactile, as well as visual perceptions, aids in avoiding vio- teric artery may have to be resected. Most care should be
lation of the bowel. In addition, the use of sharp curved taken to maintain the marginal artery. In most instances,
Mayo scissors rather than Metzenbaum scissors may aid a large bowel section will be avoided, yet if there is a
in avoiding this complication. During the dissection of a question of viability at the end of the case, any suspect
large tumor mass, it is often necessary to shift the loca- area of bowel should be resected. Vena cava obstruction
tion of the dissection when one particular area becomes may be associated with any right-sided lesions, and if this
difficult due to concerns of a clear margin or the poten- is the case, it is usually best to resect the vena cava en bloc
tial for vascular damage. A centripetal dissection allows with removal of the tumor. Often, a right nephrectomy
Chapter 39 Retroperitoneal Tumors 663

may also be required. If this maneuver is performed, it is Proportion Surviving


best to maintain vascular connection between the vena 1.0
cava and left renal vein to decrease the possibility of acute 81
0.8
or long-term renal insufficiency.
In general, the aorta can be dissected free of large 0.6
54
retroperitoneal tumors but rarely may require replace- 45
ment with a Dacron graft. During dissection or mobi- 0.4 34
lization of the great vessels, care must be taken to control 0.2
17
the lumbar vessels with ligation and division to avoid 8
problematic bleeding. While the distal vessels may be 0.0
controlled with hemoclips, it is appropriate to ligate the 0 2 5 10
lumbar vessel at its origin on its great vessel. In the case Years
of significant bleeding, the vessel tear can be controlled Complete Resection Incomplete Resection
with the use of a Judd or Allis clamp. While the possibil- Figure 39-6 Patient survival as a function of completeness of
ity of spinal devascularization exists, it is very uncommon the surgical resection. The data represent x complete
due to the significant collateral blood supply to the spine resections and y incomplete resections from collected series.
from the artery of the Adamkiewicz. In younger patients,
the potential for ejaculatory dysfunction is significant.
Reoperative surgery for the treatment of recurrent
retroperitoneal sarcoma can be of value. Over 60% of
SURGICAL OUTCOME
patients may be experienced a complete resection.88 In
The classic overall survival for patients presenting with one series, 30 patients with a previous complete resection
retroperitoneal sarcomas is poor. In a multiinstitutional of the primary lesion experienced local tumor recurrence
review, the 2-, 5-, and 10-year mean survivals for patients at a mean interval of 23 months. Sixty percent of these
with this disease were 56%, 34%, and 18%, respec- patients were rendered free of disease after reoperative
tively.21 Recent series suggest 2-year survivals of over surgery. To accomplish this, 33% of these patients
70% and 5-year survivals of 50% to 60% in patients required the resection of adjacent viscera. Those patients
without metastases.88,89 The effective surgical manage- who achieved a complete resection with a second opera-
ment of retroperitoneal tumors has been limited by their tion had a 33-month median survival compared to a 14-
size at presentation that often results in secondary organ month median survival in those patients who did not
involvement. This has a poor impact on the ability to experience a second complete resection.92
achieve negative surgical margins even with the resection The role of subtotal resection has never been clearly
of adjacent organs. Historically, a complete surgical established, yet some data suggest that this may have
resection with negative margins was achieved in 50% to some positive value compared to palliative or debulking
60% of patients. The more recent series demonstrate surgery. The argument for the possible value of incom-
complete resection rates in the 60% to 80% range, which plete resection has been made the management of
may be due to improvements in imaging, preoperative liposarcomas. In a series of 55 patients, 75% received
planning, and surgical technique. symptom relief from their surgery and partial resection
Those patients who achieve a complete resection dis- compared with biopsy or exploration alone had a signifi-
play superior survival to those patients with incomplete cant impact on increased survival (26 months versus 4
resection of their tumors (see Table 39-4). This is also months).93 In another report, 22 patients with complete
supported in laboratory models.90 In an analysis of resection of tumor displayed a similar median survival to
microscopic margins, however, positive margins at this 15 patients with subtotal resection (median survival >120
level of pathologic resolution had little impact on recur- months) compared to those only having a partial pallia-
rence-free survival.91 tive resection or exploration (12 to 20 months).94 An
On the average, the 5-year survival of patients with aggressive surgical approach in cases where near com-
completely resected tumors is 54% compared to 17% for plete resection can be obtained has also been advocated
incomplete resections at 10 years, this difference is 45% by others.24
to 17%. Thus, a nearly 40% survival advantage is seen at There is no set protocol for the postoperative monitor-
5 to 10 years in those patients with complete surgical ing of patients with primary retroperitoneal sarcoma, but
resection of their lesion (Figure 39-6). Tumor recurrence recommendations can be made given the rapidity of recur-
after complete surgical resection, however, is significant. rence, the advantage of reresection, and the lethality of
There is a 72% chance of local recurrence at 5 years and this condition. It would be appropriate to perform an
a 90% chance of recurrence at 10 years in most series. abdominal CT scan, chest x-ray, and biochemistry profile
These data imply fairly poor survival at 15 years.21 with complete blood count (CBC) every 6 months. A
664 Part VI Testis

Proportion Surviving series. It is associated with a lower rate of local recur-


1.0 rences and fewer GI complications, yet peripheral neu-
83 ropathies are more frequent, and it is difficult to
0.8 74 demonstrate a survival advantage with its use.100–104
0.6 Combined modality therapy with external beam and
54 42 radiation with surgery and IORT (15 Gy) is being
0.4 explored. It appears that such intensive regimens are tol-
0.2 24 erable up to 50.4 Gy of EBRT.105 Additionally, the use of
brachytherapy postoperatively in primary or recurrent
11
0.0 disease has been explored using different techniques up
0 2 5 10 to 32 Gy.106,107 These techniques are feasible yet signifi-
Years cant side effects can be encountered when they are
G 2,3 Tumors G 1 Tumors applied to the upper abdomen. Newer radiation delivery
Figure 39-7 Patients’ survival as a function of tumor grade in techniques, such as intensity modulated radiation therapy
cases of primary retroperitoneal sarcoma. The data represent (IMRT), are being explored for the treatment of these
50 low-grade and 80 high-grade tumors from collected lesions.108,109
series.
CHEMOTHERAPY
lengthening of this follow-up interval should be consid- Multimodal therapy for extremity sarcoma has provided
ered only after 5 years. Data support the concept that close an inference for the role of chemotherapy in retroperi-
follow-up allows for early detection less bulky recurrences toneal lesions. The majority of large-scale chemotherapy
that allow for more successful salvage surgery.95 trials, however, comprise patients with extremity disease,
The effect of tumor grade on patient survival cannot so these findings may not be directly applicable. Overall,
be underestimated. Low-grade lesions display a 50% sur- the argument for adjuvant therapy seems reasonable
vival advantage over intermediate- and high-grade since the potential for distant recurrence is significant
lesions at 5 years (74% to 24%) and a 30% survival at 10 with retroperitoneal lesions. The data in this regard have
years (42% to 11%) (Figure 39-7). This is demonstrated been somewhat contradictory, yet a metaanalysis of these
even in the face of total surgical resection. While aggres- studies suggests a slight advantage for the use of adjuvant
sive surgical resection of retroperitoneal sarcoma serves doxorubicin therapy to decrease the risk of death and
as the foundation of therapy for this condition, the high recurrence in patients with high-grade extremity
local recurrence rate and eventual mortality from this lesions.99 Such therapy is associated with toxicity, how-
disease has prompted the exploration of adjuvant thera- ever, and has not gained considerable acceptance for
peutic modalities. retroperitoneal lesions.110

RADIATION THERAPY Metastatic Disease


The role of radiation therapy in the treatment of soft Approximately 20% of patients with soft tissue retroperi-
tissue sarcomas has been established by the advances in toneal sarcoma will present with metastatic disease.
extremity lesions. Unfortunately, the radiosensitivity of Those patients who recur after treatment for localized
adjacent soft visceral organs in the retroperitoneum has disease generally do so by 60 months.111,112 Those
limited the direct transfer of such techniques to lesions patients who recur demonstrate disease at multiple sites
in this region.96,97 While extremity tumors are treated (47%) or display a local recurrence (30%). Isolated pul-
in the dose range of 60 to 64 Gy, most series of monary lesions occur in approximately 20% of patients.
retroperitoneal lesions are treated in the 40- to 55-Gy Three-year survival in patients undergoing complete
range. excision of lung-only lesions is 38%.113
In general, one sees a decrease in local recurrences Doxorubicin is the foundation for chemotherapy in
compared to historic controls at the cost of increased gas- advanced sarcoma.114,115 The general response rate is
trointestinal complications (nausea, vomiting, and enteri- 20% to 25%, but sustained complete responses are
tis). It is difficult to demonstrate a significant uncommon. Other classic agents demonstrating activity
improvement in survival, and on multivariable analysis include dacarbazine, ifosfamide, methotrexate, and
the completeness of the surgical resection and the grade cyclophosphamide. In several phase III studies, however,
of the tumor have the most significant bearing on out- combination treatments were not superior to single-
comes.98–100 Intraoperative radiation therapy (IORT) has agent doxorubicin.116–118 Other phase III studies have
been employed as a surgical adjuvant in several small not demonstrated superior response, yet they do show
Chapter 39 Retroperitoneal Tumors 665

significant increased toxicity (myelosuppression and car- Table 39-6 Common Genitourinary Sarcomas in
diac) with the addition of other agents.119 Another com- Decreasing Order of Frequency
bination regimen of mesna, adriamycin (doxorubicin), Renal
ifosfamide, and dacarbazine (MAID) has been successful
in neoadjuvant programs for extremity sarcomas com- Leiomyosarcoma
pared to historic controls, yet less data are available with
regard to other soft tissue sites.120,121 In general, salvage Liposarcoma
therapy for patients has been associated with poor Malignant fibrous histiocytoma
responses and little in the way of quality of life advan-
tages.121 Hemangiopericytoma
More recently, gemcitabine has been investigated as
an initial agent in advanced disease and for salvage ther- Rhabdomyosarcoma
apy. As a single agent the results have been disappointing Osteogenic sarcoma
with response rates below 5% in either setting.122–124
The combination of gemcitabine and docetaxel in Bladder
patients with unresectable leiomyosarcoma, however,
demonstrated a 10% complete response and an overall Leiomyosarcoma
response of 53%. It was felt to be a tolerable and active
Rhabdomyosarcoma
combination in treated and untreated patients.125
Disseminated intraperitoneal spread of disease is very Osteogenic sarcoma
difficult to treat. A novel approach is the application of
photodynamic therapy.126 In a preliminary report, Liposarcoma
patients underwent surgical debulking and were treated
Malignant fibrous histiocytoma
with photofrin and laser light. Of initial 11 patients, 5
demonstrated no evidence of disease over a range of 2 to Fibrosarcoma
17 months. The advancement of such technologies and
the implementation of small molecule therapy may pro- Prostate
vide improved outcomes for such patients in the future.
Leiomyosarcoma

ADULT URINARY TRACT SARCOMA Rhabdomyosarcoma

Primary sarcomas arising from the genitourinary system Fibrosarcoma


are extremely rare lesions overall and comprise only 1%
Spindle cell sarcoma
to 2% of urologic tumors in adults. Only 800 to 1000 such
tumors have been described from multiple genitourinary Spermatic cord, testis, paratestis
sites. Although many small clusters go unreported, prac-
tice principles in the diagnosis and treatment of these Leiomyosarcoma
tumors must therefore be derived from anecdotal pooled
Rhabdomyosarcoma
data.127,128 General treatment principles are also extrapo-
lated and applied from the clinical experience with pri- Liposarcoma
mary retroperitoneal sarcomas. These lesions behave
differently from pediatric genitourinary sarcomas, Fibrosarcoma
notably pediatric rhabdomyosarcoma, which is discussed
Malignant fibrous histiocytoma
separately in Chapter 49. Thus, treatment strategies,
especially for advanced disease, tend to be empiric.
Despite their rarity, the collective data regarding sar-
RENAL SARCOMA
comas of the genitourinary tract provide significant data
with regard to clinical presentation and clinical outcome Approximately 1% to 2% of kidney tumors are true renal
that provide rough guidelines for treatment when these sarcomas. This excludes the sarcomatoid variant of renal
uncommon lesions are encountered. The most common cell carcinoma (RCC) that demonstrates spindle cells or
sarcomatous lesions of the urinary system originate in the giant cells and a rapidly progressive clinical course. The
spermatic cord and paratesticular structures, which are most frequent histologic subtypes of sarcoma encoun-
not strictly speaking retroperitoneal. In decreasing inci- tered in the kidney are listed in Table 39-6.
dence, sarcomas of the kidney, the bladder, and the Leiomyosarcoma is the most common renal sarcoma,
prostate are encountered (Table 39-6). comprising 30% to 40% of most reported series.
666 Part VI Testis

Liposarcoma and MFH are then encountered in near mas. Liposarcoma, MFH, and osteosarcoma account for
equal frequency. Other histologies include rhabdosar- <5% each of bladder sarcoma.136,137 A recent report also
coma, osteosarcoma, and hemangiopericytoma.129–132 collected a cohort of 10 angiosarcomas the majority of
Renal sarcoma usually presents at a slightly younger age which responded poorly to therapy (Engel). The devel-
than the average patient with RCC with pain and flank opment of some bladder sarcomas has been attributed to
mass as the common symptoms. Hematuria is seen less the secondary effects of radiation therapy or chemother-
frequently with classic RCC. They have no distinguish- apy exposure.138,139
ing imaging characteristics but tend to be hypervascular. The treatment of bladder sarcoma results in excep-
When small, they are hard to distinguish from RCC, but tionally good outcomes for a tumor type generally
hypovascularity combined with contiguous tumor spread expected to perform poorly. Cystectomy and diversion in
may suggest sarcoma. combination with other treatment modalities can result
Treatment for renal sarcoma is similar to RCC, in 5-year disease-free survivals of 62%.140 Smaller,
namely, wide surgical excision. The principles for a stan- selected cystectomy series have demonstrated similar
dard radical nephrectomy conform to those for sarcoma findings.136 Significant long-term disease-free survival
surgery, and the modified thoracoabdominal approach is rates have also been achieved with less aggressive surgery.
particularly suited to large lesions. As is the case in all soft In an earlier series from Memorial Sloan Kettering
tissue sarcomas, tumor size reflected in complete Cancer Center, 14 of 15 patients with resectable bladder
resectability, and grades are the major determinants of tumors displayed no evidence of disease 1 to 9 years after
clinical outcome. There are no data to support surgery, this included 4 patients who underwent partial
chemotherapy or radiation in the adjuvant setting, and it cystectomy.127 In a more recent series from the same
is unlikely that these modalities will confer a survival institution, 7 of 10 patients with bladder sarcoma are
benefit for patients with advanced disease. The overall 5- alive without evidence of disease with over 5 years of fol-
year survival for these patients is approximately 30%. low-up, including 2 patients treated only with
Those individuals with liposarcoma or hemangiopericy- transurethral resection of 2 cm leiomyosarcomas of the
toma perform much better than patients with bladder.131 Other series have demonstrated that low-
leiomyosarcoma. The survival rate at 5 years is in the grade lesions can respond reasonably well to organ-spar-
80% to 90% range for liposarcoma yet <50% for ing therapy.141 These data suggest that bladder
leiomyosarcoma. This more than likely a reflection of preservation is a reasonable option in the treatment of
tumor grade at presentation.127,128,131 this disease especially in the case of leiomyosarcoma.
Patients with renal sarcoma should adhere to close Partial cystectomy is a reasonable option if wide (3 to 4
postoperative follow-up, including more frequent evalu- cm) margins can be obtained. Involvement of the trigone
ation of the renal bed, since local recurrence is the rule or more distal structures, however, would suggest total
rather than the exception with sarcomas. Therapy for cystectomy and continent reconstruction as an appropri-
advanced disease usually consists of single agent doxoru- ate option. Although the data are anecdotal, the use of
bicin-based chemotherapy or multiagent doxorubicin- chemotherapy and external beam radiation may be
based chemotherapy. While responses may be seen, appropriate in patients with bladder sarcoma at high risk
sustained complete responses are not reported. for local or distant recurrence. Overall 5-year survival for
patients with bladder sarcomas is 60%. Those patients
with adult rhabdomyosarcoma of the bladder have a
URINARY BLADDER
poorer prognosis than their pediatric counterparts and
In the adult, primary urinary bladder sarcomas are very adults with leiomyosarcoma. The 5-year survival for
rare with roughly 200 cases reported in the medical liter- patients with adult rhabdomyosarcoma is approximately
ature.127,128 They comprise approximately 0.1% to 0.2% 30%. Angio sarcoma is a very rare bladder lesion (10
of all primary bladder tumors, yet present in a manner reported cases) that is highly progressive. It responds best
similar to typical bladder tumors. Most patients display to multimodal oncologic therapy.142
hematuria, dysuria, or urinary frequency as the present-
ing signs and symptoms of their lesions. On imaging
PROSTATE
studies they can display upper urinary tract obstruction
or filling defects of the bladder.131–135 The diagnosis can Approximately 150 reported cases of primary sarcoma of
usually be made by transurethral resection of the bladder the prostate have been reported.127,128,135,143,144 Given the
tumor. Pelvic and abdominal imaging with CT or MRI is estimated yearly incidence of prostate cancer at over
useful in perioperative staging. The most common tumor 220,000 cases, these lesions comprise <0.1% of primary
histology in adult bladder sarcoma is leiomyosarcoma, prostate tumors. The majority of prostate sarcomas is
followed by rhabdomyosarcoma. These lesions of muscle either leiomyosarcoma (50%) or rhabdomyosarcoma
origin comprise 70% to 80% of all adult bladder sarco- (30%). The remainders of tumors are distributed over
Chapter 39 Retroperitoneal Tumors 667

multiple tissue types. In general, patients with prostate sar- older age groups and can occur anywhere along the
coma tend to present somewhat younger than the typical inguinal to paratesticular region and are often found in
patient with adenocarcinoma of the prostate. The most the inguinal region. It is unusual to have a sarcoma in
common presenting symptom is bladder outlet obstruc- the testicular parenchyma.
tion and dysuria. A combination of symptoms and a palpa- Patients with paratesticular sarcomas have a firm pal-
ble mass (often smooth) on physical examination lead to pable mass in the scrotum or the spermatic cord. Any
endoscopic evaluation and needle biopsy diagnosis. The lesions of the scrotal contents should be evaluated with
majority of patients present with regional or local disease, ultrasound and all solid lesions should be classically
while 10% to 20% of patients may have metastases. treated with inguinal exploration biopsy and radical
Cystoprostatectomy has generally been the treatment orchiectomy if indicated. Transscrotal exploration should
of choice for these tumors since most are large at the time not be performed. Further therapy for these is dictated
of diagnosis, also because usually defined tissue planes by tissue histology. Postoperative radiation therapy can
may be obliterated by these tumors. In the case of exten- be administered especially if it appears that one has not
sive retroperitoneal disease it may be preferable to per- achieved negative margins after the initial surgery.149
form a total pelvic exenteration to achieve complete local Rhabdomyosarcoma and leiomyosarcoma generally
control. Since the potential for total continent recon- perform poorly. It is necessary to assess these patients for
struction of both the urinary and lower GI tract exists, the presence of metastatic disease, particularly retroperi-
this aggressive approach is a more favorable option than toneal lymph node involvement. In the absence of gross
double ostomies or palliative therapy.145 As in the case of disease, a retroperitoneal lymph node dissection is appro-
bladder sarcoma, a pelvic lymph node dissection should priate, especially in the case of rhabdomyosarcoma where
accompany the exenterative procedure since nodal the incidence of positive retroperitoneal nodes may be
involvement, especially in rhabdomyosarcoma, can exist. >50%.149–151 It has also been advocated for MFH and
In general, patients with prostate sarcoma have an fibrosarcomas of the cord. If gross nodal or distant metasta-
unfavorable outcome. The average 5-year survival is tic disease is detected, doxorubicin-based chemotherapy is
approximately 25%. Patients with leiomyosarcoma have the classic treatment for advanced disease and is generally
a 40% 5-year survival, while those with rhabdomyosar- employed in the presence of microscopic metastasis. Those
coma have a 0% to 10% 5-year survival. A recent series patients with liposarcoma generally show nearly complete
of these lesions demonstrated a 3- and 5-year survival of cure through local excision.151 Ninety percent of patients
43% and 38%. The factors most associated with a good may be treated in this fashion, but local recurrence have
outcome were the absence of metastatic disease at pres- been reported and can result in fatalities.
entation and the finding of negative surgical margins.146 True primary sarcoma of the testis is exceedingly rare
It is difficult to assess the value of combined adjuvant with only 16 cases reported in the literature the mean
therapy, but in the case of high-grade or extensive dis- patient age was 32 with a range from 3 to 86. The major-
ease, a doxorubicin-based protocol and the addition of ity of lesions were leiomyosarcomas, and spindle cell sar-
external beam radiation would be appropriate. comas were the next most frequent histology.85
Carcinosarcoma is a very rare lesion. It is a malignant
mixed tumor with sarcomatous and carcinomatous com-
PARATESTICULAR TUMORS
ponents. There are <100 reported cases, and the majority
The most common primary genitourinary sarcomas are of these affect the urinary bladder.152,153 It appears that
those of the paratestis, spermatic cord, and testis these lesions have a clonal origin similar to bladder
proper.127,128,131,147,148 Almost 300 cases of such lesions tumors but that additional unique changes lead to this
have been reported. The majority of these lesions from aggressive phenotype.154 This has been reported in the
muscle origin and are either rhabdomyosarcoma or prostate and kidney. Patient outcomes are uniformly
leiomyosarcoma (50% of tumors). Liposarcomas and poor, and treatment for this disease beyond radical sur-
fibrosarcomas are commonly seen, along with a distribu- gery is anecdotal.
tion of other rare lesions. The most common benign
tumor of the spermatic cord, however, is a lipoma, while
SUMMARY
the most common benign lesion of the paratesticular
region is an adenomatoid tumor. Genitourinary sarcomas are exceedingly rare lesions,
These lesions segregate into an adolescent-early which are not amenable to controlled clinical trials.
adult and older age group (over 50) with the majority of Precise standards for clinical care are therefore difficult
rhabdomyosarcomas concentrated in the younger to establish. While the management of renal sarcomas
patients. Rhabdomyosarcomas also have a greater differs little from the classic management of renal
predilection for occurring in an intrascrotal area. tumors, it is interesting to note that bladder sarcoma is a
Liposarcoma and leiomyosarcoma generally occur in very treatable disease, in which cure may be obtained in
668 Part VI Testis

many cases without the loss of function. Prostate sarco- 16. Coran AG, Crocker DW, Wilson RE: A twenty-five year
mas are best treated with very aggressive surgery, and the experience with soft tissue sarcomas. Am J Surg 1970;
outcome of paratesticular sarcomas is very much affected 119:288.
by primary histology. In urologic sarcomas, there appears 17. Oriana S, Bonardi P, Preda F: Primary retroperitoneal
tumors. Tumori 1977; 63:397.
to be a role for lymph node dissections in conjunction
18. McGrath PC, Neifeld JP, Lawrence W Jr, et al: Improved
with primary therapy, which is different than the clinical
survival following complete excision of retroperitoneal
recommendations for primary retroperitoneal sarcomas. sarcomas. Ann Surg 1984; 200:200.
Adult rhabdomyosarcoma generally responds less well 19. Dalton RR, Donohue JH, Mucha P, et al: Management of
than its pediatric counterpart even when successful pedi- retroperitoneal sarcomas. Surgery 1989; 106:725.
atric, therapeutic protocols are employed. In all instances 20. Jacques DP, Coit DG, Hajdu SI, Brennan MF:
of genitourinary sarcoma, the overall lack of sustained Management of primary and recurrent soft-tissue sarcoma
responses to doxorubicin-based therapy suggests that of the retroperitoneum. Ann Surg 1990; 212:51.
patients with advanced disease should be enrolled in 21. Storm FK, Mahvi DM: Diagnosis and management of
novel protocols. retroperitoneal soft-tissue sarcoma. Ann Surg 1991; 214:2.
22. Glenn J, Sindelar WF, Kinsella T, et al: Results of
multimodality therapy of resectable soft-tissue sarcomas of
the retroperitoneum. Surgery 1985; 97:316.
REFERENCES 23. Zornig C, Weh HJ, Krull A, et al: Retroperitoneal sarcoma
in a series of 51 adults. Eur J Surg Oncol 1992; 18:475.
1. McGrath PC: Retroperitoneal sarcomas. Semin Surg 24. Karakousis CP, Velez AF, Emrich LJ: Management of
Oncol 1994; 10:364. retroperitoneal sarcomas and patient survival. Am J Surg
2. Jemal A, Thomas A, Murray T, Thun M: Cancer 1985; 150:376.
statistics, 2002. CA Cancer J Clin 2002; 52:23–47. 25. Rossi CR, Nitti M, Foletto S, et al: Management of
3. Mack TM: Sarcomas and other malignancies of soft primary sarcomas of the retroperitoneum. Eur J Surg
tissue, retroperitoneum, peritoneum, pleura, heart, Oncol 1993; 19:355.
mediastinum, and spleen. Cancer 1995; 75:211. 26. Kilkenny JW III, Bland KI, Copeland EM, et al:
4. Pack GT, Tabah EJ: Primary retroperitoneal tumors: a Retroperitoneal sarcoma: the University of Florida
study of 120 cases. Surg Gynecol Obstet 1954; 99: experience. J Am Coll Surg 1996;182: 329.
209–231, 313–341 (International Abstracts of Surgery). 27. Rooser B, et al: Malignant fibrous histiocytoma of soft
5. Woodruff JM: Peripheral nerve tumors showing glandular tissue. Cancer 1991; 67:499.
differentiation (glandular schwannoma). Cancer 1976; 32: 28. Pezzi CM, Rawlings MS, Esgro JJ, et al: Prognostic
2399. factors in 227 patients with malignant fibrous
6. Arlen M, Higinbotham NL, Huvos AG, et al: Radiation- histiocytoma. Cancer 1992; 69:2098.
induced sarcoma of bone. Cancer 1971; 28:1087. 29. Crozat A, et al: Fusion of CHOP to a novel RNA-binding
7. Bailar JC: How dangerous is dioxin? N Engl J Med 1991; protein in human myxoid liposarcoma. Nature 1993;
324:260. 363:640.
8. Laskin WB, Silverman TA, Enzinger FM: Postradiation 30. Ladanyi M: The emerging molecular genetics of sarcoma
soft tissue sarcomas: an analysis of 53 cases. Cancer 1988; translocations. Diagn Mol Pathol 1995; 4(3):162.
62:2330. 31. Panagopoulos I, Hoglund M, Mertcus F, et al: Fusion of
9. Hardell L, Sandstrom A: Case-control study: soft tissue the EWS and CHOP genes in Myxoid lipo sarcoma.
sarcoma and exposure to phenoxyacetic acids or Oncogene 1996; 12:489–496.
chlorophenols. Br J Cancer 1979; 39:711. 32. Berner JM, Forus A, Elkakoun A, et al: Separate amplified
10. Zahn SH, Fraumeni JF, Semin J: The epidemiology of regions encompassing CDK4 and MDM2 in human
soft tissue sarcomas. Oncology 1997; 24:504–513. sarcomas. Genes Chrom Cancer 1996; 17:254.
11. Greenwald P, Kovasznay B, Collins DN, et al: Sarcomas 33. Elkahloun AG, Bittner M, Heskius K, et al: Molecular
of soft tissues after Vietnam service. JNCI 1984; cytogenetic characterization and physical morphology of
73:1107. Rg-13-15 amplification in human cancers. Genes
12. Suruda AJ, Ward EM, Fingerhut MA: Identification of Chromosomes Cancer 1996; 17(4):205–214.
soft tissue sarcoma deaths in cohorts exposed to dioxin 34. Pilotti S, Della Torre G, Lavarino C, Sozzi G, et al:
and to chlorinated naphthalenes. Epidemiology 1993; Molecular abnormalities in liposarcoma: role of MDM2
4:14. and CDK4-containing amplicons at 12q13-22. J Pathol
13. Economou JS, Sondak VK, Eilber FR: General 1998; 185:188–190.
considerations, In Eilber FR (ed): The Soft Tissue 35. DeWeerd JH, Dockerty MB: Lipomatous retroperitoneal
Sarcomas, p 3. Orlando, Grune and Stratton, 1987. tumors. Am J Surg 1952; 84:397.
14. Bose B: Primary malignant retroperitoneal tumors: 36. Mowat AP, Clark CG: Presacral lipomata. Br J Surg 1961;
analysis of 30 cases. Can J Surg 1979; 22:215. 49:230.
15. Armstrong JR, Cohn I Jr: Primary malignant 37. Heyns CF: Pelvic lipomatosis: a review of its diagnosis
retroperitoneal tumors. Am J Surg 1965; 110:937. and management. J Urol 1991; 146:267.
Chapter 39 Retroperitoneal Tumors 669

38. Yalla SV, Ivker M, Burros HM, et al: Cystitis glandularis 57. Weiss SW: Malignant fibrous histiocytoma: a
with perivesical lipomatosis: frequent association of two reaffirmation. Am J Surg Pathol 1982; 6:773.
unusual proliferative conditions. Urology 1975; 5:383. 58. Coindre JM, Mariani O, Chibbon F, Mairal A, et al: Most
39. Klein FA, Vernon-Smith MJ, Kasenetz I, et al: Pelvic malignant fibrous histiocytomas developed in the
lipomatosis: 35 year experience. J Urol 1988; 139:998. retroperitoneum are dedifferentiated liposarcomas: a
40. Andac N, Baltacioglu F, Cimsit NC, Tuney D, et al: Fat review of cases initially diagnosed as malignant fibrous
necrosis mimicking liposarcoma in a patient with pelvic histiocytoma 2003; 16:256–262.
lipomatosis. CT findings. J Urol 2003; 27:109–111. 59. Costa MJ, Weiss SW: Angiomatoid malignant fibrous
41. Sanders R, Nabil B, Curry N, et al: Clinical spectrum of histiocytoma: a follow-up study of 108 cases with
adrenal myelolipoma: analysis of 8 tumors in 7 patients. evaluation of possible histologic predictors of outcome.
J Urol 1995; 153:1791. Am J Surg Pathol 1990; 14:1126.
42. Chenkt F: Extra-adrenal myelolipoma. Am J Clin Pathol 60. Bizer LS: Fibrosarcoma: report of 64 cases. Am J Surg
1982; 78:386. 1971; 121:586.
43. Fowler MR, Williams RB, Alba JM, et al: Extra-adrenal 61. Horn RC, Enterline HT: Rhabdomyosarcoma: a
myelolipomas compared with extramedullary clinicopathological study of 39 cases. Cancer 1958;
hematopoietic tumors: a case of presacral myelolipoma. 11:181.
Am J Surg Pathol 1992; 6:363. 62. Cavazzana AO, Schmidt D, Ninfo V, et al: Spindle cell
44. Clement PH: Intravenous leiomyomatosis of the uterus. rhabdomyosarcoma: a prognostically favorable variant of
Pathol Annul 1988; 23:153. rhabdomyosarcoma. Am J Surg Pathol 1992; 16:229.
45. Kindblom LG, Angervall L, Svendsen P: Liposarcoma: a 63. Enzinger FM, Smith BH: Hemangiopericytoma: an
clinicopathologic, radiographic and prognostic study. Acta analysis of 106 cases. Hum Pathol 1976; 7:61.
Pathol Microbiol Scand 1975; 253:1. 64. Zirkin RM: Retroperitoneal hemangiopericytoma. Int
46. Spittle MF, Newton KA, Mackenzie DH: Liposarcoma: a Surg 1977; 62:395.
review of 60 cases. Br J Cancer 1971; 24:696. 65. Douglas JL, Mitchell R, Short DW: Retroperitoneal
47. Weiss SW, Rao VK: Well-differentiated liposarcoma hemangiopericytoma. Br J Surg 1966; 53:31.
(atypical lipoma) of deep soft tissue of the extremities, 66. Cohan RH, Baker ME, Cooper C, et al: Computed
retroperitoneum and miscellaneous sites: a follow-up tomography of primary retroperitoneal malignancies.
study of 92 cases with analysis of the incidence of J Comput Assit Tomogr 1988; 12:804.
“dedifferentiation”. Am J Surg Pathol 1992; 16:1051. 67. Fong Y, Coit DG, Woodruff JM, Brennan MF: Lymph
48. Enterline HT, Culberson JD, Rochlin DB, et al: node metastasis from soft tissue sarcoma in adults: analysis
Liposarcoma. A clinical pathological study of 53 cases. of data from a prospective database of 1772 sarcoma
Cancer 1960; 13:932–950. patients. Ann Surg 1993; 217:72.
49. Forus A, Larramendy ML, Meza-Zepeda LA, Bjerkehagen 68. Neifeld JP, Walsh JW, Lawrence W Jr: Computed
B, et al: Dedifferentiation of a well-differentiated tomography in the management of soft tissue tumors.
liposarcoma to a highly malignant metastatic Surg Gynecol Obstet 1982; 155:535.
osteosarcoma: amplification of 12q14 at al stages and gain 69. Chang AE, Matory YL, Dwyer AJ, et al: Magnetic
of 1q22-q24 associated with metastases. Cancer Genet resonance imaging versus computed tomography in the
Cytogenet 2001; 125:100–111. evaluation of soft tissue tumors of the extremities. Ann
50. Pilotti S, Della Torre G, Lavarino C, Sozzi G, et al: Surg 1987; 205:340.
Molecular abnormalities in liposarcoma: role of MDM2 70. Bland KI, McCoy DM, Kinard RE, et al: Application of
and CDK4-containing amplicons at 12q13-22. J Pathol magnetic resonance imaging and computerized
1998; 185:188–190. tomography as an adjunct to the surgical management of
51. Dalcin P, Sciot R, Panagopoulouz I, et al: Additional soft tissue sarcomas. Ann Surg 1987; 205:473.
evidence of a t(R:22) with EWS/CHOP fusion in myxoid 71. Barth RJ, Merino MJ, Solomon, et al: A prospective study
liposarcoma. J. Path 1997; 182:437. of the value of core needle biopsy and fine needle
52. Deitos AP, Dogliani C, Piccinin S, et al: Molecular aspiration in the diagnosis of soft tissue masses. Surgery
abnormalities of the p53 pathway in dedifferentiated 1992; 112:536.
liposarcoma. J Pathol 1997; 181:8. 72. Lopez-Rios F, Perez-Barrios A, Alberti N, Vargas J, et al:
53. Shmookler BM, Lauer DH: Retroperitoneal Fine-needle aspiration biopsy of the retroperitoneum: a
leiomyosarcoma: a clinicopathologic analysis of 36 cases. series of 111 cases not including specific organs. Diagn
Am J Surg Pathol 1983; 7:269. Cytopathol 2002; 27:85–89.
54. Demers ML, Curley SA, Romsdahl MM: Inferior vena 73. Leyvraz S, Costa J: Histological diagnosis and grading of
cava leiomyosarcoma. J Surg Oncol 1992; 51:89. soft-tissue sarcomas. Semin Surg Oncol 1988: 4:3.
55. Cardell BS, McGill DAF, Williams F: Leiomyosarcoma of 74. Kulander BG, Polissar L, Yang CY, et al: Grading of soft
inferior vena cava producing Budd-Chiari syndrome. tissue sarcomas: necrosis as a determinant of survival.
J Pathol 1971; 104:283. Mod Pathol 1989; 2:205.
56. Ozzello L, Stout AP, Murray MR: Cultural characteristics 75. Benjmark S, Hafstrom L, Jonsson PE, et al:
of malignant histiocytomas and fibrous xanthomas. Retroperitoneal sarcoma treated by surgery. J Surg Oncol
Cancer 1963; 16:331. 1980; 14:307.
670 Part VI Testis

76. Catton CN, O’Sullivan B, Kotwall C, et al: Outcome and 94. Shiloni E, Szold A, White DE, Freund HR: High-grade
prognosis in retroperitoneal soft tissue sarcoma. Int retroperitoneal sarcomas: role of an aggressive palliative
J Radiat Oncol Biol Phys 1994; 29:1005. approach. J Surg Oncol 1993; 53:197.
77. Cody II HS, Turnbull AD, Fortner JG, et al: The 95. Gupta AK, Cohan RH, Francis IR, Sondak VK, et al:
continuing challenge of retroperitoneal sarcomas. Ann CT of recurrent retroperitoneal sarcomas. AJR 2000,
Surg 1984; 200:200. 174:1025–1030.
78. Singer S, Corson JM, Demetri GD, et al: Prognostic 96. Suit HD, Mankin HJ, Wood WC, Proppe KH:
factors predictive of survival of truncal and Preoperative, intraoperative, and postoperative radiation
retroperitoneal soft-tissue sarcoma. Ann Surg 1995; in the treatment of primary soft tissue sarcomas. Cancer
221:185. 1985; 55:2659.
79. Bevilacqua RG, Rogatoko A, Hajdu SI, Brennan MF: 97. O’Sullivan B, Ward I, Catton C: Recent advances in
Prognostic factors in primary retroperitoneal soft-tissue radiotherapy for soft-tissue sarcoma. Curr Oncol Rep
sarcomas. Arch Surg 1991; 126:328. 2003; 5:274–281.
80. Alvarenga JC, Ball AB, Fisher C, et al: Limitations of 98. Youssef E, Fontanesi J, Mott M, Kraut M, et al:
surgery in the treatment of retroperitoneal sarcoma. Long-term outcome of combined modality therapy in
Br J Surg 1991; 78:912. retroperitoneal and deep-trunk soft-tissue sarcoma;
81. Ferrario T, Karakousis CP: Retroperitoneal sarcomas: analysis of prognostic factors. Int J Radiat Oncol Biol
grade and survival. Arch Surg 2003, 138:248–251. Phys 2002; 54:514–519.
82. Wurl E, Lautenschlager C, Meye A, Schmidt H, et al: 99. Gilbeau L, Kantor G, Stoeckle E, Lagarde P, et al:
A multifactorial prognostic mode for adult soft tissue Surgical resection and radiotherapy for primary
sarcoma considering clinical, histopathological and retroperitonea soft tissue sarcoma. Radiother Oncol
molecular data. Anticancer Res 2000; 20:2065–2072. 2002; 65:133–136.
83. Bautista N, Su W, O’Connell T: Retroperitoneal soft- 100. Petersen IA, Haddock MG, Donohue JH, Nagorney
tissue sarcomas: prognosis and treatment of primary and DM, et al: Use of intraoperative electron beam
recurrent disease. Am Surg 2000, 66:832–836. radiotherapy in the management of retroperitoneal soft
84. Malerba M, Doglietto GB, Pacelli F, Carriero C, et al: tissue sarcomas. Int J Radiat Oncol Biol Phys 2002;
Primary retroperitoneal soft tissue sarcomas: results of 52:469–475.
aggressive surgical treatment. World J Surg 1999, 101. Sindelar WF, Kinsella TJ, Chen PW, et al:
23:670–675. Intraoperative radiotherapy in retroperitoneal sarcomas.
85. Russo P, Kim Y, Ravindran S, Huang W, Brennan MF: Final results of a prospective, randomized, clinical trial.
Nephrectomy during operative management of Arch Surg 1993; 128:402.
retroperitoneal sarcoma. Ann Surg Oncol 1997; 102. Willett CG, Suite HD, Tepper JE, et al: Intraoperative
4:421–424. electron beam radiation therapy for retroperitoneal soft
86. Fernandez-Trigo V, Surgarbaker PH: Sarcomas involving tissue sarcoma. Cancer 1991; 68:278.
the abdominal and pelvic cavity. Tumori 1993; 79:77. 103. Gieschen HL, Spiro IJ, Sutt HD, Mark PJ, et al: Long-
87. Skinner DG: Considerations for management of large term results of intraoperative electron beam radiotherapy
retroperitoneal tumors: use of the modified for primary and recurrent retroperitoneal soft tissue
thoracoabdominal approach. J Urol 1977; 117:605. sarcoma. Int J Radiat Oncol Biol Phys 2001; 50:127–131.
88. Lewis JJ, Leung D, Woodruff JM, Murray F, et al: 104. Alektiar KM, Hu K, Anderson L, Brennan MF: High-
Retroperitoneal soft-tissue sarcoma. Ann Surg 1998; dose-rate intraoperative radiation therapy (HDR-IORT)
228:355–365. for retroperitoneal sarcomas. Int J Radiat Oncol Biol
89. Stoeckle E, Coindre JM, Bonvalot S, Kantor G, et al: Phys 2000; 47:157–163.
Prognostic factors in retroperitoneal sarcoma. Cancer 105. Pisters PW, Ballo MT, Fenstermacher MJ, Feig BW:
2001; 92:359–368. Phase I trial of preoperative concurrent doxorubicin and
90. Tsivian A, Lev-Chelouche D, Shtabsky A, Issakov J, et al: radiation therapy, surgical resection, and intraoperative
Aggressive resection of retroperitoneal sarcoma is superior electron-beam radiation therapy for patients with
to simple excision: an animal study using a novel model localized retroperitoneal sarcoma. J Clin Oncol 2003;
for retroperitoneal sarcoma. J Surg Oncol 2002; 21:3092–3097.
81:144–147. 106. Jones JJ, Catton CN, O’Sullivan B, Couture J: Initial
91. Stojadinovic A, Leung D, Hoos A, Jaques DP, et al: results of a trial of preoperative external-beam radiation
Analysis of the prognostic significance of microscopic therapy and postoperative brachytherapy for
margins in 2084 localized primary adult soft tissue retroperitoneal sarcoma. Ann Surg Oncol 2002;
sarcomas. Ann Surg 2002; 235:424–434. 9:346–354.
92. Wang YN, Zhu WQ, Shen ZZ, et al: Treatment of locally 107. Classen J, Hehr T, Lamprecht U, Zumbragel A, et al:
recurrent soft tissue sarcomas of the retroperitoneum: Hyperfractionated 192Ir brachytherapy for recurrent
report of 30 cases. J Surg Oncol 1994; 56:213. retroperitoneal sarcoma: a technique for delivery of local
93. Shibata D, Lewis JJ, Leung DH, Brennan MF: Is there a tumor boost dose. Strahlenther Onkol 2003;
role for incomplete resection in the management of 179:118–122.
retroperitoneal liposarcomas? J Am Coll Surg 2001, 108. Koshy M, Landry JC, Lawson JD, Esiashvili N, et al:
193:373–379. Potential for toxicity reduction using intensity
Chapter 39 Retroperitoneal Tumors 671

modulated radiation therapy (IMRT) for retroperitoneal 124. Svancarova L, Blay JY, Judson IR, van Hoesel QG, et al:
sarcoma. Int J Radiat Oncol Biol Phys 2003; 57:448–449. Gemcitabine in advanced adult soft-tissue sarcomas.
109. Greiner RH, Munkel G, Blattman H, et al: Conformal A phase II study of the EORTC Soft Tissue and Bone
radiotherapy for unresectable retroperitoneal soft tissue Sarcoma Group. Eur J Cancer 2002; 38:556–559.
sarcoma. Int J Radiat Oncol Biol Phys 1992; 22:333. 125. Hensley ML, Maki R, Venkatraman E, Geller G, et al:
110. Zalupski MM, Ryan JR, Hussein ME, et al: Systemic Gemcitabine and docetaxel in patients with unresectable
adjuvant chemotherapy for soft tissue sarcomas of the leiomyosarcoma: results of a phase II trial. J Clin Oncol
extremities. Surg Oncol Clin NA 1993; 2:621. 2002; 20:2824–2831.
111. Potter DA, Glenn J, Kinsella T, et al: Patterns of 126. Bauer TW, Hahn SM, Spitz FR, Kachur A, et al:
recurrence in patients with high grade soft-tissue Preliminary report of photodynamic therapy for
sarcomas. J Clin Oncol 1985; 3:353. intraperitoneal sarcomatosis. Ann Surg Oncol 2001;
112. Weingrad DW, Rosenberg SA: Early lymphatic spread of 8:254–259.
osteogenic and soft-tissue sarcomas. Surgery 1978; 127. Herr HW: Sarcoma of the urinary tract. In Dekernion
84:231. JB, Paulson DF (eds): Genitourinary Cancer
113. Putnam JB Jr, Roth JA: Resection of sarcomatous Management, p 259. Philadelphia, Lea & Febiger,
pulmonary metastases. Surg Oncol Clin North Am 1993; 1987.
2:673. 128. Tsukamoto T, Lieber MM: Sarcomas of the kidney,
114. Gottlieb JA, Baker LJ, O’Bryan RM, et al: Adriamycin urinary bladder, prostate, spermatic cord, paratestis, and
(NSC 123127) used alone and in combination for soft testis in adults. In Raaf JH (ed): Soft Tissue Sarcomas.
tissue and bone sarcoma. Cancer Chemother Rep 1975; Diagnosis and Treatment, p 201. St. Louis, Mosby,
6:271. 1993.
115. Borden EC, Amato DA, Rosenbaum C, et al: 129. Grignon DJ, Ayala AG, Ro JY, et al: Primary sarcoma of
Randomized comparison of three Adriamycin regimens the kidney. A clinicopathologic and DNA flow
for metastatic soft tissue sarcomas. J Clin Oncol 1987; cytometric study of 17 cases. Cancer 1990; 65:1611.
5:840. 130. Mayes DC, Fechner RE, Gillenwater JY: Renal
116. Yap BS, Baker LH, Sinkovics JG, et al: liposarcoma. Am J Surg Pathol 1990; 14:268.
Cyclophosphamide, vincristine, Adriamycin and DTIC 131. Russo P, Brady MS, Conlon K, et al: Adult urological
(CyVADIC) combination chemotherapy for the sarcoma. J Urol 1992; 147:1032.
treatment of advanced sarcomas. Cancer Treat Rep 1980; 132. Sen SE, Malek RS, Farrow GM, et al: Sarcoma and
64:93. carcinosarcoma of the bladder in adults. J Urol 1985;
117. Edmonson JH, Ryan LM, Blum RH, et al: Randomized 133:29.
comparison of doxorubicin alone versus ifosfamide plus 133. Taylor RE, Busuttil A: Case report: adult
doxorubicin or mitomycin, doxorubicin, and cisplatin rhabdomyosarcoma of bladder, complete response to
against advanced soft tissue sarcomas. J Clin Oncol 1993; radiation therapy. J Urol 1989; 142:1321.
11:1269. 134. Mills SE, Bova GS, Wick MR, Young RH:
118. Antman KH, Crowley J, Balcerzak SP, et al: An Leiomyosarcoma of the urinary bladder. A
intergroup phase III randomized study of doxorubicin clinicopathologic and immunohistochemical study of 15
and dacarbazine with or without ifosfamide and mesna in cases. Am J Surg Pathol 1989; 13:480.
advanced soft tissue and bone sarcomas. J Clin Oncol 135. Ahlering TE, Weintraub P, Skinner DG: Management of
1993; 11:1276. adult sarcomas of the bladder and prostate. J Urol 1988;
119. Santoro A, Tursz T, Mouridsen H, et al: Doxorubicin 140:1397.
versus CYVADIC versus doxorubicin plus ifosfamide in 136. Harrison GSM: Malignant fibrous histiocytoma of the
first-line treatment of advanced soft tissue sarcomas: a bladder. Br J Urol 1986; 58:457.
randomized study of the European organization for 137. Oesterling JE, Epstein JI, Brendler CB: Malignant
research and treatment of cancer soft tissue and bone fibrous histiocytoma of the bladder. Cancer 1990;
sarcoma group. J Clin Oncol 1995; 113:1537. 66:1836.
120. DeLaney TF, Spiro IJ, Suit HD, Gebhardt MC, et al: 138. Kerr KM, Grigor KM, Tolley DA: Rhabdomyosarcoma
Neoadjuvant chemotherapy and radiotherapy for large of the adult urinary bladder after radiotherapy for
extremity soft-tissue sarcomas. Int J Radiat Oncol Biol carcinoma. Clin Oncol 1989; 1:115.
Phys 2003, 56:1117–1127. 139. Kawamura J, Sakurai M, Tsukamoto K, Tochigo H:
121. O’Sullivan B, Bell RS: Has “MAID” made it in the Leiomyosarcoma of the bladder eighteen years after
management of high-risk soft-tissue sarcoma? Int J cyclophosphamide therapy for retinoblastoma. Urol Int
Radiat Oncol Biol Phys 2003; 56:1117–1127. 1993; 51:49.
122. Okuno S, Edmonson J, Mahoney M, Buckner JC, et al: 140. Rosser CJ, Slaton JW, Izawa JI, Levy LB, et al: Clinical
Phase II trial of gemcitabine in advanced sarcomas. presentation and outcome of high-grade urinary bladder
Cancer 2002, 94:3225–3229. leiomyosarcoma in adults. Urology 2003; 61:1151–1155.
123. Okuno S, Ryan LM, Edmonson JH, Priebat DA, et al: 141. Martin SA, Sears DL, Sebo TJ, Lohse CM, et al:
Phase II trial of gemcitabine in patients with advanced Smooth muscle neoplasms of the urinary bladder: a
sarcoma: (E1797): a trial of the Eastern Cooperative clinicopathologic comparison of leiomyoma and
Oncology Group. Cancer 2003, 97:1969–1973. leiomyosarcoma. Am J Surg Pathol 2002; 26:292–300.
672 Part VI Testis

142. Engel JD, Kuzel TM, Moceanu MC, Oefelein MG, et retroperitoneal lymphadenectomy in management.
al: Angiosarcoma of the bladder: a review. Urology 1998; J Urol 1983; 130:577.
52:778–784. 150. Torosian MH, Wein AJ: Liposarcoma of the spermatic
143. Yum M, Miller JC, Agarwal BL: Leiomyosarcoma arising cord: case report and review of the literature. J Surg
in atypical fibromuscular hyperplasia (phyllodes tumor) Oncol 1987; 34:179.
of the prostate with distant metastasis. Cancer 1991; 151. Wiener ES, Anderson JR, Ojimba JI, et al: Controversies
68:910. in the management of paratesticular rhabdomyosarcoma:
144. Palmer MA, Viswanath S, Desmond D: Adult prostatic is staging retroperitoneal lymph node dissection
rhabdomyosarcoma. Br J Urol 1993; 71:489. necessary for adolescents with resected paratesticular
145. Skinner DG, Sherrod A: Total pelvic exenteration with rhabdomyosarcoma? Semin Pediatr Surg 2001;
simultaneous bowel and urinary reconstruction. J Urol 10:146–152.
1990; 144:1433. 152. Grossman HB, Sonda LP, Lloyd RV, Gikas PW:
146. Sexton WJ, Lance RE, Reyes AO, Pisters PW, et al: Carcinosarcoma of bladder: evaluation by electron
Adult prostate sarcoma: the M.D. Anderson Cancer microscopy and immunohistochemistry. Urology 1984;
Center experience. J Urol 2001; 166:521–525. 24:387.
147. Merimsky O, Terrier P, Bonvalot S, Le Pechoux C, et al: 153. Wick MR, Young RH, Malvesta R, et al: Prostatic
Spermatic cord sarcoma in adults. Acta Oncol 1999; carcinosarcomas. Am J Clin Pathol 1989; 92:131.
38:635–638. 154. Halachmi S, DeMarzo AM, Chow NH, et al: Genetic
148. Coleman J, Brennan MF, Alektiar K, Russo P: Adult alterations in urinary bladder carcinosarcoma: evidence
spermatic cord sarcomas: management and results. Ann of a common clonal origin. Eur Urol 2000; 37:350–357.
Surg Oncol 2003; 10:669–675.
149. Sclama AD, Berger BM, Cherry JM, et al: Malignant
fibrous histiocytoma of the spermatic cord: role of
C H A P T E R

40 Urethral Cancer
Robert C. Flanigan, MD

Urethral cancer is a rare disorder in both men and urethra and bulbomembranous urethra are squamous cell
women. Because of the differences in etiology, presenta- cancers in 90% and 80% of cases, respectively.5 An
tion, cell type, and treatment between the two sexes, this understanding of the normal and abnormal urethral
chapter will address male and female urethral cancer urothelium is, therefore, important to the understanding
separately. of cancer development, local invasion, and metastases.

MALE URETHRAL CANCER PATHOLOGY


Normal Urothelium
Cancer of the male urethra typically occurs in the 5th
decade of life but cases have been described from ages 13 Normal transitional urothelium is composed of 3 to 7
to 90.1 The etiologies that have been shown to be related layers of transitional cells with large umbrella cells over-
include chronic infection or inflammation, urethral stric- lapping the cells of the intermediate cell layers that in
ture, sexually transmitted disease, and human papillo- turn rest on a basal cell layer. Below the epithelium lies
mavirus 16 (HPV 16).2 The incidence of urethral stricture the lamina propria and submucosal layer. The submu-
in men who develop urethral cancer ranges from 24% to cosal layer contains the muscularis mucosa and multiple
76% and most often involves the bulbomembranous ure- vascular spaces. Below the submucosal layer lies the ure-
thra. Because the development of cancer in a chronic stric- thral muscle. The diminished barrier to tumor extension
ture patient is often insidious, the urologist should have a presented by the thin muscular layer explains the ready
high index of suspicion and a low threshold to biopsy a access of tumor to the paraurethral space.6
urethral stricture in this setting, especially when the stric-
ture is rapidly recurrent or does not otherwise respond
Abnormalities of the Urothelial Layer
well to treatment. It is estimated that nearly one-quarter of
patients with urethral cancer have a history of sexually Abnormalities of the urothelial layer can range from
transmitted disease and that over 90% of these patients are hyperplasia to carcinoma. Hyperplasia may be further
symptomatic at presentation.3 The most common present- subdivided into epithelial hyperplasia, inverted papil-
ing clinical signs of urethral cancer are bleeding, perineal loma, von Brunn’s nests, and cystitis cystica.
pain, decreased force of stream due to stricture or obstruc-
tion, urinary frequency or dysuria, or urinary fistula.
Metaplasia
Location and histologic type are the keys to catego-
rization of male urethral strictures.4 The frequency of Metaplastic lesions of the urothelium include cystitis
cancer by site is bulbomembranous urethra (60%), penile glandularis (believed to be a precursor of urothelium ade-
urethra (30%), and prostatic urethra (10%). Eighty per- nocarcinoma), nephrogenic adenoma, and squamous
cent are squamous cancers, 15% transitional cell, and 5% metaplasia.
adenocarcinomas or undifferentiated tumors. The histo-
logic subtypes, as expected, vary with anatomic site,
Dysplasia
based largely on the typical lying cells of that area (Figure
40-1). Thus, carcinomas of the prostatic urethra are typ- Dysplasia of the urethra may take the form of atypical
ically transitional cell (90%), while cancers of the penile hyperplasia, mild atypia, moderate atypia, or severe

675
676 Part VII Urethra and Penis

tional cell epithelium may occur in the absence of visible


bladder tumor, its association with either a concurrent or
prior bladder tumor significantly increases the risk of
progression to invasion.
The cystoscopic appearance of carcinoma in situ is
often misleading in that a normal-appearing epithelium
is the most common finding. A reddened or velvety patch
of erythematous mucosa should, however, suggest the
possibility of carcinoma in situ. Carcinoma in situ, like
papillary urothelial cancer, is more common in men than
in women. Because carcinoma in situ frequently presents
with irritative voiding symptoms, the patient is often
misdiagnosed as suffering from prostatism, urinary
tract infection, or neuropathic bladder dysfunction.
Cytopathologic studies of voided urine are positive in
85% to 90% of patients with carcinoma in situ.
The natural history of carcinoma in situ is unpre-
dictable. Early in its evolution, carcinoma in situ may
produce no symptoms, and, in fact, patients with focal
asymptomatic carcinoma in situ have been reported to
generally display a protracted clinical course with low
likelihood of development of invasive cancer. In contrast,
patients with more diffuse or symptomatic carcinoma in
situ, especially in association with a history of bladder
cancer, often display a poor prognosis despite definitive
therapy.
Figure 40-1 Anatomic regions of the male urethra and
corresponding histology and histopathology. Papillary Transitional Cell Carcinoma
TCC is generally graded on a scale of 1 to 3 (some sys-
tems grade from 1 to 4). Grade 1 TCCs differ from nor-
atypia. In addition, leukoplakia of the urethra may occur. mal epithelium by having an increased number of
Dysplastic urothelium exhibits histologic changes that epithelial cell layers. With increasing grade, abnormali-
are intermediate between normal urothelium and carci- ties of nuclear morphology, loss of cellular polarity,
noma in situ. It is generally felt that lesions on the severe abnormalities of the normal cellular maturation from
end of this spectrum are neoplastic rather than dysplastic. the basal to superficial layers, nuclear crowding, and
increased nuclear-to-cytoplasmic ratio are seen. In
addition, prominent nucleoli with clumping of the
Superficial Carcinomas
chromatin and an increased number of mitoses may be
Superficial carcinomas of the urethral urothelium include appreciated.
carcinoma in situ and cancers involving the epithelium
and submucosal layers. Histologically, these cancers are
Squamous Cell Carcinoma and Adenocarcinoma
either transitional cell, squamous, or adenocarcinomas.
In the United States, squamous cell carcinomas account
for approximately 80% of urethral cancers. These tumors
Carcinoma In Situ
are commonly associated with a chronic inflammatory
Histologically, carcinoma in situ is characterized by a process involving the urothelium, including the presence
poorly differentiated cancer involving only the urothe- of a chronic foreign body (e.g., an indwelling catheter,
lium. A loss of cellular cohesiveness, widening of the urinary tract calculi, or recurrent urinary tract infec-
intracellular spaces, separation of cells from the basement tions). Generally, a greater proportion of patients with
membrane, and a loss of the superficial umbrella cell squamous cell carcinoma, as compared with TCC, pres-
layer is characteristic. Carcinoma in situ may occur ent with advanced disease at the time of diagnosis.
focally or diffusely within the urothelium. It is often Adenocarcinoma of the urethra is an uncommon
associated with high-grade and high-stage transitional lesion accounting for very small percentage of urethral
cell cancers. Although carcinoma in situ of the transi- cancers.
Chapter 40 Urethral Cancer 677

NATURAL HISTORY extent of involvement of the surrounding tissues and can


vary from transurethral excision or local resection to par-
Numerous chromosomal abnormalities have been
tial or total penile amputation. The treatment of urethral
found to be associated with the development of TCC,
cancer can also be divided into three areas of urethral
including chromosome 9 loss, c-myc amplification, Y
involvement: the penile urethra and fossa navicularis, the
chromosome loss, and p53, p21, and erb-B-2 abnormal-
bulbomembranous urethra, and the prostatic urethra.
ities. Of these, chromosome 9 loss and p53 abnormali-
The prognosis of urethra cancers is dependent on the
ties may be the most important. Loss of heterozygosity
type of cancer, site of the lesion (distal better than proxi-
of chromosome 9 seems to be an early event associated
mal), the depth of invasion, and the ability to achieve
with the formation of papillary tumors, whereas p53
complete local control.9 A summary of prognosis by site
gene deletion seems to be associated with the formation
of involvement is given in Table 40-2.
of dysplasia and carcinoma in situ. By the time a TCC
invades the lamina propria, both of these genetic events
have typically occurred. Chromosome 9 deletion is felt Penile Urethra and Fossa Navicularis Cancers
to result in a homozygous deletion of the MTS-1 gene
When distal urethral cancer is superficial, papillary, or in
(a tumor-suppressor gene), and it is a frequent event in
situ, transurethral resection and fulguration may be
TCC but not a driving force in tumor progression. P53
employed. When the cancer invades the corpus spongio-
is a tumor-suppressor gene located at 17p13.1, which
sum, more aggressive therapy is indicated and may
codes for a DNA-binding protein involved in the regu-
include urethrectomy with sparing of the corpora caver-
lation of transcription. Deletion of this gene results in
nosa (penile-sparing), partial penectomy with an ade-
an overexpression of the p53 (missense mutation), lead-
quate margin (2 cm), and total penectomy. Partial
ing to an extension of the half-life of the protein. p53
penectomy is most useful in cancers that involve the dis-
and another transcription-mediating gene, p21, are
tal half of the penis. In any case, the advantages of tissue-
associated with progression of TCC.
conserving approaches must be balanced against the
The natural history and molecular biology of squa-
probability of local recurrence and death from metastatic
mous cell and adenocarcinomas of the urothelium are
disease.10 Ilioinguinal lymphadenectomy is indicated in
less well understood than that of TCC. Squamous cell
the presence of palpable nodes but the value of prophy-
carcinoma is an invasive lesion with a nodular infiltrative
lactic node dissection has not been demonstrated in this
growth pattern. It is postulated that nitrosamines pro-
disease.
duced by bacteria in infected urine act as carcinogens and
tumor promoters. A recent study of cytogenetics in squa-
mous cell urethra cancer has demonstrated involvement Bulbomembranous Cancers
of chromosomes Y, 2, 3, 4, 6, 7, 8, 11, and 20 but of inter-
Superficial lesions of the bulbomembranous urethra are
est, not chromosomes 9 and 17, which are widely involved
quite rare but have been successfully managed by
in transitional cell cancer.7
transurethral resection and local excision with end-to-
end urethral reanastomosis. In general, radical excision
Evaluation and Staging seems to be the best approach, employing radical cysto-
The TNM staging system is based on the depth of invasion of prostatectomy combined with total penectomy and bilat-
the primary tumor, the presence of regional lymph node eral pelvic lymphadenectomy. In severe cases, extending
involvement, and the presence of distant metastases (Table 40-1). the en bloc excision to include the pubic rami and uro-
The diagnosis and staging of urethral cancer begins with phys- genital diaphragm has been recommended.11,12
ical examination of the external genitalia, urethra, rectum, and
perineum. Cystoscopy and bimanual examination-under- Prostatic Urethral Cancers
anesthesia are often useful. Transurethral or percutaneous
needle biopsy of the lesion is performed. Cytologic studies of Primary carcinoma of the prostatic urethra is rare. In this
first voided urine may be helpful. Local involvement, includ- setting, transitional cell and adenocarcinomas are the
ing involvement of regional lymphatics and contiguous struc- most common histologies. It is imperative to rule out
tures, as well as estimation of the extent of local disease, is best extension of these cancers from the bladder, as this pres-
determined by MR or CT imaging. MR is particularly help- entation is far more common than tumor arising in the
ful in determining invasion of the corporal bodies.8 prostatic urethra. Patients typically present with hema-
turia or obstructive symptoms and rarely with prostatic
induration on digital rectal exam (a sign of advanced dis-
Treatment
ease). Diagnosis may require both transurethral biopsy
Surgical excision remains the primary therapy for male and transrectal ultrasound-guided needle biopsy.
urethral cancers. The extent of resection depends on the Superficial papillary or in situ cancers may be managed
678 Part VII Urethra and Penis

Table 40-1 Urethral Cancer TNM Staging System


Primary tumor (T) (male and female)

Tx Primary tumor cannot be assessed

T0 No evidence of primary tumor

Ta Noninvasive papillary, polypoid, or verrucous carcinoma

Tis Carcinoma in situ

T1 Tumor invades subepithelial connective tissue

T2 Tumor invades any of the following: corpus spongiosum, prostate, periurethral muscle

T3 Tumor invades any of the following: corpus cavernosum, beyond prostatic capsule, anterior vagina, bladder neck

T4 Tumor invades other adjacent organs

Transitional cell carcinoma of the prostate

Tis-pu Carcinoma in situ, involvement of the prostatic urethra

Tis-pd Carcinoma in situ, involvement of the prostatic ducts

T1 Tumor invades subepithelial connective tissue

T2 Tumor invades any of the following: prostatic stroma, corpus spongiosum, periurethral muscle

T3 Tumor invades any of the following: corpus cavernosum, beyond prostatic capsule, bladder neck (extra prostatic exten-
sion)

T4 Tumor invades other adjacent organs (invasion of the bladder)

Regional lymph nodes (N)

Nx Regional lymph nodes cannot be assessed

N0 No regional lymph node metastasis

N1 Metastasis in a single lymph node, 2 cm or less in greatest dimension

N2 Metastasis in a single lymph node, more than 2 cm but <5 cm in greatest dimension; or in multiple nodes, none >5 cm

N3 Metastasis in a lymph node >5 cm in greatest dimension

Distant metastasis (M)

Mx Presence of distant metastasis cannot be assessed

M0 No distant metastasis

M1 Distant metastasis

OPERATIVE TECHNIQUES FOR MALE URETHRA


successfully by transurethral resection in select
CANCERS
patients.13 In the great majority of cases, however, the
Partial and Total Urethrectomy in the Male Patient
cancer involves the prostatic stroma and is typically
with Primary Urethral Carcinoma
treated by cystoprostatectomy with total urethrectomy
and bilateral pelvic lymphadenectomy. The outlook in Distal urethral carcinoma in the male includes all tumors
this setting is poor, with reported 5-year survival ranging distal to the bulbous urethra.15 These tumors may be
from 6% to 26%.14 managed by transurethral resection, partial penectomy,
Chapter 40 Urethral Cancer 679

Table 40-2 Treatment Survival for Urethral Cancer


Site Treatment No. Patients Survival

Fossa navicularis Partial penectomy 12 11 (92%)

Penile Partial or total penectomy 100 34 (34%)

Bulbomembranous Total penectomy, urethrectomy, 64 10 (16%)


cystoprostatectomy

Prostatic superficial Transurethral resection 45 39 (87%)

Invasive Cystoprostatectomy, urethrectomy 78 29 (37%)

are incised and the bulbocavernosus muscle divided in


the midline and reflected laterally to expose the bulbous
urethra. A perineal ring retractor (e.g., Turner-Warwick
or Omni) provides good exposure. After a segment of
the urethra is isolated by incising Buck’s fascia overlying
the inferior lateral grooves between the corpus spongio-
sum and the corpus cavernosum and dividing its supero-
medial attachments, a Penrose drain is advanced through
the defect and used to create countertraction on the cor-
pus spongiosum during the remaining distal dissection
(Figure 40-3B). The plane between corpus spongiosum
and corpus cavernosus is remarkably easy to develop in
most cases, although inadvertent laceration of the tunica
albuginea of the corpora cavernosa may produce consid-
erable hemorrhage, which is most readily controlled
with continuous 3-0 chromic or polyglycolic acid (PGA)
sutures.
During distal dissection, the penis becomes inverted
and the plane between the corpus spongiosum and cor-
pora cavernosa is lost. A decision must be made whether
or not to preserve the fossa navicularis at this point.
Preservation of the fossa navicularis may be helpful in
Figure 40-2 A, Patient position for radical allowing for more adequate placement and seating of the
cystoprostatectomy. Umbilicus is placed over break of table, tips of a penile prosthesis at a later date. If the fossa nav-
and table is fully flexed and tilted into Trendelenburg’s icularis is to be resected, the penile meatus is circum-
position, until legs are parallel to floor. B, Patient position for scribed using a tear-shaped incision and the fossa
urethrectomy. Leg braces are elevated until hips are flexed 60 navicularis is mobilized from the glans penis and pulled
degrees and knees are fully extended. into the perineal incision (Figure 40-5). Defects in the
glans penis are closed with interrupted 2-0 chromic or
or partial penectomy with perineal urethrostomy. Standard PGA sutures. Isolation of the proximal urethra at the bul-
positioning techniques are pictured in Figure 40-2. bomembranous junction is the most difficult aspect of
For patients with posterior urethral cancers, total ure- the procedure. Dissection is carried in a posterior and
threctomy with cystoprostatectomy is typically under- lateral direction with special care to identify and secure
taken. The patient is placed in the lithotomy position branches of the pudendal arteries entering at the
with special attention to abduction of the legs. After rad- 4 o’clock and 8 o’clock positions (Figure 40-4). Small
ical cystectomy and lymphadenectomy is completed, the hemoclips or 3-0 chromic/PGA sutures are particularly
membranous urethra is dissected from the urogenital useful for this maneuver. The urethrectomy is completed
diaphragm using finger dissection. Attention is then by mobilizing the most proximal bulbous urethral spon-
directed to the perineum. A midline or inverted Y inci- giosum and by dissecting the remaining membranous
sion is made in the anterior perineum overlying the cen- urethra from the urogenital diaphragm. At this point, the
tral tendon (Figure 40-3). The superficial and deep fascia appropriate plane of dissection may be best appreciated
680 Part VII Urethra and Penis

Figure 40-3 Total urethrectomy with cystoprostatectomy.


Chapter 40 Urethral Cancer 681

Figure 40-4 Control of the bulbar arteries fudging urethrectomy.

Figure 40-5 Inverted T-incision on glans penis and dissection


of glanular urethra.

by combined perineal and abdominal palpation and


inspection.
The perineal wound is carefully irrigated and a suc-
tion or Penrose drain is positioned in the bed of the
bulbous urethra and brought out through a separate
stab wound (Figure 40-6). Reapproximation of the bul-
bocavernosus muscle is undertaken in the midline using
2-0 chromic or PGA sutures. The subcutaneous tissue
is closed with interrupted 3-0 chromic or PGA sutures
and the skin is closed with interrupted 3-0 nylon
sutures. The shaft of the penis is wrapped loosely with
gauze or is covered with a self-adhering dressing of Figure 40-6 Closure of the perineal incision and placement
some variation. Exposure of the tip of the glans penis in of the Jackson-Pratt drain.
682 Part VII Urethra and Penis

order to permit prompt identification of postoperative FEMALE URETHRAL CANCERS


ischemia is important.
Urethral cancer is more common in women than men
(ratio 4:1). Risk factors include race (Caucasian more
Total Urethrectomy in the Male Patient in common), age (>50 years most common), a history of
Association with Cystectomy chronic inflammation or irritation, urinary tract infec-
tions, or proliferative lesions, including caruncles,
The procedure for concomitant urethrectomy at the time papillomas, adenomas, and leukoplakia. Most women
of cystoprostatectomy is very similar to that described present with urethral bleeding, urinary symptoms, and a
previously for the treatment of primary urethral cancer. palpable mass or induration. Tumors may be papillary,
The bulbomembranous and prostatic urethral segments fungating, or ulcerating and may cause induration of the
are those most often involved by extension of the primary anterior vaginal wall. Odor producing necrosis and dis-
bladder cancers; thus careful dissection of these areas is charge may occur. Local extension is not uncommon
essential. This part of the dissection can be considerably and may involve the bladder neck, vagina, or vulva.
more difficult when urethrectomy follows cystoprostate- Metastases to regional lymphatics are common, occur-
ctomy by more than several weeks.16 In such cases, a ring in up to one-third of patients at presentation.
Foley catheter must be placed to the proximal end of the Anterior urethral cancers drain into the superficial and
urethra and secured in place by suturing to the meatus, deep inguinal nodes whereas posterior urethral tumors
and dissection carried to the proximal extent of the drain into the external iliac, hypogastric, and obturator
catheter. Dissection of the proximal urethra should be nodes. Female urethral cancer often produces systemic
done carefully and methodically because extensive trac- metastases without regional node involvement. Generally,
tion on the urethra may avulse the proximal portion as a therefore, groin dissection is reserved for those patients
result of extensive fibrosis in this area. Drainage of the who have palpable lymphadenopathy without distant
proximal urethral bed using a Jackson-Pratt or Penrose spread of cancer.19
drain is suggested.

Pathology
Radiation Therapy and Multimodal Therapy
As with male urethral cancer, the urethral cancer cell type
Radiation therapy alone may be adequate to control in women is related to the site of urethra involved and
small lesions in the distal urethra.17 Radiation may be may be squamous, transitional cell, or adenocarcinomas
delivered using brachytherapy or intracavitary tech- (Figure 40-7). Carcinomas of the proximal or posterior
niques. Chemotherapy using MVAC or paclitaxel and urethral are typically locally advanced and high grade,
ifosfamide have recently been reported to successfully whereas distal cancers are, in general, low grade and less
treat transitional cell cancers of the urethra.18 extensive. Overall, squamous cell cancers are most common

Figure 40-7 Anatomic regions of the female urethra and


corresponding histology and histopathology.
Chapter 40 Urethral Cancer 683

(60%), followed by transitional cell (20%), adenocarci- rior exenteration with wide resection of the vagina in
noma (10%), undifferentiated or sarcomatous (8%), and most cases. Partial and total urethrectomy can be useful
melanoma (2%). in selected cases of female urethral cancer.

Evaluation and Staging OPERATIVE TECHNIQUES FOR PARTIAL AND


TOTAL URETHRECTOMY IN THE FEMALE
Pelvic examination combined with cystoscopy and biopsy
Partial and Total Urethrectomy in the Female
is the cornerstones of diagnosis. The TNM staging sys-
Patient with Primary Urethral Cancer
tem is similar to that of male urethral cancer and is
shown in Table 40-1. Prognosis has been reported in var- The female patient with primary urethral cancer is pre-
ious studies to be dependent on tumor size, location in pared for anterior exenteration with mechanical and
the urethra (posterior versus anterior), histology, and antibiotic bowel preparation, systemic antibiotics, and
extent of disease (early versus advanced) (Table 40-3).20 intravenous hydration. In those patients presenting with
In one series of 76 patients, 5-year disease-specific sur- tumors of the distal third of the urethra that are con-
vival was 89% for low-stage tumors compared to 33% for fined to the mucosa, submucosa, or periurethral muscu-
high-stage tumors.21 lature, a distal urethrectomy may be appropriate. Distal
urethrectomy is performed with the patient in the litho-
tomy position after full vaginal and abdominal prepara-
Treatment
tion. A weighted speculum is used to improve
Although local excision may suffice for low grade, low- visualization of the urethral orifice. A traction suture
stage tumors, single modality therapy often fails to ade- ligature of 0 or 2-0 silk is passed through the urethral
quately control advanced cancer and is associated with a meatus or the surrounding periurethral tissue. An encir-
20% 5-year survival and a local recurrence rate exceeding cling incision is then made around the meatus (Figure
65%.22 As with male urethral cancer, treatment options 40-8). After sharp dissection of the urethral meatus, a
vary considerably with site of urethral involvement: distal longitudinal incision is made on the vaginal mucosa
or proximal. overlying the epithelium to approximately 2 cm beyond
the point of palpable induration. Sharp dissection then
circumscribes the urethra, which is transected at the
Distal Urethral Cancer
previously determined proximal extent of dissection.
Distal cancers, as stated above, are more often superficial Frozen sections of the proximal urethral margin should
and well localized with infrequent lymph node metas- be evaluated to document the absence of microscopic
tases. Local excision is often sufficient and is accom- involvement of the urethra at that point. Mucosal edges
plished by circumferential excision of the involved of the remaining urethra may then be approximated to
urethra with adjacent portions of the anterior vagina. the vaginal mucosa using 3-0 chromic or PGA sutures.
Spatulation of the remaining urethra with approxima- A 16 to 18 French Foley catheter is inserted into the
tion to the adjacent vagina and labia is associated with bladder to provide urinary drainage. Vaginal repair is
preservation of continence in most cases. then completed using an interrupted or running 2-0 or
3-0 chromic or PGA suture. Povidone-iodine soaked
vaginal packing is left within the vaginal vault and is
Proximal Urethral Cancer
removed on postoperative day 1.
Proximal urethral cancers are most often aggressive and In those patients in whom tumor extends microscopi-
invasive and require extensive resection, including ante- cally beyond the point of palpable induration, and/or

Table 40-3 Prognosis for Early and Advanced Tumors


Stage Treatment No. Patients Survival

Early Radiation therapy 140 94 (67%)

Surgery 24 20 (83%)

Radiation therapy plus surgery 5 4 (80%)

Advanced Radiation therapy 157 54 (34%)

Radiation therapy plus surgery 39 21 (54%)


684 Part VII Urethra and Penis

Figure 40-9 In extensive pelvic/perineal exenteration, the


urethra and external genitalia in continuity with the pelvic
structures are removed. The pubic rami are identified and
prepared for excision by freeing the overlying subcutaneous
Figure 40-8 Technique for resection of the distal third of the tissue, the suspensory ligament of the penis, and the various
female urethra. muscular attachments. The outlines of exenteration of the
bone are indicated by the dotted lines.
those patients with proximal urethral cancers in whom
proximal urethral involvement of the tumor is established report, 6 women were managed with high-dose rate
preoperatively, total urethrectomy, vulvectomy, clitorec- brachytherapy, anterior exenteration, and external beam
tomy, and anterior exenteration may be required to radiation therapy with 4 patients receiving adjuvant plat-
remove the tumor with an adequate margin.23 In selected inum-based chemotherapy. Despite this approach, 3 of 6
cases, resection of the pubic rami may also be necessary.24 patients had distant metastases at a mean follow-up of 21
The female candidate for total urethrectomy is prepared months.27 In another series, surgery was combined with
in a fashion similar to that described for partial urethrec- either neoadjuvant or adjuvant radiation therapy in 12
tomy. The operation is generally begun abdominally, patients, resulting in a 44% 5-year cause-specific sur-
where radical cystectomy and pelvic lymph node dissec- vival.20 A single case of squamous cell cancer successfully
tions are performed in the standard fashion. In addition, managed with external beam radiation therapy and cis-
salpingo-oophorectomy and hysterectomy are also per- platin, and 5-fluorouracil has been reported.28
formed in most cases. Resection of the anterior urethra
en bloc with the abdominal structures proceeds along the
lines described previously. Complete excision of the ante- REFERENCES
rior compartment of the perineum may require a gracilis
flap to provide for adequate pelvic floor support (Figure 1. Weiner JS, Liu ET, Walther PJ: Oncogenic human
40-9).25 Resection of the pubic rami is undertaken only in papillomavirus type 16 is associated with squamous cell
very extensive tumors because it may be associated with cancer of the male urethra. Cancer Res 1992;
significant complications, including sacroiliac joint dis- 52:5018–5023.
ruption and perineal hernia formation.26 It is important 2. Cupp MR, Reza MS, Goellner JR, et al: Detection of
to preserve vulvar skin whenever possible and to remove human papillomavirus DNA in primary squamous cell
the specimen en bloc so as to avoid contamination of the carcinoma of the male urethra. Urology 1996;
operative field by tumor spillage. After completion of the 48:551–555.
extirpative aspects of the procedure, urinary diversion is 3. Dalbagni G, Zhang ZF, Lacombe L, Herr HW: Male
urethral carcinoma: analysis of treatment outcome.
completed.
Urology 1999; 53:1126–1132.
4. Mostofi FK, Davis CJ, Sesterhenn IA: Carcinoma of the
Multimodal Therapy male and female urethra. Urol Clin North Am 1992;
19:347–358.
Because of the dismal results with single modality ther- 5. Grigsby PW, Herr HW: Urethral tumors. In
apies in female urethral cancers, attempts to provide a Volgelzang N, Scardino PT, Shipley WU, Coffey DS
multimodal approach have been undertaken. In a recent (eds): Comprehensive Textbook of Genitourinary
Chapter 40 Urethral Cancer 685

Oncology, pp 1133–1139. Baltimore, Williams & 18. VanderMolen LA, Sheehy PF, Dillman RO: Successful
Wilkins, 2000. treatment of transitional cell carcinoma of the urethra
6. Flanigan RC, Kim FJ: Neoplastic disease of the pelvis, with chemotherapy. Cancer Invest 2002; 20(2):206–207.
ureter, bladder and urethra. In Textbook of Nephrology, 19. Grigsby PW, Herr HW: Urethral tumors. In Volgelzang
4th edition. Philadelphia, Lippincott, Williams & N, Scardino PT, Shipley WU, Coffey DS (eds):
Williams,2001. Comprehensive Textbook of Genitourinary Oncology, pp
7. Fadl-Elmula I, Gorunova L, Mandahl N, Elfving P, Heim 1133–1139. Baltimore, Williams & Wilkins, 2000.
S: Chromosome abnormalities in squamous cell carcinoma 20. Grigsby PW: Carcinoma of the urethra in women. Int J
of the urethra. Genes, Chromosomes Cancer 1998; Radiat Oncol Biol Phys 1998; 41(3):535–541.
23:72–73. 21. Dalbagni G, Zhang ZF, Lacombe L, Herr HW: Female
8. Vapnek JM, Hricak H, Carroll PR: Recent advances in urethral carcinoma: an analysis of treatment outcome and
imaging studies for staging of penile and urethral a plea for a standardized management strategy. Br J Urol
carcinoma. Urol Clin North Am 1992; 19:257–266. 1998; 82:835–841.
9. Zeidman EJ, Desmond P, Thompson I: Surgical treatment 22. Narayan P, Konety B: Surgical treatment of female urethral
of carcinoma of the male urethra. Urol Clin North Am carcinoma. Urol Clin North Am 1992; 19:373–382.
1992; 19:359–372. 23. Grabstald H, Hilaris B, Henschke U, Whitmore WF:
10. Davis JW, Schellhammer PF, Schlossberg SM: Cancer of the female urethra. JAMA 1966; 197:835–842.
Conservative surgical therapy for penile and urethral 24. Klein FA, Whitmore WF, Herr HW, et al: Inferior pubic
carcinoma. Urology 1999; 53(2):386–392. rami resection with en bloc radical excision for invasive
11. Mackenzie AR, Whitmore WF: Resection of the pubic proximal urethral carcinoma. Cancer 1983; 51:1238–1242.
rami for urologic cancer. J Urol 1968; 100:546–551. 25. Johnson DE, Lo RR: Tumors of the penis, urethra and
12. Klein EA, Herr HW, Whitmore WF, et al: Inferior pubic scrotum. In deKernion JB, Paulson DF (eds):
rami resection with en bloc radical excision for invasive Genitourinary Cancer Management, pp 219–270.
proximal urethral carcinoma. Cancer 1983; 51:1238–1242. Philadelphia, Lea & Febiger, 1987.
13. Bretton PR, Herr HW: Intravesical BCG for in situ 26. MacKenzie AR, Whitmore WF: Resection of the pubic
transitional cell carcinoma involving the prostatic urethra. rami for urologic cancer. J Urol 1968; 100:546–551.
J Urol 1989; 141:853–856. 27. Dalbagni G, Donat SM, Eschwege P, Herr HW, Zelefsky
14. Hall RR, Robinson MC: Transitional cell carcinoma of MJ: Results of high dose rate brachytherapy, anterior
the prostate. Eur Urol Update Series 1998; 7:1–7. pelvic exenteration and external beam radiotherapy for
15. Mandler JI, Pool TL: Primary carcinoma of the male carcinoma of the female urethra. J Urol 2001;
urethra. J Urol 1966; 96:67–72. 166(5):1759–1761.
16. Brendler CB: Urethrectomy. In Walsh PC, Retik AB, 28. Lee KC: Carcinoma of the female urethra responsive to
Stamey TA, Vaughan ED Jr (eds): Campbell’s Urology, 6th moderate dose chemoradiotherapy. J Urol 2000;
edition, pp 2774–2781. Philadelphia, WB Saunders, 1992. 163:905–906.
17. Sailer SL, Shipley WU, Wang CC: Carcinoma of the
female urethra: a review of results with radiation therapy.
J Urol 1988; 140:1–5.
C H A P T E R

41 Urethrectomy
Sanjaya Kumar, MD

INDICATIONS
involving the urethra. The prostatic urethra is usually
Over the years, the indications for urethrectomy have involved and this determination is made from prior stag-
changed. The trend has shifted towards preservation of the ing resection and biopsy. A frozen section analysis of the
urethra. The indications for preserving the urethra have membranous urethra is always undertaken at the time of
been liberalized for several reasons. These include the cystoprostatectomy. Positive findings will necessitate
widespread utilization of orthotopic neobladders in both abandoning the orthotopic neobladder and performing a
men and women, better understanding of the anatomy of complete urethrectomy. If the patient is not able to tol-
the female urethra, and importantly the reduced risk of erate the simultaneous procedure, the urethrectomy can
urethral recurrence following orthotopic urinary diver- be performed at a later date.
sion.1 Currently, the absolute indication to remove the For a concomitant radical cystoprostatectomy, the
urethra includes the involvement of the urethra by carci- patient is positioned in a lithotomy position to allow
noma. The single most important predictor of recurrent access to the abdomen, pelvis, and the perineum.
disease in the male urethra is transitional cell carcinoma Modified stirrups or boots are usually used that allow the
(TCC) involvement of the prostatic urethra, specially the position of the patient to be modified in a sterile fashion
stroma.2–4 The single most important predictor of recur- during the urethrectomy (Figure 41-1). All pressure
rent disease in the female urethra is involvement of tumor points must be carefully padded and protected. A two-
at the bladder neck.5,6 However, the absolute indication team approach may be more expeditious. Once the dis-
for a prophylactic urethrectomy includes the histologic section for the cystoprostatectomy is complete, attention
evidence of cancer at the urethral margin at the time of is directed towards performing an en bloc urethrectomy.
cystectomy in both men and women. At this point the dorsal venous complex and other attach-
A urethrectomy may be performed simultaneously dur- ments have been divided and the bladder and prostate are
ing a cystoprostatectomy for transitional cell cancer, if the in continuity with the membranous urethra. The mem-
prostatic urethra is involved by tumor at the time of the branous urethra is dissected from the urogenital sinus
cystectomy, or at a later time, should there be a recurrence using blunt and sharp dissection. An umbilical tape is
of tumor in the urethra. Ideally, a complete urethrectomy passed under the urethra, below the apex of the prostate.
is performed, both in males and females. However, in Gentle traction on the tape facilitates dissection in a plane
males, under certain circumstances, such as when there is a between the urethral smooth muscle and the striated mus-
superficial isolated solitary recurrence, following an ortho- cle of the urogenital diaphragm. The dissection is per-
topic neobladder, a partial or segmental urethrectomy may formed carefully to visualize the bleeders and preserve the
be performed, provided it is surgically feasible. neurovascular bundles located posterolateral to the ure-
thra (Figure 41-2). The use of electrocautery should be
minimized if a nerve-sparing technique is being adopted.7
OPERATIVE TECHNIQUE
Once the membranous urethra has been completely
Urethrectomy in the Male
mobilized from the urogenital diaphragm, attention is
Simultaneous Urethrectomy with
directed towards dissecting the anterior urethra.
Cystoprostatectomy
Secondary urethrectomy is difficult at this location
Pelvic dissection. A simultaneous urethrectomy with due to scarring from previous cystoprostatectomy.
cystoprostatectomy is performed for TCC of the bladder The procedure can result in shredding of the urethra,

686
Chapter 41 Urethrectomy 687

nosum to pass a quarter inch Penrose drain around the


urethra (Figure 41-5). The dorsum of the urethra is usu-
ally adherent to the corpus cavernosum. Traction on the
Penrose and sharp dissection in this plane allows separa-
tion of the urethra from the corpus cavernosum. The
pointed tip electrocautery on a low setting works well in
this midline plane (Figure 41-6). Cavernosal bleeding
and minor rents can be closed with 2-0 Vicryl/monofila-
ment absorbable sutures. Distal dissection and traction
on the urethra eventually results in inversion of the penis
into the wound (Figure 41-7A,B). Once the dissection
reaches the urethral tip, the fossa navicularis and the
meatus are cored out. Alternatively, the penis is straight-
ened, and either a wedge or a T-shaped incision made of
the ventral aspect of the glans penis. The fossa navicularis
and the meatus can now be dissected (Figure 41-8). Strict
hemostasis is ensured. The glans penis is reconstructed
with 3-0 chromic catgut or monofilament absorbable
sutures. For cosmetic reasons, under certain circum-
stances, the fossa navicularis can be preserved.
Once the distal urethra is completely mobilized and
Figure 41-1 A, Patient position for radical disconnected from the glans penis, the proximal urethra
cystoprostatectomy. Umbilicus is placed over break of table, is freed from the perineal investments. The bulbar ure-
and table is fully flexed and tilted into Trendelenburg’s thral arteries are posterolateral to the bulb. These are
position, until legs are parallel to floor. B, Patient position for carefully secured (Figure 41-9). Anteriorly, the urethra
urethrectomy. Leg braces are elevated until hips are flexed 60
is dissected under the pubic symphysis (Figure 41-10).
degrees and knees are fully extended. (From Brendler CB,
Eventually, dissections around the urethra, from the
Schlegel PN, Walsh PC: J Urol 1990; 144:270–273.)
pelvis above and the perineum below, meet up in a com-
mon plane (when combined with cystoprostatectomy),
incomplete resection, and risk injury to bowel. It is there-
through the urethra genital diaphragm. It is important
fore important at this time to dissect the urethra as far
to dissect close to the urethra to avoid injury to the
distally as is safely possible. Attention is now directed
erector nerves. The urethra is now completely mobi-
towards the perineum. However, if a frozen section spec-
lized. It can either be removed separately or pulled from
imen of the urethra is necessary, the urethra should be
the abdominal side and removed en bloc with the cys-
tied before transaction to prevent tumor spillage. The
tectomy specimen. In a secondary urethrectomy, the
urethra may be deliberately divided here if the patient is
very last proximal end of the urethra is surrounded by
unable to withstand anesthesia for the additional time
scar tissue and is carefully dissected. The operator must
necessary to perform the anterior urethrectomy. The
safeguard against urethral avulsion, at the pelvis, by
urethrectomy can then be performed as a secondary pro-
avoiding undue traction on the urethra. The urethra
cedure several weeks later.
must be removed in its entirety (Figure 41-11A,B) while
Perineal dissection. The lithotomy position can now the surgeon is cognizant of potential injury to underly-
be exaggerated by tilting the boots up. An inverted Y- ing bowel.
shaped incision is made in the skin of the perineum The wound is irrigated with antibiotic solution, strict
between the scrotum and the anal verge (Figure 41-3). hemostasis ensured and closed in layers. The bulbocav-
The subcutaneous tissue is divided to expose the bulbo- ernous muscles and the urogenital diaphragm are reap-
cavernous muscle. The bulbocavernous muscle is divided proximated with 2-0/3-0 absorbable sutures. A number 7
in the midline (Figure 41-4). Two Richardson retractors french Jackson-Pratt drain is left indwelling for 24 to 48
or ring retractors are used to keep the wound open. hours in the urethral bed and brought out through a sep-
The corpus spongiosum is now dissected. The urethra arate stab incision (Figure 41-12). The subcutaneous
can be palpated through the urethral catheter. In a sec- tissue is closed with 3-0 absorbable sutures. Additional
ondary urethrectomy, a metal sound (Van Buren) can be layers may need to be reapproximated in the pelvic area
used instead of a catheter. Pinch the catheter surrounded to close all dead space. The skin is closed with 4-0
by the urethra by the fingers and using a Metzenbaum Monocryl. Either Steri-Strips or Dermabond can be used
scissors expose the dorsal aspect of the urethra. Create for additional support. A gentle compressive dressing
adequate space between the urethra and corpus caver- with Kerlex sponges is then applied.
688 Part VII Urethra and Penis

Figure 41-2 A, Urethra is ligated with 1-0 silk suture to prevent spillage of urine
around catheter (NVB, neurovascular bundle; Pros, prostate). B, Mobilization of
membranous urethra from urogenital diaphragm with Kitner dissector. C, Further
mobilization of membranous urethra. D, Lateral view shows membranous urethra (Ur)
fully mobilized with neurovascular bundles displaced posterolaterally. E, Urethra is
transected, and catheter is drawn cephalad into wound. Neurovascular bundles are
seen intact, lateral to the urethra. (From Brendler CB, Schlegel PN, Walsh PC: J Urol
1990; 144:270–273.)

dure are similar to those mentioned in the section on


Total Anterior Urethrectomy in the Male
perineal dissection.
Complete anterior urethrectomy may be performed as a The extent of urethral resection for nontransitional cell
secondary procedure for TCC or as a primary procedure cancer of the urethra depends on the pathology and stage
for nontransitional cell cancers of the anterior urethra. of the cancer. Most of the distal urethral tumors are low
This procedure is performed for transitional cell cancer stage and proximal urethral tumors are high stage.8 Thus,
of the prostatic urethra, as a staged procedure, if a cysto- surgery for tumors in the anterior urethra may vary from
prostatectomy with concomitant urethrectomy cannot be transurethral resection, segmental resection, and partial
completed in a primary setting. Total anterior urethrec- penectomy to total penectomy with perineal urethros-
tomy is also performed for recurrent transitional cell tomy. Tumors of the bulbomembranous and prostatic ure-
cancer of the anterior urethra. The details of the proce- thra are generally advanced at the time of presentation and
Chapter 41 Urethrectomy 689

Figure 41-5 Mobilization of urethra off corpus cavernosa


facilitated using a Penrose drain. (From Graham SD Jr (ed):
Glenn’s Urologic Surgery, 5th edition, pp 461–466.
Philadelphia, Lippincott, Williams and Wilkins, 1998.)

Figure 41-3 Perineal skin incision for urethrectomy. (From


Hinman F, et al: Atlas of Urologic Surgery, 2nd edition.
Amsterdam, Elsevier Science, 1998.)

Figure 41-6 Use of electrocautery to dissect the urethra from


Figure 41-4 Division of bulbocavernous muscle and the corpus cavernosum. (From Hinman F, et al: Atlas of
exposure of urethra. (From Graham SD Jr (ed): Glenn’s Urologic Surgery, 2nd edition. Amsterdam, Elsevier Science,
Urologic Surgery, 5th edition, pp 461–466. Philadelphia, 1998.)
Lippincott, Williams and Wilkins, 1998.)
690 Part VII Urethra and Penis

Figure 41-7 Distal dissection of the urethra resulting in inversion of the penis. (A from Hinman F, et al: Atlas of Urologic
Surgery, 2nd edition. Amsterdam, Elsevier Science, 1998; B courtesy Sanjaya Kumar, M.D.)

are treated with total penectomy and urethrectomy,


Urethrectomy for Small Distal Tumors
prostatectomy and continent cutaneous urinary diver-
sion. It has recently been proposed that locally advanced The patient is placed in a lithotomy position.
male anterior urethral carcinoma be treated with multi- Trendelenburg position helps better visualization of the
modal treatment, including chemoradiation and extirpa- anterior vaginal wall. A Foley catheter is inserted in the
tive surgery.9 urethra and a weighted speculum is placed in the vagina.
The labia are retracted with silk sutures (Figure 41-13).
The urethral meatus is circumscribed with a purse
Urethrectomy in the Female
string sutures. A circumferential incision is made
Primary urethral cancer in women is rare. Although around the urethra (Figure 41-14). Palpation of the
women have a lower incidence of transitional cell cancer Foley catheter allows the urethra to be dissected out in
than men, urethral cancer per se is 4 times more common a plane between the full thickness of the urethra and the
in women than men. The common urethral cancers surrounding tissues. The anterior vaginal wall may be
include squamous cell (60%), transitional cell (20%), and incorporated en bloc in the posterior dissection to
adenocarcinoma (15%). Primary transitional cell cancer obtain negative margin. Proximal dissection of the ure-
of the urethra is very rare. In patients with transitional thra is complete if a reasonable margin (5 mm) proximal
cell cancer of the bladder a total urethrectomy is usually to the tumor is achieved. The urethra is tied with a silk
performed at the time of the cystectomy, unless the suture to prevent tumor spillage and the distal urethra
patient is a candidate for an orthotopic neobladder. transected (Figure 41-15). Negative margins are con-
Distal urethrectomy is performed in patients with tumors firmed by frozen section. The proximal cut edge of the
of the distal one-third of the urethra, preferably for urethra is now approximated with 3-0 absorbable
tumors close to the meatus. sutures to the vaginal mucosa to create a neoorifice
Chapter 41 Urethrectomy 691

Figure 41-8 A, Incision used for dissection of the urethral Figure 41-10 Dissection of the proximal urethra at the
meatus and the glandular urethra (B). Note that the penis has urogenital diaphragm. (From Brendler CB, Schlegel PN,
been placed in its normal anatomic position. Walsh PC: J Urol 1990; 144:270–273.)

(Figure 41-16). The anterior vaginal wall is also closed


with absorbable sutures. The proximal urethra and the
vaginal mucosa may need to be mobilized to achieve
a tension-free anastomosis. A Foley catheter is left
indwelling for 7 days and a vaginal pack for about 24 hours.

Treatment of Larger Distal or Proximal Urethral


Tumors
Extensive dissection of the mid to proximal urethra can
result in incontinence. If the tumor is not involving the
bladder neck, then a distal urethrectomy is performed,
including a cuff of the bladder neck. The bladder is pre-
served, augmented, and a continent cutaneous urinary
diversion performed. The patient will need to catheterize
the augmented continent bladder. This approach is
employed for squamous cell carcinoma and adeno carci-
noma of the urethra. Bladder preserving techniques can-
not be performed for transitional cell cancer, as they are
Figure 41-9 Anatomy and ligation of bulbar urethral arteries.
a field defect change mandating a cystectomy.
A, B, Ligation of bulbar urethral arteries with hemoclips. C,
Lateral view shows relationship between internal pudendal
The patient is positioned in a lithotomy position.
and bulbar arteries, and ischium and inferior ramus of pubis. There are 3 steps to the operation. The first step includes
The bulbar arteries should not be fulgurated to prevent injury an abdominal part where the bladder neck and proximal
to internal pudendal arteries from which they arise and which urethra are dissected. However, if there is involvement of
provide arterial supply to corpora cavernosa. (From Brendler the bladder neck by tumor, a radical cystectomy may be
CB, Schlegel PN, Walsh PC: J Urol 1990; 144:270–273.) necessary. Rarely, resection of the pubic ramus, vulva,
692 Part VII Urethra and Penis

Figure 41-11 A, Completed proximal urethrectomy. Dissection of bulbar urethra is completed without difficulty, because the
membranous urethra has previously been mobilized through the pelvis. B, Specimen: total anterior urethra, including fossa
navicularis. (A from Brendler CB, Schlegel PN, Walsh PC: J Urol 1990; 144:270–273; B courtesy Sanjaya Kumar, M.D.)

Figure 41-12 Closure of the perineal incision and placement Figure 41-13 Exposure of urethra and vagina.
of the Jackson-Pratt drain. [From Walsh PC (ed): Campbell’s
Urology, 6th edition. Philadelphia, WB Saunders, 1992.]
Chapter 41 Urethrectomy 693

Figure 41-14 Placement of suture around urethral meatus Figure 41-15 Transection of distal urethra.
and incision of urethra and anterior vaginal wall.

and pelvic tissues may be necessary. In the face of lymph


node involvement or extensive tumor, given the poor
prognosis, only palliative procedures should be per-
formed. The perineal dissection is next. It is similar but
more extensive than that for distal urethrectomy.
Sometimes, a more extensive dissection of the anterior
vaginal wall is necessary to achieve complete tumor
resection. If the anterior vaginal tissue cannot be suffi-
ciently mobilized to close the defect, gracilis flaps can be
raised to close the defect. The third stage is closure of the
bladder neck, bladder augmentation, and creation of a
continent reservoir. Details of these procedures have
been described elsewhere in this book.

SUMMARY
Urethrectomy is well tolerated in both males and Figure 41-16 Creation of neourethral orifice and closure of
females. Postoperative edema and ecchymosis is common anterior vaginal wall.
but obtaining meticulous hemostasis and anatomic clo-
sure can minimize postoperative hematomas and infec-
tion. Overall cosmetic results are excellent. Urethral REFERENCES
stenosis in females can be easily managed with dilations.
Cancer control depends on the histology, stage, and 1. Naitoh J, Aronson WJ, DeKernion JB: Urethral cancer in
grade of the tumor. In general, urethrectomy alone women. In Graham SD Jr (ed): Glenn’s Urologic Surgery,
results in excellent cure rates for low stage and low-grade 5th edition, pp 461–466. Philadelphia, Lippincott,
tumors. The cure rate for more advanced tumors is lim- Williams and Wilkins, 1998.
ited despite extensive surgery.
694 Part VII Urethra and Penis

2. Kakizoe T, Tobisu K: Transitional cell cancer of the 7. Brendler CB, Schlegel PN, Walsh PC: Urethrectomy
urethra in men and women associated with bladder with preservation of potency. J Urol 1990; 144:270–273.
cancer. Jpn J Clin Oncol 1998; 28(6):357–359. 8. Gheiler EL, Tfilli MV, Tiguert R, et al: Management of
3. Hardeman SW, Soloway MS: Urethral recurrence primary urethral cancer. Urology 1998; 52(3):488–493.
following radical cystectomy. J Urol 1990; 144:666–669. 9. Dalbagni G, Zhang ZF, Lacombe L, Herr HW: Male
4. Levinson KA, Douglas EJ, Wishnow KI: Indications for urethral carcinoma: analysis of treatment outcome.
urethrectomy in an era of continent urinary diversion. Urology 1999; 53(6):1126–1132.
J Urol 1990; 144:73–75. 10. Aronson WJ, Naitoh J, DeKernion JB: Carcinoma of the
5. Stenzl A, Draxl H, Posch B, et al: The risk of urethral male urethra. In Graham SD Jr (ed): Glenn’s Urologic
tumors in female bladder cancer: can the urethra be used Surgery, 5th edition, pp 467–474. Philadelphia,
for orthotopic reconstruction of the urinary tract? J Urol Lippincott, Williams and Wilkins, 1998.
1995; 153:950–955.
6. Freeman JA, Tarter TA, Esrig D, et al: Urethral
recurrence in patients with orthotopic ileal neobladders. J
Urol 1996; 156:1615–1619.
C H A P T E R

42Squamous Cell Carcinoma of the


Penis: Diagnosis and Staging
Kevin R. Loughlin, MD, MBA

INCIDENCE
These authors reported that the risk of penile cancer
The incidence of squamous cell carcinoma of the penis among men who reported a history of genital warts was
varies widely according to geographic distribution. In the 5.9 times that of men who reported no such history.6
United Sates and most industrialized countries, squa- Beyond the link between a history of condylomata and
mous cell carcinoma of the penis is a rare malignancy, subsequent penile cancer, there is strong molecular bio-
accounting for <1% of all male cancers.1,2 The incidence logic evidence demonstrating an association between
in the United States has been estimated to be 1 to 2 cases human papillomavirus (HPV) infection and subsequent
per 100,000 males per year.2,3 However, in societies that penile cancer. Iwasawa et al.8 examined 111 untreated
are more agrarian and where circumcision is less com- penile carcinomas retrospectively and found 70 to be
mon, the incidence of squamous cell cancer of the penis positive for HPV DNA by polymerase chain reaction. In
is much higher. Reddy et al.4 have reported that penile situ hybridization analysis found the HPV to be localized
cancer comprises 16.7% of all cancer in parts of India, in the nuclei of the malignant cells.
and Dodge and Linsell5 have reported that penile cancer Similar findings have been published by Scinicariello
accounts for 12.2% of all cancers diagnosed in Uganda. et al.9 They demonstrated that HPV 16 DNA was incor-
porated into the host’s genome in a primary squamous
cell carcinoma and its lymph node metastases using
EPIDEMIOLOGY
Southern blot analysis and two-dimensional gel elec-
Squamous cell carcinoma of the penis is virtually trophoresis. These findings suggest a causal relationship
unknown in populations where neonatal circumcision is between HPV and penile squamous cell carcinoma.
routinely practiced. Maden et al.6 have reported that the Other reports have shown a similar relationship between
risk for penile cancer was 3.2 times greater in men who HPV 16 and cervical cancer.10 Such relationships raise
were never circumcised and 3.0 times greater in men who the question of whether penile and cervical carcinomas
underwent circumcision after the neonatal period. These may, in some instances, be considered venereal disease.11
authors also identified a 2.8-times-increased risk for
penile cancer among current smokers compared to men
DIFFERENTIAL DIAGNOSIS
who never smoked.6
Penile squamous cell carcinoma most typically occurs The clinician must be aware of several pathologic condi-
in men in the sixth and seventh decades of life; however, tions of the penis that may mimic squamous cell carci-
it can occur in men below the age of 40.3 Racial predilec- noma. Buschke-Löwenstein tumors may grossly be
tion has not been clearly proven, and any observed racial indistinguishable from squamous cell carcinoma.
differences are more likely due to socioeconomic or envi- Buschke-Löwenstein tumors also resemble condyloma
ronmental factors.7 acuminatum and have been referred to as “giant condy-
There appears to be an infectious component to the loma.” However, they differ in that Buschke-Löwenstein
etiology of squamous penile cancer. Maden et al.6 have tumors can invade and penetrate tissue, whereas condy-
reported a strong correlation between a history of geni- loma acuminatum cannot. Despite the ability to invade
tal warts and subsequent development of penile cancer. locally, Buschke-Löwenstein tumors rarely metastasize.

695
696 Part VII Urethra and Penis

However, as with condylomata acuminatum and squa- nodes, lung, and bone. Cavernosography, lymphangiog-
mous cell carcinoma, the etiology of some Buschke- raphy, computed tomography (CT), ultrasonography,
Löwenstein tumors appears to be viral. HPV types 6 and and magnetic resonance imaging (MRI) have all been uti-
11 have been identified in some of these lesions.12 lized to stage the local lesion and metastatic extent of
Erythroplasia of Queyrat and Bowen’s disease are both penile cancer. Despite reports of its accuracy, caver-
considered carcinoma in situ of the penis. Erythroplasia nosography is normally not performed because of the
was first described by Queyrat13 as a red, velvety lesion invasive nature of the technique.22
found on the glans penis or prepuce. The lesion may be Horenblas et al.23 examined the role of lymphangiog-
ulcerative or painful. Histologically, the mucosa appears raphy, CT, and fine-needle aspiration in penile cancer
to be replaced by atypical, hyperplastic cells with hyper- staging. In 98 patients with penile cancer, they found that
chromatic nuclei and mitotic figures. CT scan provided accurate local staging in 74% of the
Bowen’s disease refers to an intraepithelial neoplasm patients. Lymphangiography was utilized in 19 patients
of the skin associated with a high occurrence of subse- to assess regional (inguinal and pelvic) lymph node status.
quent internal malignancy. 14 However, the terms All 6 patients with pathologically proven negative nodes
“Bowen’s disease of the penis” and “erythroplasia of had negative lymphangiograms; however, of the 13
Queyrat” are used interchangeably. Visceral disease is patients with pathologically proven positive nodal
not associated with carcinoma in situ of the penis. HPV involvement, 9 had negative lymphangiograms and 4
has also been identified in carcinoma in situ of the were positive, for a sensitivity of 31% and a specificity of
penis.15 100%. Fine-needle aspiration was used to evaluate the
inguinal nodes only in 18 patients. In this cohort, aspira-
tion cytology had a sensitivity of 71% and a specificity of
PRESENTATION
100%.
Squamous cell carcinoma of the penis occurs almost The use of ultrasound in the staging of primary penile
exclusively in uncircumcised men. The cancer typically cancer was first reported in 2 patients in 1989.24 A larger
begins as a small lesion on the glans. The primary lesion experience was reported by Horenblas et al.25 In their
may be either exophytic or flat and ulcerative. The local group of 16 patients, accurate assessment of the depth
lesion can grow to invade the entire glans, shaft, and cor- and extent of the primary lesion was only achieved in 7
pora. The only reliable means of diagnosis is biopsy and patients (44%). Ultrasound was most limited in differen-
histologic examination. No definite therapy, surgical or tiating invasion of subepithelial tissue from corpus spon-
medical, should be instituted prior to histologic diagno- giosum in the glans. However, determination of invasion
sis. It is not unusual for many of these patients to delay into the corpus cavernosum was clearly demonstrated in
seeking medical attention, and it has been reported that all cases where it was present.
15% to 50% of patients have the penile lesion for >1 year MRI has also been used to stage penile carcinoma.
prior to diagnosis.16,17 Pain is usually not a presenting deKervilier et al.26 reported their preliminary experience
complaint, although weakness, weight loss, and fatigue with MRI and staging penile cancer. They found that
can be present as a result of chronic suppuration. MRI correctly staged local tumors in 7 of 9 cases. T2-
The physical examination should note the size and weighted sequences were most useful, and they recom-
extent of the local lesion. In addition, there should be mended that spin-echo T2-weighted sequences should
careful palpation of the inguinal areas to determine if be used in evaluating such patients.
adenopathy is present. Aspiration of nodes under either fluoroscopic or com-
puted tomographic guidance has been reported as an
additional staging technique. However, like other biopsy
LABORATORY STUDIES AND STAGING
techniques, false negatives can occur.27
There are no tumor markers for penile carcinoma. In addition to radiographic staging of penile cancer,
However, several investigators have reported an associa- Cabanas28 introduced the concept of “sentinel node
tion between penile carcinoma and hypercalcemia.18,19 biopsy.” This concept was based on the belief that penile
Sklaroff and Yagoda19 reported that 17 of 81 patients cancer first spreads to a group of nodes located supero-
with penile carcinoma treated at Memorial Sloan- medial to the junction of the saphenous and femoral
Kettering were hypercalcemic. Hypercalcemia seemed to veins in the area of the superficial epigastric vein.
be related to the bulk of the disease and may resolve with However, subsequent reports29–31 have failed to confirm
surgical excision of inguinal metastases.20 Parathormone- the efficacy of the sentinel node biopsy and such a stag-
like substances can be produced by the primary tumor or ing procedure is no longer recommended. Finally, in
metastases.21 addition to staging the local lesion and regional lymph
The most common sites of metastatic spread of penile nodes, all patients with penile cancer should have a base-
cancer are the inguinal lymph nodes, the pelvic lymph line chest x-ray and bone scan.
Chapter 42 Squamous Cell Carcinoma of the Penis 697

A more recent staging system that incorporates the


STAGING SYSTEMS
histologic degree of differentiation and the extent of local
Two major staging systems have been commonly invasion of the primary tumor has been recently pro-
employed to stage penile cancer. The Jackson system, posed by Heyns et al.33 Their staging system appears in
introduced in 1966,32 is the oldest system used (Table Table 42-3. It provides a predictive distinction between
42-1). A newer system, the TNM classification, is widely T1 and T2-4 tumors and indicates that lymphadenec-
accepted and is now more commonly used than the tomy can be avoided in T1 tumors but should be per-
Jackson system (Table 42-2). formed in all patients with T2 to T4 tumors.

Table 42-1 Classification for Carcinoma of the Penis Table 42-3 Heyns Classification of Penile
Stage I (A) Tumors confined to glans, prepuce, or both Carcinoma-Primary Lesion
T1 Grade 1–2 with invasion through dermis
Stage II (B) Tumors extending onto shaft of penis
T2 Any grade with invasion through the corpus
Stage III (C) Tumors with inguinal metastasis that spongiosum or cavernosum
are operable
T3 Any grade with invasion of the urethra
Stage IV (D) Tumors involving adjacent structures;
tumors associated with inoperable inguinal T4 Grade 3, regardless of invasion
metastasis or distant metastasis

Table 42-2 TNM Classification of Penile Carcinoma SUMMARY


Primary tumor (T) Penile cancer is a rare malignancy in regions where cir-
cumcision is routinely practiced. A careful physical exam-
Tx Primary tumor cannot be assessed
ination and radiographic imaging are necessary to stage
T0 No evidence of primary tumor local and distant involvement of the tumor. A biopsy and
pathologic confirmation of the diagnosis must be
Tis Carcinoma in situ obtained prior to initiating treatment.
Ta Noninvasive verrucous carcinoma

T1 Tumor invades subepithelial connective tissue REFERENCES


T2 Tumor invades corpus spongiosum or cavernosum
1. Grabstald H: Cancer of the penis. J Cont Ed Urol 1979;
T3 Tumor invades urethra or prostate 18:15.
2. Sufrin G, Huben R: Benigh and malignant lesions of the
T4 Tumor invades other adjacent structures penis. In Gillenwater JY, Grayback JT, Howard SS,
Duckett JW (eds): Adult and Pediatric Urology, 2nd
Regional lymph nodes (N) edition, pp 1643–1681. St. Louis, Mosby, Year Book, 1991.
3. Schellhammer PF, Jordon GH, Schlossberg SM:
Nx Regional lymph nodes cannot be assessed Tumors of the penis. In Walsh PC, Retik AB, Stamey TA,
Vaughan ED Jr (eds): Campbell’s Urology, 6th edition,
N0 No regional lymph node metastasis
pp 1264–1298. Philadelphia, WB Saunders Co, 1992.
N1 Metastasis in a single superficial, inguinal lymph node 4. Reddy CRRM, Raghavaigah NV, Mouli KC: Prevalence
of carcinoma of the penis with special reference to India.
N2 Metastases in multiple or bilateral superficial Int Surg 1978; 60:470.
inguinal lymph nodes 5. Dodge OG, Linsell CA: Carcinoma of the penis in
Uganda and Kenya Africa. Cancer 1963; 16:1255.
N3 Metastasis in deep inguinal or pelvic lymph node(s) 6. Maden C, Sherman KJ, Beckmann AM, et al: History of
unilateral or bilateral circumcision, medical conditions, sexual activity and risk
of penile cancer. J Natl Cancer Inst 1993; 85:19.
Distant metastases (M) 7. Hall NEL, Schottenfeld D: Penis. In Shottenfeld D,
Fraumens JF Jr (eds): Cancer Epidemiology and
Mx Distant metastasis cannot be assessed Prevention, p 964. Philadelphia, WB Saunders, 1982.
8. Iwasawa A, Kumanoto Y, Fujnaga K: Detection of human
M0 No distant metastases
papilloma virus deoxyribonucleic acid in penile carcinoma
M1 Distant metastases by polymerase chain reaction and in situ hybridization.
J Urol 1993; 149:59.
698 Part VII Urethra and Penis

9. Scinicariello F, Rudy P, Saltzstein D, et al: Human 22. Raghavaiah NV: Corpus cavernosogram in the evaluation
papilloma virus 16 exhibits a similar integration pattern in of carcinoma of the penis. J Urol 1978; 120:423.
primary squamous cell carcinoma of the penis and its 23. Horenblas S, Van Tinteren H, Delemarre JF, et al:
metastasis. Cancer 1992; 70:2143. Squamous cell carcinoma of the penis: accuracy of tumor,
10. Rohan T, Mann V, McLaughlin J, et al: PCR detected nodes and metastasis classification system and role of
genital papilloma virus infection: prevalence and lymphangiography, computerized tomography scan and
association with risk factors for cervical cancer. Int J fine needle aspiration cytology. J Urol 1991; 146:1279.
Cancer 1991; 49:856. 24. Yamashita T, Ogawa A: Ultrasound in penile cancer. Urol
11. Loughlin KR: Penile cancer. Curr Opin Urol 1993; 3:415. Radiol 1989; 11:174.
12. Boshart M, Zor Hausen H: Human papilloma virus (HPV) 25. Horenblas S, Kroger R, Gallee MPW, et al: Ultrasound in
in Buschke-Löwenstein tumors: physical state of the DNA squamous cell carcinoma of the penis: a useful addition to
and identification of a tandem duplication in the non- clinical staging. Urology 1994; 43:702.
coding region of a HPV 6 subtype. J Virol 1986; 58:963. 26. deKervilier E, Ollier P, Desgrandchamps F, et al:
13. Queyrat L: Erythroplasia du gland. Sec Franc Dermatol Magnetic resonance imaging in patients with penile
Syphilol 1911; 22:378. carcinoma. Br J Radiol 1995; 68:704.
14. Bowen J: Precancerous dermatoses: a review of two cases 27. Scappini P, Piscioli F, Pusiol T, et al: Penile cancer
of chronic atypical epithelial proliferation. J Cutan Dis aspiration biopsy cytology for staging. Cancer 1986;
1912; 30:241. 58:1526–1533.
15. Pfister H, Haneke E: Demonstration of human papilloma 28. Cabanas R: An approach to the treatment of penile
virus type 2 DNA in Bowen’s disease. Arch Dermatol Res carcinoma. Cancer 1977; 39:456.
1984; 276:123. 29. Catalona WJ: Role of lymphadenectomy in carcinomas of
16. Hardner J, Bhanalaph T, Murphy GP, et al: Carcinoma of the penis. Urol Clin North Am 1980; 7:785.
the penis: analysis of therapy in 100 consecutive cases. 30. Perinetti EP, Crane DC, Catalona WJ: Unreliability of
J Urol 1972; 108:428. sentinel lymph node biopsy for staging penile carcinoma.
17. Gursel EO, Georgourtzos C, Uson AC, et al: Penile J Urol 1980; 124:734.
cancer. Urology 1973; 1:569. 31. Wespes E, Simon J, Schulmann CC: Cabanas’ approach:
18. Rudd FV, Rott RK, Skoglund RW, Ansell JS: Tumor Is sentinel node biopsy reliable for staging of penile
induced hypercalcemia. J Urol 1972; 107:986. carcinoma? Urology 1986; 28:278.
19. Sklaroff RB, Yagoda A: Penile cancer: natural history and 32. Jackson SM: The treatment of carcinoma of the penis.
therapy. In Chemotherapy and Urological Malignancy, pp Br J Surg 1966; 53:33.
98–105. New York, Springer, 1982. 33. Heyns CF, VanVollenhaven P, Steenkamp JW, Allen FJ,
20. Block NL, Rosen P, Whitmore WF: Hemipelvectomy for Van Velden DJ: Carcinoma of the penis – appraisal of a
advanced penile cancer. J Urol 1973; 110:703. modified tumor-staging system. Br J Urol 1997;
21. Malakoff AF, Schmidt JD: Metastatic carcinoma of penis 80:307–312.
complicated by hypercalcemia. Urology 1975; 5:519.
C H A P T E R

43 Superficial Carcinoma of the Penis:


Management and Prognosis
Jay S. Belani, MD, and Gerald L. Andriole, MD

Penile cancer is a rare entity in developed nations. In the ventrally. Buck’s fascia provides a dense layer of covering
United States only 0.3% of all cancers diagnosed are around the corpora. Superficial to Buck’s fascia lie the
malignancies of the penis.1 The annual incidence rate in superficial penile fascia and skin.
the U.S. and Europe is about 0.5 to 1 per 100,000 men,1 The arterial supply of the penis is derived from
which has remained constant over the past 5 decades.2 In branches of the penile artery, which arises from the
other countries the incidence varies based on hygienic pudendal artery. The penile artery divides into the bul-
standards and culture. The incidence is much higher in bourethral artery, deep artery (AKA: cavernosal artery),
developing countries, such as Mexico, China, India, and and dorsal penile artery.5 Three venous systems drain the
many African countries. In addition, as socioeconomic penis: the superficial, intermediate, and deep.6 The
conditions improve in developing countries, the rates of superficial system drains the skin of the dorsal penis and
cancer have declined. prepuce. The intermediate system is made up of the deep
Cancer of the penis usually occurs in elderly men. The dorsal veins and circumflex veins, which drain into
rate of cancer abruptly increases at about age 60 and Santorini’s plexus along the prostate. The deep system
peaks at 80 years old.1,3 This trend seems to be evident consists of the cavernosal and crural veins, which are the
worldwide; however, those countries with a higher inci- main drainage system of the corpora cavernosa and drain
dence of the disease seem to have an earlier age of onset. into the internal pudendal vein.
In the U.S., blacks are more commonly diagnosed than Penile lymph vessels drain into one of two inguinal
whites by about 2:1.1 However, decreased rates of cir- nodal groups: superficial or deep. In general, most lym-
cumcision and a lower socioeconomic status may be con- phatic channels from the body of the penis and prepuce
founding factors in the statistics.1,2 travel to the superficial inguinal nodes. The glans may
The most common malignancy of the penis is squa- drain into the superficial or deep inguinal nodes.5
mous cell carcinoma (SCC), accounting for about 95% of More proximal lymphatic drainage generally occurs in
cases.3,4 Other rare neoplasms include malignant a stepwise fashion. The superficial nodes drain into the
melanoma, basal cell carcinoma, sarcoma, Paget’s disease, deep inguinal nodes, which, in turn, drain into the pelvic
lymphoreticular malignancy, and metastasis from other lymph nodes. Lymphatic drainage is not uniform and sig-
sources. In addition, transitional cell carcinoma (TCC) of nificant cross-communication occurs. Hence, tumors
the urethra can involve the penis by local spread. may spread to ipsilateral, contralateral, or bilateral lymph
node chains (Figures 43-1 and 43-2).7
ANATOMY
SQUAMOUS CELL CARCINOMA
Knowledge of penile anatomy is essential in compre-
Presentation, Signs and Symptoms
hending the pathophysiology, staging, and management
options of penile cancer. The penis is made up of two Carcinoma of the penis may present as an area of ery-
corpora cavernosa dorsally and the corpus spongiosum thema, induration, ulceration, or nonhealing sore. Some

699
700 Part VII Urethra and Penis

Superficial dorsal vein About 50% of patients will present with inguinal lym-
Deep dorsal vein Dorsal artery
phadenopathy.11
Physical examination is important in the diagnosis of
penile cancer. Size, location, fixation, and extent of cor-
poral body involvement must be evaluated. Both inguinal
regions should be palpated for lymphadenopathy and the
Buck’s fascia
penile base and scrotum should be inspected.10
Rarely, the primary tumor may be concealed by the
Cavernosal artery prepuce, and a patient may present with inguinal suppu-
Urethra Corpus cavernosum ration, ulceration, or hemorrhage as a result of lymph
node metastasis. Other rare presentations are urinary fis-
Corpus sponglosum tula, urinary retention, and destruction or self-amputa-
tion of the phallus.
Figure 43-1 Cross-sectional diagram of penile shaft.
Penile carcinoma exhibits two growth patterns: fun-
gating and exophytic, or ulcerative and infiltrative.12,13
Fungating tumors usually present as a papillary verrucous
lesion on the glans or prepuce. Ulcerating tumors pres-
ent as a small, erythematous ulcer, usually on the glans.
As ulcerating lesions grow they infiltrate into deeper tis-
sues and are more likely to metastasize than fungating
tumors. Lateral destruction occurs as the tumor involves
the deeper tissues.

NATURAL HISTORY
SCC usually begins as a superficial growth along the
glans or prepuce and spreads locally. As it grows, Buck’s
fascia provides a strong barrier for deeper infiltration;
however, if left untreated, the tumor will erode into the
corpus cavernosum. Hematogenous spread usually does
not occur despite the extensive vascular network within
the corpus cavernosa. Fistula formation may result with
urethral involvement. Metastasis to the superficial
inguinal nodes is more likely to occur if the prepuce is
initially involved, while metastasis to both the superficial
and deep inguinal nodes occurs with glans involvement.
Spread then goes to the pelvic nodes. Bilateral or cross-
inguinal lymph node involvement is possible given the
rich anastomoses between lymphatics in the subcutaneous
Figure 43-2 Diagram of penile lymphatic drainage. tissue that cross the midline.13
At presentation about 50% of patients have inguinal
patients experience persistent bleeding or eventual lymphadenopathy on examination. About 55% of these
sloughing and self-amputaion.10 About 47% present with patients have inflammatory inguinal lymphadenopathy
a mass or nodule, 35% with an ulcer, and 17% with an from infection of the primary tumor, while 45% have
inflammatory lesion or bleeding.11 Only 0.7% of cases metastatic tumor.11,13 Less than 2% have visceral spread
are diagnosed incidentally on a circumcision specimen. of disease at presentation.13
The site of origin is more commonly distal, with 48%
occurring on the glans, 21% on the prepuce, 9% in both
DIAGNOSIS
locations, and 6% along the coronal sulcus.11 Direct
extension of the distal tumor to the shaft occurs in 14% Lynch and Krush showed that patients with penile cancer
of cases.11 delay seeking medical attention longer than patients with
A delay in diagnosis between the initial finding of a other cancers.14 Gursel and Hardner each found that
penile lesion and presentation to the physician is typically between 15% and 50% of patients delay diagnosis for
between 3 and 26 months, with an average of 10 months greater than 1 year.15,16 Moreover, Hardner showed a
and 60% of lesions are larger than 2 cm at diagnosis. worse prognosis for patients with delayed presentation.15
Chapter 43 Superficial Carcinoma of the Penis: Management and Prognosis 701

Given the fact that most tumors begin as superficial grades 1, 2, and 3 tumors is between 0% to 29%, 26% to
lesions, and that many patients delay presentation, it is 65%, and 80% to 100%, respectively.20–23
vitally important to biopsy all suspicious lesions at time A second histologic classification system has been pro-
of presentation, especially if the lesion fails to resolve posed by Cubilla et al,24 which divides penile cancers into
after a short course of conservative treatment. The four categories: superficial spread, vertical growth, verru-
biopsy is done to confirm the diagnosis of carcinoma and cous, and multicentric. Table 43-2 describes each type.
determine the extent of invasion. If the lesion is small, an Villavicencio et al.20 studied 81 patients and defined
excisional biopsy may be performed. If it is large, an inci- prognostic values with each category. About 70% of the
sional biopsy, taken from the periphery to include normal patients evaluated had a superficial spreading SCC.
epithelium, is adequate. About 66% of these patients were found to be disease
If the lesion is suspected to be hidden by a phimotic free at an average of 61 months after treatment, and 35%
foreskin, then a preputial slit should be performed. There were found to have lymphatic spread. Vertical growth
have been no reported cases of spread as a result of the implies a worse prognosis. About 90% of these tumors
dorsal slit or biopsy.10,17 Lastly, in some cases, circumcision are usually high-grade lesions, and only 30% are disease
may be done to obtain an adequate tissue sample. free at 5 years. Verrucous growth tumors usually are of
low grade and all cases have been found to be disease free
at 5 years follow-up.
GRADING AND STAGING
Histology
Staging
Histologically, SCC of the penis has been categorized by
extent of malignant transformation, i.e., carcinoma in Staging of penile SCC was introduced by Jackson in
situ (CIS) and invasive SCC. CIS represents a full thick- 196625 and is shown in Table 43-3. Stage I lesions are
ness of atypical epithelial squamous cells. Both Bowen’s confined to the glans or prepuce. Stage II lesions extend
disease and erythroplasia of Queryat represent premalig- along the penile shaft. Stage III tumors involve the
nant lesions that have this growth pattern. In addition,
CIS is often found in association with invasive SCC.18
Two classification systems, described by Broder and Table 43-2 Classification for SCC of the Penis
Cubilla, for invasive SCC have been proposed. Broder’s Category Description
classification of SCC of the skin has been applied to penile
lesions. Table 43-1 summarizes the system. Increasing Superficial spreading Centrifugal growth usually
restricted to superficial layers
grade has been associated with an increased risk of inguinal
metastasis. The chance of spread to inguinal nodes for Vertical growth Spread is vertical with little radial
growth

Verrucous Usually large, exophytic lesions


Table 43-1 The Broder’s Classification System For
Grading SCC Multicentric Two or more foci of histologic
Grade Histology spread

I Cells well differentiated with keratinization From Cubilla AL, Barreto J, Caballero C, et al: Am J Surg Pathol
1993; 17:753.
Keratin pearls

II–III Increased nuclear atypia and mitotic activity Table 43-3 Jackson Staging System
Fewer keratin pearls Stage Description

IV Markedly increased nuclear pleomorphism I Tumor confined to glans or prepuce


and mitotic activity
II Invasion into shaft or corpora; no nodal or distant
Necrosis metastasis

No keratin pearls III Tumor confined to penis; operable inguinal nodal


metastasis
Lymphatic and perineural invasion
IV Tumor involves adjacent structures; distant
Deeply invasive metastasis and/or inoperable inguinal nodes

From Broders AC: Ann Surg 1921; 73:141. From Jackson SM: Br J Surg 1966; 53:33.
Chapter 43 Superficial Carcinoma of the Penis: Management and Prognosis 703

spread beyond the foreskin requires a more aggressive cases, the patient may be better served by a total penec-
resection. More proximal lesions may require a more tomy and perineal urethrostomy.34
extensive resection and multiple biopsies of the margins Studies have shown a local recurrence rate up to 19%
are warranted to rule out residual disease.8 Moreover, in with a 2-cm proximal tumor-free margin.42 McDougal32
patients with severe phimosis or chronic balanoposthitis, showed that 90% of patients with superficial, local dis-
Huben recommends 5000 cGy of radiation therapy eases will be cured after partial penectomy. Moreover,
over 5 weeks to eradicate any occult disease after Hardner et al.15 illustrated that partial penectomy is as
circumcision.34 effective as total penectomy as long as an adequate mar-
gin is obtained.
PARTIAL PENECTOMY
Complications
Lesions of the glans penis can be managed with partial
penectomy as long as a 1.5- to 2.0-cm margin is obtained Complications of partial penectomy include infection,
to limit local recurrence.8,37,38 Simple wedge resection hemorrhage, and stenosis of the urethral meatus.
has a higher recurrence rate. Local excision is associated Infection can occur as a result of spread of bacteria from
with a 40% to 50% recurrence rate and is not adequate the perineum or inoculation from the primary tumor
for any T2 or larger lesion.34,38,39 itself. Infection is minimized by perioperative and post-
operative antibiotics covering skin flora. Wound infec-
tions are best managed by open drainage and allowing
Surgical Technique
the area to heal by secondary intention. Bleeding is best
The penis is thoroughly scrubbed with Betadine and the avoided by attention to hemostasis after the tourniquet is
distal penis containing the tumor is covered with a sterile removed. Meatal stenosis is minimized with a wide spat-
surgical glove or condom. A tourniquet is applied to the ulation of the urethra. The problem is best managed with
base of the penis to minimize blood loss. dilation using meatal sounds or meatotomy.40
An incision is made circumferentially 1.5 cm proximal
to the tumor margin at the dorsal aspect and about 2.0 to
TOTAL PENECTOMY
2.5 cm proximal to the tumor at the ventral aspect. The
subcutaneous vessels are coagulated and the superficial Total penectomy is usually reserved for proximal lesions
dorsal vein is ligated and divided. Buck’s fascia is incised where an adequate proximal margin would not be
and the deep dorsal artery and vein are ligated and divided. obtained via partial penectomy. It is the most extensive
The cavernosal bodies are then transected about 1.5 cm means of local surgical excision and has the least risk of
proximal to the tumor margin, and the cavernosal arteries local recurrence, amongst all surgical options.
are ligated. The urethra should be dissected free with a 1-
cm stump left in place distal to the cavernosal bodies.
Surgical Technique
The specimen is then evaluated by frozen section to
exclude tumor at the margins. If tumor is found, then a Preparation of the penis is similar to that of partial penec-
more proximal resection is done. Once resection is com- tomy, and the tumor is covered in a sterile glove or con-
plete, the corpora are closed with a horizontal mattress dom. The anus is draped outside the operative field. A
suture. The tourniquet is loosened and any remaining vertical elliptical incision is made around the base of the
bleeding is controlled. The urethra is spatulated and the penis 2 cm from the tumor margin. Ventrally, Buck’s fascia
excess skin is brought over the exposed cavernosa. The is incised and the corpus spongiosum is separated from the
urethra is anastomosed to a buttonhole in the skin. A corpus cavernosum. The urethra is isolated and divided.
Foley catheter is usually left in place for 5 days while the Dorsally, the suspensory ligament is divided and the deep
anastomosis heals.35,40 dorsal arteries and veins are ligated and divided at the level
of the urogenital sinus. The corpus cavernosa are tran-
sected 2 cm proximal to the tumor margin. The corpora
Results
are then closed in similar fashion as described for partial
Usually patients have excellent control of their urinary penectomy. A 1.5-cm elliptical incision is made in the per-
stream and adequate length for intercourse if at least 2 to ineum and the skin and subcutaneous tissue are excised.
3 cm of the shaft is left in place. Jensen41 showed that The urethra is tunneled through the subcutaneous tissue
45% of patients with a 4- to 6-cm and 25% of patients to this opening, ensuring that it has not kinked or twisted.
with a 2- to 3-cm stump are able to have sexual inter- It is then spatulated and anastomosed to the skin with
course. If too short a phallic stump is left, it may be interrupted absorbable suture. The skin over the penile
engulfed in the suprapubic fat pad and the patient will defect is closed elevating the scrotum anteriorly and an 18
have difficulty voiding in the standing position. In these French silicone catheter is left in place.35,40
704 Part VII Urethra and Penis

Complications excised until no tumor is seen microscopically. Using


microscopic mapping, all tumor margins are inspected
Similar to partial penectomy, complications include
and normal tissue is spared.34,44
infection, hemorrhage, and perineal urethrostomy stric-
ture. Bleeding and wound infection are less common
than with partial penectomy.34 Strictures of the perineal Fixed Tissue Technique
urethrostomy are usually more difficult to manage, and
The technique is similar to the fresh tissue technique; how-
conservative management with intermittent catheteriza-
ever, a fixative paste, zinc chloride, is used to mark the sur-
tion may fail. Surgical revision may be required. Lastly,
face of the specimen. Excision is then performed and
delayed, extended lymphatic drainage has been
processed with horizontal sectioning. The technique is
described, and is usually managed with conservative
repeated until no neoplastic cells are seen. The difference
management.34
is that the patient returns each consecutive day for fixative
application and serial sectioning. The process can take
IMPACT OF TOTAL AND PARTIAL PENECTOMY between 1 to 7 days depending on the extent of the lesion.44
Opjordsmoen and Fossa43 showed that men with radia-
tion therapy had improved sexual function than those Effectiveness
who had undergone partial penectomy. About 83% of
Five-year cure rate is about 74%, which Mohs suggests is
patients had normal or slightly reduced sexual function
an improvement given that most tumors he treated were
after radiation therapy, while 66% of patients had
large and recurrent after other treatment methods.44
severely reduced sexual function after partial penectomy.
Mohs describes an 86% cure rate for stage I tumors, 62%
In addition, the study suggested that those patients who
for stage II, and 0% for stage III disease. These rates are
had local excision with laser therapy had worse sexual
higher than other local treatment options.45 Because of
function than those after radiation therapy. None of the
the inability to cure stage III disease, Mohs’ micro-
patients studied underwent Mohs’ micrographic surgery.
graphic surgery is best reserved for small stages I and II
Despite the impaired sexual function, Opjordsmoen and
lesions.
Fossa43 showed that 7 out of 25 patients preferred a com-
promise in survival to impaired sexual function, while 17
out of 25 preferred long-term survival to penis-sparing Complications
modalities. Hence, when treating patients with SCC of
Risks of Mohs’ surgery are minimal. Infection and bleed-
the penis, all options should be discussed when choosing
ing have been described. In addition, delayed healing can
a course of management.
occur, as 80% of lesions are usually left to heal by sec-
ondary intention.46
MOHS’ MICROGRAPHIC SURGERY
Mohs’ studies showed that SCC of the skin sends out LASER THERAPY
thin sheets of tumor cells along the path of least resist-
ance, like nerve sheaths, blood vessel adventitia, or fascial In 1976, Hofstetter and Frank47 first used laser energy to
planes.44 This explains why some tumors recur after exci- treat penile carcinoma and preserve the phallus. Recent
sion with a clearly visualized negative margin. Hence, studies have suggested that, if done with attention to
Mohs described a technique of maximal tissue sparing meticulous technique, laser therapy provides as good or
and complete tumor excision—Mohs’ micrographic sur- better tumor control compared to partial and total
gery. Studies have shown cure rates and tissue sparing to penectomy without the functional impairment and poor
be superior than other tissue-sparing techniques.44–46 cosmetic result.48,49

Surgical Technique Surgical Technique


Two techniques have been described: fresh tissue tech- The technique of laser excision involves one of two
nique and fixed tissue technique. types of lasers: the Nd:YAG laser or carbon dioxide
laser. The Nd:YAG laser penetrates the tissue and
causes coagulation up to 3 to 4 mm deep. It does not
Fresh Tissue Technique
vaporize much tissue at the surface, and keeps the struc-
The main tumor mass is debulked. Residual tumor is tural integrity intact. On the other hand, the carbon
then serially excised and evaluated microscopically in dioxide laser minimally penetrates into deeper layers. It
horizontal sectioning. The areas of residual disease are is a better cutting laser than the Nd:YAG and creates
mapped and the adjacent areas are excised. All tissue is less of a defect. Hence, studies have suggested that the
Chapter 43 Superficial Carcinoma of the Penis: Management and Prognosis 705

carbon dioxide laser is better for superficial, noninva- within 3 mm of the surface. Moreover, Tietjen and
sive tumors (i.e., CIS), while the Nd:YAG laser is better Malek51 have suggested that laser therapy probably leaves
suited for more invasive disease (i.e., T1 or possibly T2 some dysplastic premalignant tissue behind, hence it is
disease).50 important to follow these patients closely for evidence of
Prior to treatment the tumor site is biopsied to deter- recurrence, and treat any recurrence expeditiously.
mine the depth of penetration and grade of tumor.
Circumcision may be performed if the tumor is on the
RADIATION THERAPY
foreskin or glans. The extent of the lesion is mapped with
5% acetic acid for 20 minutes. The acid helps demarcate The effectiveness of radiation therapy for penile cancer
suspicious areas by staining them white. The laser energy treatment is controversial since there are multiple differ-
is then applied to the marked areas with a 3-mm margin ent modalities and no uniform dosage has been used.
around the perimeter of the tumor site. Most data come from small, retrospective studies.
Proponents of radiation therapy for penile cancer
advocate that it preserves penile function and eliminates
Effectiveness
the distress of possible amputation.54 Surgical excision,
Recurrence rates have ranged between 6% and 26%, either partial or total penectomy, results in significant
depending on the histologic grade and tumor stage. functional and psychologic morbidities.43,54–56 T1 and
Unfortunately, Tietjen and Malek51 suggest that dysplas- T2 penile carcinomas have been cured by radiation ther-
tic tissue may be left behind and up to a 26% of patients apy; however, significant complications can occur,
recur. Overall, though, they found an 8.3% local recur- depending on the type of radiation given.
rence for CIS and a 14.3% local recurrence rate for T1 Radiation therapy can be used for the primary lesion
lesions, compared to a rate as low as 6% and 10% in the via external beam or interstitial therapy. The advantage
literature, respectively (Table 43-5). of radiation therapy is that it provides local control, and
in cases of failure, salvage surgical control may be per-
formed. Moreover, healing after salvage surgery is usu-
Complications
ally not impaired, despite the local radiation.
Complications from laser treatment of penile cancers are In treating the primary lesion, some suggest that all
minimal. Pain at the site may persist especially if infec- patients undergo circumcision to evaluate the extent of
tion develops. Preputial lymphedema has also been the primary tumor and possibly provide debulking.
described in uncircumcised men. Circumcision minimizes some of the complications, by
Frimberger et al.49 showed that all patients treated decreasing the edema and mucosal irritation and reduc-
with laser coagulation were satisfied with the cosmetic ing the risk of secondary infection.57 Ravi et al.58 have
result. Seventy-five percent of patients had functional suggested that circumcision is not necessary if the fore-
erections for intercourse, no patients developed meatal skin can be retracted easily and kept in that position
strictures, and no patient needed psychologic counseling while the radiation is given. However, if phimosis is pres-
as a result of the treatment. ent or the lesion cannot be assessed accurately, then cir-
The disadvantages of laser therapy include the lack of cumcision or a dorsal slit is warranted.
accurate pathologic staging and identification of tumor-
free margins. Instead, a biopsy at the base of excision is
BRACHYTHERAPY
performed to rule out any further extension. Hence, van
Bezooijen et al.50 have argued that laser therapy should Interstitial brachytherapy is the use of radiation sources
only be used for superficial disease or if the depth is implanted within the site of the tumor. The tumor site is

Table 43-5 Local Recurrence after Laser Ablative Therapy


Follow-Up (months) Tumor Stage (n) Recurrence (%)

Tietjen and Malek51 12–117 Tis (17), T1 (14) 8.3, 14.3

Windahl and Hellsten52 8–94 T1 (7), T2 (8) 14, 12

Rothenberger53 13–36 T1(9), T2 (8) 22, 0

Van Bezooijen et al.50 6–75 Tis (19) 26

Frimberger et al.49 7–168 Tis (17), T1(10) 5.8, 10


706 Part VII Urethra and Penis

transfixed with needles containing a radioactive sub- Local control rates for external beam radiation ther-
stance. Historically, this has been radon, however, more apy ranges from 50% to 90%.65 Sarin et al.66 studied 59
recently other radioisotopes, including iridium-192, patients who received external beam therapy and found a
cobalt-60, tantalum-111, and gold-190, have been used. 5- and 10-year local control rate of 51% and 48%,
Usually, a dose of 70 Gy is given over 5 to 7 days. The respectively. McLean et al.67 found a recurrence rate of
technique of implantation involves using a Plexiglas tem- 28% (5 of 18) for T1 and T2 tumors. Thirty-three per-
plate with drilled holes to guide the needles into proper cent of patients required penectomy because of local
position. This ensures accuracy for a homogenous radia- recurrence in 5 patients and complications in 1 patient.
tion dose to the entire tumor bed. Table 43-7 summarizes recent studies and the effective
Brachytherapy has been most successful in T1 and control rate.
T2 diseases. Studies have shown a local recurrence rate Complications include acute skin irritation and
of between 12% and 25%, a penile conservation rate of desquamation and penile soreness or swelling, acutely.
67% to 83%, and a disease-free survival rate of 85% to Long-term complications include urethral stricture and
95% (Table 43-6). 59–63 Most failures occurred in meatal stenosis, which is usually managed with dilation.
lesions >4 cm. Kiltie et al.61 reported a local failure rate Uncommonly, penile necrosis may develop and only a
of 60% for tumors >4 cm and 14% for tumors <4 cm. few patients will have a loss of potency.65,68
Mazeron et al.64 showed a local failure rate of 11% for
tumors <2 cm, 26% for tumors 2 to 4 cm and 50% for
MANAGEMENT OF INGUINAL NODES
tumors >4 cm. Hence, brachytherapy is mainly recom-
mended for tumors <4 cm and superficial or moder- Inguinal lymph node status provides important prog-
ately infiltrating.64 nostic information in managing penile carcinoma.
Complications of brachytherapy include skin necrosis, Overall, those patients without lymph node metasta-
urethritis, urethral stricture disease, pigmentation, or sis have a 5-year survival of about 85%, while those
telangiectasias. Sexual potency is usually preserved.65 with metastasis only have a 50% 2-year survival.11 The
dilemma, however, revolves around the inaccuracy of
determining inguinal node spread. About 50% of
EXTERNAL BEAM RADIATION
patients with palpable inguinal nodes will have metas-
External beam radiation has been used for superficial tasis and about 20% of patients without palpably
invasive carcinoma or CIS. It is used to deliver a low dose enlarged nodes will have spread.15,32,56 These data
of radiation therapy to a superficial area with a 1- to 2 cm incorporate both stage I and stage II diseases. Patients
margin. Gerbaulet and Lambin57 have argued that a low with superficial SCC are unlikely to have inguinal
dose of therapy with a high total dose effectively treats spread. McDougal et al.32 showed that patients with
the lesion and minimizes the risk of fibrosis. For exam- stage I disease approach a 100% survival rate free of
ple, a 2-Gy fraction can be given per day to a total of 65 tumor after local excision alone. Stage II disease is
to 70 Gy. Problems with external beam radiation include more likely to have indolent lymphatic metastasis, and
a prolonged treatment time of 6 to 7 weeks and difficulty an inguinal lymphadenectomy may be prudent.32,33,71
in reproducing the area of radiation. Figure 43-3 provides an algorithm for management of

Table 43-6 Effectiveness of Brachytherapy


Disease-Free
% Local Recurrence Survival (%)
Number of Cases (Years Follow-Up) Penile Conservation (%) (Years Follow-Up)

Crook et al.59 30 15 (2) 83 95 (2)

24 (5) 89 (5)

Kiltie et al.61 31 25 (5) 74.8 85.4 (5)

Rozan et al.62 184 12 (3) 78 88 (5)

15 (5)

Soria et al.63 72 25 82

Delannes et al.60 51 18 67
Chapter 43 Superficial Carcinoma of the Penis: Management and Prognosis 707

Table 43-7 Effectiveness of External Beam Therapy


Sarin et al.66 McLean et al.67 Sagerman et al.69 Fossa et al.70 Grabstald et al.68

Number of patients 59 18 12 11 10

Local control rate (%) 51 (5) 65 66 73 90 (6–10)


(years follow-up)

48 (10)

REFERENCES
Superficial, well-differentiated tumor*

1. Gloeckler-Ries LAHB, Edwards BK (eds): Cancer


Statistics Review 1973–1987, National Cancer Institute,
Palpable nodes Nonpalpable nodes
National Institutes of Health Publication No. 90-2789.
Bethesda, National Institute of Health, 1990.
2. Huben RP: Biology of penile cancer. In Ragavan D, Scher
Treat with antibiotics HI, Leibel SA, et al (eds): Principles and Practice of
for 4−6 weeks Observe** Genitourinary Oncology, pp 921–936. Philadelphia,
Lippincott-Raven, 1997.
3. Lucia MS, Miller GJ: Histopathology of malignant lesions
of the penis. Urol Clin North Am 1992; 19:227.
Nonpalpable nodes Palpable nodes 4. Chesney TM, Murphy WM (eds): Diseases of the Penis
and Scrotum, p 392. Philadelphia, WB Saunders, 1989.
5. Dewire D, Lepor H: Anatomic considerations of the penis
Observe** Lymph node dissection and its lymphatic drainage. Urol Clin North Am 1992;
19:211.
6. Moore KL, Dalley AF: Clinically Oriented Anatomy.
* Primary tumor is T1 after circumcision, partial or total penectomy.
Philadelphia, Lippincott Williams & Wilkins, 1999.
** Exam every 2 months for 2 years, then every 6 months. 7. Johnson DE, Costello AJ: Anatomy of the penis and
groin. In Ragavan D, Scher HI, Leibel SA, et al (eds):
Figure 43-3 Algorithm for management of lymphadenopathy Principles and Practice of Genitourinary Oncology, pp
for superficial penile cancer. 913–920. Philadelphia, Lippincott-Raven, 1997.
8. Bissada NK: Conservative extirpative treatment of cancer
of the penis. Urol Clin North Am 1992; 19:283.
inguinal nodes in patients with low stage SCC of the 9. Clemente CD Anatomy: A Regional Atlas of the Human
penis. Body, 4th edition, p 414. Philadelphia, Lea & Febiger,
1987.
10. Burgers JK, Badalament RA, Drago JR: Penile cancer.
SUMMARY Clinical presentation, diagnosis, and staging. Urol Clin
North Am 1992; 19:247.
Penile cancer rarely occurs in the North America and 11. Sufrin G, Huben RP: Benign and malignant lesions of the
European nations. SCC accounts for 95% of cases penis. In Gillenwater JY, Grayhack JT, Howards SS, et al:
and superficial tumors have an excellent prognosis, Adult and Pediatric Urology, Vol 2, 3rd edition, pp
especially if the patient presents without inguinal lym- 1997–2042. St. Louis, Mosby, 1996.
phadenopathy. 12. Huben RP: Clinical presentation and staging of penile
The gold standard for management includes surgical cancer. In Ragavan D, Scher HI, Leibel SA, et al:
excision. Partial penectomy may be performed if a 1.5-cm Principles and Practice of Genitourinary Oncology, pp
margin is obtained, otherwise, total penectomy is indi- 937–942. Philadelphia, Lippincott-Raven, 1997.
cated. Because of the functional and psychologic impair- 13. Ayala AG, Ro JY: Pathology of penile cancer. In Ragavan
ment that results from surgical excision, Mohs’ D, Scher HI, Leibel SA, et al: Principles and Practice of
Genitourinary Oncology, pp 927–936. Philadelphia,
micrographic surgery, laser ablation, and radiation ther-
Lippincott-Raven, 1997.
apy have also been performed. Each has its own advan- 14. Lynch HT, Krush AJ: Delay factors in detection of cancer
tages and complications. Overall, though, each approach of the penis. Nebr State Med J 1969; 54:360.
works well for small, stage I or stage II disease and hold 15. Hardner GJ, Bhanalaph T, Murphy GP, et al: Carcinoma
the advantage that if tumor recurs, surgical excision can of the penis: analysis of therapy in 100 consecutive cases.
be done at a later date. J Urol 1972; 108:428.
708 Part VII Urethra and Penis

16. Gursel EO, Georgountzos C, Uson AC, et al: Penile 36. Young HH: A radical operation of the cure of cancer of
cancer. Urology 1973; 1:569. the penis. J Urol 1931; 26:295.
17. Dean AL: Epithelioma of the penis. J Urol 1935; 33:252. 37. Kline EA: Partial and total penectomy for cancer. Urol
18. Humphrey PA: Pathology of carcinoma of the penis. In Clin North Am 1991; 18:161.
Hamdy FC, Basler JW, Neal DE, et al: Management of 38. Lynch DF Jr, Pettaway CA: Tumors of the penis.
Urologic Malignancies, pp 507–511. London, Churchill In Walsh PC, Retik AB, Vaughan ED Jr, et al: Campbell’s
Livingstone, 2002. Urology, 8th edition, Vol 4, p 2945. Philadelphia, WB
19. Broders AC: Squamous cell epithelioma of the skin. Ann Saunders, 2002.
Surg 1921; 73:141. 39. Hanash KA, Furlow WL, Utz DC, et al: Carcinoma of
20. Villavicencio H, Rubio-Briones J, Regalado R, et al: the penis: a clinicopathologic study. J Urol 1970; 104:291.
Grade, local stage and growth pattern as prognostic 40. Andriole GL, Miller DC, Colberg JW, et al: Surgical
factors in carcinoma of the penis. Eur Urol 1997; management of penile cancer. In Vogelzang NJ,
32:442. Scardino PT, Shipley WU, et al: Comprehensive
21. Solsona E, Iborra I, Ricos JV, et al: Corpus cavernosum Textbook of Genitourinary Oncology, 2nd edition, p.
invasion and tumor grade in the prediction of lymph node 1057. Philadelphia, Lippincott, Williams & Wilkins,
condition in penile carcinoma. Eur Urol 1992; 22:115. 2000.
22. Fraley EE, Zhang G, Manivel C, et al: The role of 41. Jensen MO: Cancer of the penis in Denmark 1942 to
ilioinguinal lymphadenectomy and significance of 1962 (511 cases). Dan Med Bull 1977; 24:66.
histological differentiation in treatment of carcinoma of 42. Horenblas S, van Tinteren H, Delemarre JF, et al:
the penis. J Urol 1989; 142:1478. Squamous cell carcinoma of the penis. II. Treatment of
23. Horenblas S, van Tinteren H, Delemarre JF, et al: the primary tumor. J Urol 1992; 147:1533.
Squamous cell carcinoma of the penis. III. Treatment of 43. Opjordsmoen S, Fossa SD: Quality of life in patients
regional lymph nodes. J Urol 1993; 149:492. treated for penile cancer. A follow-up study. Br J Urol
24. Cubilla AL, Barreto J, Caballero C, et al: Pathologic 1994; 74:652.
features of epidermoid carcinoma of the penis. A 44. Mohs FE, Snow SN, Larson PO: Mohs micrographic
prospective study of 66 cases. Am J Surg Pathol 1993; surgery for penile tumors. Urol Clin North Am 1992;
17:753. 19:291.
25. Jackson SM: The treatment of carcinoma of the penis. 45. Mohs FE, Snow SN, Messing EM, et al: Microscopically
Br J Surg 1966; 53:33. controlled surgery in the treatment of carcinoma of the
26. Harmer MH: TNM Classification of Malignant Tumours, penis. J Urol 1985; 133:961.
3rd edition. Geneva, International Union Against Cancer, 46. Brown MD, Zachary CB, Grekin RC, et al: Penile
1978. tumors: their management by Mohs micrographic surgery.
27. Beggs JH, Spratt JS: Epidermoid carcinoma of the penis. J Dermatol Surg Oncol 1987; 13:1163.
J Urol 1964; 91:166. 47. Hofstetter A, Frank F: The Neodymium-YAG Laser in
28. Derrick FC Jr, Lynch KM Jr, Kretkowski RC, et al: Urology. Basel, Hoffman-LaRoche, 1980.
Epidermoid carcinoma of the penis: computer analysis of 48. Pettaway CA: Nd:YAG laser therapy for penile carcinoma.
87 cases. J Urol 1973; 110:303. Editorial comment. J Urol 2002; 168:2421.
29. Hoppmann HJ, Fraley EE: Squamous cell carcinoma of 49. Frimberger D, Hungerhuber E, Zaak D, et al: Penile
the penis. J Urol 1978; 120:393. carcinoma. Is Nd:YAG laser therapy radical enough?
30. Yu HH, Lam P, Leong CH, et al: Carcinoma of the J Urol 2002; 168:2418.
penis: report of 52 cases with reference to lymphography 50. van Bezooijen BP, Horenblas S, Meinhardt W, et al: Laser
and ilioinguinal block dissection. Clin Oncol 1978; therapy for carcinoma in situ of the penis. J Urol 2001;
4: 47. 166: 1670.
31. Abi-Aad AS, deKernion JB: Controversies in ilioinguinal 51. Tietjen DN, Malek RS: Laser therapy of squamous cell
lymphadenectomy for cancer of the penis. Urol Clin dysplasia and carcinoma of the penis. Urology 1998;
North Am 1992; 19:319. 52:559.
32. McDougal WS, Kirchner FK Jr, Edwards RH, et al: 52. Windahl T, Hellsten S: Laser treatment of localized
Treatment of carcinoma of the penis: the case for primary squamous cell carcinoma of the penis. J Urol 1995;
lymphadenectomy. J Urol 1986; 136:38. 154:1020.
33. McDougal WS: Carcinoma of the penis: improved 53. Rothenberger KH: Value of the neodymium-YAG laser in
survival by early regional lymphadenectomy based on the the therapy of penile carcinoma. Eur Urol 1986; 12(Suppl
histological grade and depth of invasion of the primary 1):34.
lesion. J Urol 1995; 154:1364. 54. Narayana AS, Olney LE, Loening SA, et al: Carcinoma of
34. Huben RP: Surgical management of penile cancer. In the penis: analysis of 219 cases. Cancer 1982; 49:2185.
Ragavan D, Scher HI, Leibel SA, et al: Principles and 55. Wajsman Z, Moore RH, Merrin CE, et al: Surgical
Practice of Genitourinary Oncology, pp 949–955. treatment of penile cancer: a follow-up report. Cancer
Philadelphia, Lippincott-Raven, 1997. 1977; 40:1697.
35. Das S: Penile amputations for the management of primary 56. Kossow JH, Hotchkiss RS, Morales PA: Carcinoma of
carcinoma of the penis. Urol Clin North Am 1992; penis treated surgically. Analysis of 100 cases. Urology
19:277. 1973; 2:169.
Chapter 43 Superficial Carcinoma of the Penis: Management and Prognosis 709

57. Gerbaulet A, Lambin P: Radiation therapy of cancer of 65. Krieg R, Hoffman R: Current management of unusual
the penis. Indications, advantages, and pitfalls. Urol Clin genitourinary cancers. Part 1: Penile cancer. Oncology
North Am 1992; 19:325. (Huntingt) 1999; 13:1347.
58. Ravi R, Chaturvedi HK, Sastry DV: Role of radiation 66. Sarin R, Norman AR, Steel GG, et al: Treatment results
therapy in the treatment of carcinoma of the penis. Br J and prognostic factors in 101 men treated for squamous
Urol 1994; 74:646. carcinoma of the penis. Int J Radiat Oncol Biol Phys
59. Crook J, Grimard L, Tsihlias J, et al: Interstitial 1997; 38:713.
brachytherapy for penile cancer: an alternative to 67. McLean M, Akl AM, Warde P, et al: The results of
amputation. J Urol 2002; 167:506. primary radiation therapy in the management of
60. Delannes M, Malavaud B, Douchez J, et al: Iridium-192 squamous cell carcinoma of the penis. Int J Radiat Oncol
interstitial therapy for squamous cell carcinoma of the Biol Phys 1993; 25:623.
penis. Int J Radiat Oncol Biol Phys 1992; 24:479. 68. Grabstald H, Kelley CD: Radiation therapy of penile
61. Kiltie AE, Elwell C, Close HJ, et al: Iridium-192 cancer: six to ten-year follow-up. Urology 1980; 15:575.
implantation for node-negative carcinoma of the penis: 69. Sagerman RH, Yu WS, Chung CT, et al: External-beam
the Cookridge Hospital experience. Clin Oncol (R Coll irradiation of carcinoma of the penis. Radiology 1984;
Radiol) 2000; 12:25. 152:183.
62. Rozan R, Albuisson E, Giraud B, et al: Interstitial 70. Fossa SD, Hall KS, Johannessen NB, et al: Cancer of the
brachytherapy for penile carcinoma: a multicentric survey penis. Experience at the Norwegian Radium Hospital
(259 patients). Radiother Oncol 1995; 36:83. 1974–1985. Eur Urol 1987; 13:372.
63. Soria JC, Fizazi K, Piron D, et al: Squamous cell 71. Lynch DF Jr, Schellhammer PF: Contemporary concepts
carcinoma of the penis: multivariate analysis of prognostic in ilioinguinal lymphadenectomy for squamous cell
factors and natural history in monocentric study with a carcinoma of the penis. AUA Update Series, 1997; 16:256.
conservative policy. Ann Oncol 1997; 8:1089. 72. Cabanas RM: Anatomy and biopsy of sentinel lymph
64. Mazeron JJ, Langlois D, Lobo PA, et al: Interstitial nodes. Urol Clin North Am 1992; 19:267.
radiation therapy for carcinoma of the penis using iridium
192 wires: the Henri Mondor experience (1970–1979). Int
J Radiat Oncol Biol Phys 1984; 10:1891.
C H A P T E R

44Invasive Carcinoma of the Penis:


Management and Prognosis
Shahin Tabatabaei, MD, and W. Scott McDougal, MD

EPIDEMIOLOGY
Carcinoma of the penis is an uncommon malignancy in been circumcised neonatally.5,6 It is believed that neona-
the United States comprising <1% of all malignancies in tal circumcision has a protective effect, particularly for
males. This translates into 1 to 2 cases per 100,000 pop- invasive carcinoma. Tseng et al.7 discovered that neona-
ulation per annum. The American Cancer Society pre- tal circumcision was inversely associated with invasive
dicts that 1400 new cancer cases will occur in 2004. The carcinoma (odds ratio [OR] = 0.41; 95% confidence
peek incidence is in the sixth and seventh decades of life. interval [CI] = 013 to 1.1) but not carcinoma in situ
In other portions of the world, particularly in Asia, (CIS).
Africa, and South America, however, it is a more com-
mon disease. There appear to be no racial predilections. Phimosis
Almost one-half of individuals with cancer of the penis
ETIOLOGY have had a history of phimosis. Hellberg et al.8 found a
The etiology is somewhat controversial; however, several 65-fold relative risk for penile SCC among males with
facts are quite clear. The presence of a foreskin, phimosis, phimosis in a Swedish case control study. Phimosis is one
chronic inflammatory conditions, treatment with psoralen of the strongest predictors of invasive carcinoma.6–8 It is
and ultraviolet A (PUVA) photochemotherapy, exposure hypothesized that men with phimosis are more likely to
to human papillomavirus (HPV), and history of smoking retain smegma. Smegma can cause epithelial hyperplasia
appear to have the most evidence suggesting that they may and mild to moderate atypia of the squamous epithelium
play a role in the development of this disease. of the preputial sac in men with phimosis.9

Lack of Circumcision/Presence of Foreskin Chronic Inflammatory Conditions


Circumcision has been established as a prophylactic The association of chronic inflammation and irritation,
measure that reduces the risk of penile cancer.1–4 Penile burned areas, preexisting scars, and draining sinuses is
squamous cell carcinoma (SCC) is rare among Jews and well established as predisposing factors for SCC in other
Muslims, who practice circumcision during the neonatal parts of the body. The same observation has been made
period and childhood, respectively. While penile cancer for penile cancer.10 Hellberg et al.8 reported that 45% of
is common in Africa, it is rare among the Ibos of Nigeria, patients had at least one episode of balanitis, while 8% of
who practice ritual male circumcision soon after birth.5 controls were affected by balanitis.
Indeed it is rare to find cancer of the penis in a patient Lichen sclerosis et atrophicus (LSA) is a chronic
who has been circumcised at birth. In a case control inflammatory skin condition of unknown etiology. The
study, neonatal circumcision was associated with a 3-fold autoimmune response that is triggered by trauma, injury,
decreased risk, albeit 20% of penile cancer patients had or infection has been suggested as its predisposing

710
Chapter 44 Invasive Carcinoma of the Penis 711

cause.5,11 Nasca et al.12 reported that 5.8% of their 86 types of HPV in the development of intraepithelial neo-
patients with LSA developed penile cancer in a 10-year plasia and carcinoma of the female cervix is well sup-
follow-up study. Bissada et al.13 reported the develop- ported by experimental and epidemiologic data. The
ment of SCC in postcircumcision scars in 15 patients. relative risk patterns of the 15 most common HPV types
implicated in cervical neoplasm have been assessed and
categorized. These include low-risk group (HPV 6, 11,
Smoking 42, 43, 44), intermediate-risk group (HPV 31, 33, 35, 51,
A history of smoking is an independent risk factor for the 52, 58), and high-risk group (HPV 16, 18, 45, 56).
development of penile cancer.6–8,14,15 The incidence of The DNA of high-risk HPV types has been detected
penile cancer among men who had ever smoked ciga- in a substantial subset of penile SCC. Dillner et al.5 noted
rettes was 2.4 times that of men who had never smoked.7 up to 40% of penile cancer lesions were positive for HPV
Although the exact cause is not known, the accumulation with PCR method in two large series, with the majority
of nitrosamines in genital secretions has been suggeted.8 of cases positive only for HPV 16 or 18. So far, a few
seroepidemiologic studies indicate that exposure to HPV
is indeed a major risk factor for penile cancer. We could
Ultraviolet Light Irradiation not find any study to confirm the causal effect of this
Treatment with PUVA photochemotherapy has been association.
considered as a strong risk factor for penile cancer. In a The presence of HPV E6 and E7 oncoproteins may
12.3-year prospective study of 892 men in a cohort of explain the carcinogenic effect of the viral infection.
patients with psoriasis, who had been treated with oral E6/E7 genes immortalize human keratinocyte on trans-
methotrexate and PUVA, Stern16 identified 14 patients fection and directly stimulate cell proliferation.
(1.6%) with 30 genital neoplasms. Recently Aubin et al.17 Furthermore, E6/E7 oncoproteins bind to the tumor
questioned the high prevalence of penile cancer in men suppressor proteins p53 and Rb (retinoblastoma), respec-
treated by PUVA and suggested the carcinogenesis is tively. E6 binds to ubiquitin-dependent proteinase,
probably dose dependent. which promotes the degradation of p53. E7 displaces the
transcription factor E2F from the Rb protein and alters
the cell cycle.27 However, we are not aware of any study
Cervical Cancer in the Partner that has investigated the expression of E6 and E7 onco-
An association of penile cancer and cervical cancer in proteins in penile cancer specimens.
partners of patients with penile cancer has been sug- Chromosomal instability,29 cooperation with activated
gested by several authors.18–21 Hellberg, however, has oncogenes, methylation of viral and cellular DNA sites,
clearly showed that these studies had methodologic flaws and telomerase activation are some of the other potential
and his review of 1064 penile cancer cases in Sweden did mechanisms of HPV oncogenicity.30–32
not show any association with cervical cancer in their
partners.22 Other recent studies also found either weak or PATHOLOGY
no risk elevation of penile cancer in men who had part-
ners with cervical cancer.23–25 Worldwide reports indicate that 95% of penile cancers
are SCC. Sarcomas account for 4% to 5% of the remain-
ing cancers. Rarely, other cancers, such as melanoma and
Human Papillomavirus Infection basal cell carcinoma, arise in the penis.
HPV infection is a sexually transmitted disease with a
very high rate of infectivity. The infection is character- Premalignant Lesions
ized by a high rate of spontaneous clearance. At least 75
different types of HPV have been identified and these The terminology of premalignant penile lesions is one
types vary not only in their DNA sequences but also in of the major areas of confusion in the nomenclature of
their clinical manifestations.26,27 penile lesions. Premalignant lesions of the penis may be
Different HPV types have been demonstrated in asso- categorized into two groups as follows. (1) Lesions that
ciation with different genital lesions. Clinically, 85% of are sporadically associated with SCC of the penis:
grossly visible genital condylomas (condyloma acumi- bowenoid papulosis of the penis, balanitis xerotica
nata) or anogenital warts contain HPV type 6 and/or obliterans (BXO), cutaneous horn of the penis and
HPV type 11.26 In contrast, microscopic lesions identi- Buschke-Löwenstein tumor (Verrucous carcinoma,
fied in the male partners of women with cervical dyspla- Giant condyloma acuminatum). (2) Lesions that are at
sia were shown to contain HPV types 16 or 18 in 60% to risk of developing into invasive SCC of the penis: CIS
75% of the cases.26,28 These are the lesions that become of the penis (erythroplasia of Queyrat and Bowen’s
visible only by acetowhitening. The causal role of certain disease).
712 Part VII Urethra and Penis

Bowenoid Papulosis
lesions are considered premalignant and one-third of
Bowenoid papulosis, although histologically similar to cases reported have been malignant at presentation.45,46.
CIS, usually has a benign course.33,34 Patterson et al.33 Surgical excision with a margin of normal tissue
evaluated 51 patients with bowenoid papulosis and around the base of the lesion has been very successful in
showed that the lesion usually occurs in young men treating it. Most malignant penile horns are of low grade,
(mean age 29.5 years). Lesions occur most commonly on but metastasis has been reported. Therefore, wide local
the penile shaft and are usually multicentric, pigmented excision and close follow-up to detect early metastasis is
papules ranging from 2 to 30 mm. Smaller lesions may suggested for malignant forms.46–48
coalesce into larger ones.33
The etiology of bowenoid papulosis is unknown,
Buschke-Löwenstein Tumor, Verrucous Carcinoma,
although viral (particularly HPV), chemical, and
Giant Condyloma Acuminatum
immunologic causes have been suggested.33,35
The diagnosis is confirmed by excisional biopsy. Whether the Buschke-Löwenstein tumor or giant condy-
Histologically, bowenoid papulosis is characterized by loma and verrucous carcinoma are the same or different
varying degrees of hyperkeratosis, parakeratosis, irregu- lesions is controversial. Histologically, the tumor is com-
lar acanthosis, and papillomatosis. As mentioned previ- posed of wide, circular rete pegs that usually extend to
ously, histologically these lesions are similar to CIS, but the deep tissue. There is no sign of anaplasia in the pegs’
bowenoid papulosis shows more maturation of ker- squamous cells. The pegs are surrounded with inflamma-
atinocytes and displays different growth patterns relative tory cells. These lesions are locally invasive and may be
to CIS.33,36–38,39 quite aggressive. They destroy adjacent structures by
Treatment includes surgical excision or elimination of local invasion. They rarely, if ever, metastasize.
the lesion by electrodesiccation, cryotherapy, laser fulgu- Treatment is directed at eradicating the local disease.
ration, or topical 5-fluorouracil cream. Spontaneous This can be achieved by local excision of the tumor and
regression has been reported.33 avoiding extensive excision of normal penile tissue. In
rare cases, total penectomy is indicated for large, infiltra-
tive lesions.
Balanitis Xerotica Obliterans
BXO is the term applied to LSA of the glans penis and
Carcinoma In Situ, Bowen’s Disease, Erythroplasia
prepuce. This disorder most often occurs in uncircum-
of Queyrat
cised, middle-aged men and may precede, coexist with,
or progress to the penile SCC.40–43 Bowen’s disease and erythroplasia of Queyrat are the
Although initially asymptomatic, most patients com- same histologically but occur in different locations.
plain of penile discomfort and/or pain, difficult urination Erythroplasia of Queyrat is CIS of the penile glans or
secondary to meatal stricture, and painful erection. On prepuce, while Bowen’s disease is CIS of the shaft or
examination, BXO presents as a well-defined marginated remainder of the genitalia and perineal area. CIS of the
white patch on the glans penis or prepuce that may penis is a velvety red, well-circumscribed lesion that
involve the urethral meatus. In chronic cases, the lesion usually involves the glans, or less frequently, the pre-
is firm due to a thick underlying fibrosis. puce or shaft of the penis. Histologically, they are
Diagnosis is made by biopsy. Histologically, active described as atypical hyperplastic cells in a disordered
lesions show pronounced orthokeratotic hyperkeratosis array, with vacuolated cytoplasm and mitotic figures at
accompanied by striking atrophy of the epidermis, which all levels. The epithelial rete extends into the submu-
is very distinctive. Rete pegs are usually lost and collagen cosa and appears elongated and widened. Increased
on the upper third of the dermis is homogenized. microvascular density with surrounding inflammatory
The treatment of BXO is usually difficult and depend- infiltrates, predominantly with plasma cells, present in
ing on the severity consists of surgical excision, topical the submucosal layer.
steroid cream, and/or laser therapy. Meatal stenosis may Up to one-third of patients with CIS of the penis may
need repeated dilatations or even formal meatoplasty. also have invasive carcinoma of the penis.49
Diagnosis is based on adequate biopsies of the lesion
with sufficient depth to rule out invasion. These lesions
Cutaneous Horn
respond well to limited local excision with minimal inter-
A cutaneous horn is an overgrowth and cornification of ference with penile anatomy. Circumcision is usually an
the epithelium that forms a solid protuberance. This is adequate treatment for CIS of the prepuce. It seems that
characterized by severe hyperkeratosis, dyskeratosis, and local fulguration with electrocautery is not able to ade-
acanthosis. Penile horn is a rare form of cutaneous horn, quately eradicate the tumor. Radiation therapy has been
with only 18 cases reported in North America.44 These successfully used for this tumor.50
Chapter 44 Invasive Carcinoma of the Penis 713

Topical use of 5-fluorouracil at 5% concentration has that flat, ulcerative tumors are usually less differentiated
shown excellent results.51,52 Successful use of CO2 or and are overall associated with earlier nodal metastases.
Nd:YAG laser and liquid nitrogen have also been SCC of the penis is considered a locoregional malig-
reported with excellent cosmetic results.53–56 nancy. The lymphatic system is the primary route for
metastases. The disease first spreads to the superficial
and deep inguinal nodes, followed by the pelvic nodes,
Invasive Carcinoma of the Penis
long before distant metastases occur. Enlarged inguinal
Ninety-five percent of cancers of the penis are SCC. lymph nodes occur in 30% to 60% of patients at presen-
Histologically, the lesion contains heavy keratinization, tation.72–75 Half of these patients actually harbor cancer
epithelial pearl formation, increased mitotic activity, and in the regional nodes. Lymph node involvement with the
hyperchromatic, enlarged nuclei. Various histologic pat- cancer may also be present in 20% of patients with neg-
terns, such as classic, basaloid, verrucous, sarcomatoid ative palpable nodes.76–78 Up to 60% of the patients may
and adenomatoid, may be present. have tumor metastasis to the contralateral inguinal
SCC may be divided histologically into well differen- nodes.79
tiated, moderately differentiated and poorly differenti- The presence and extent of inguinal lymph node
ated grades according to the classification of Broders.57 metastases are the most important prognostic factors in
This classification was recently confirmed by Maiche et men with SCC of the penis.72,80–83 Lymph node metasta-
al.,58 who proposed four grades but with similar prog- sis causes chronic infection and skin necrosis. Untreated,
nostic significance. Several authors have also studied the the majority of patients die within 1 year of diagnosis
type of spread. Cubilla et al.59 studied a whole-organ-sec- from sepsis, hemorrhage secondary to tumor erosion into
tion of 66 patients with SCC of the penis and identified the femoral vessels, and/or inanition.84 Distant metas-
four types of growth: superficially spreading squamous tases to the lung, liver, bone, or brain are uncom-
carcinoma (42%), vertical invasive carcinoma (32%), ver- mon.76,78,85
rucous carcinoma (18%), and multicentric carcinoma
(8%). They found that 82% of patients with vertical
PRESENTATION
growth versus 42% of patients with superficially spread-
ing growth have inguinal lymph node metastasis. The presence of penile lesion is the first presentation.
The distinction as to degree of differentiation is par- This could range from a subtle small papule or pustule
ticularly important as a potential predictor for metastatic that does not heal to a large exophytic, fungating lesion.
disease to the groin.60–62 It is even more powerful when Sometimes it presents as superficial, erythematous ero-
combined with tumor depth of invasion.60 sion. These lesions occur most commonly on the glans
The remaining 5% of malignancies involving the and prepuce and less commonly on the coronal sulcus
penis include the sarcomas (angiosarcoma, fibrosar- and penile shaft.
coma, myelosarcoma, Kopasi’s sarcoma), 63–66 If the primary lesions were ignored due to their loca-
melanoma,67,68 basal cell carcinoma,69,70 and lym- tion under a phimotic foreskin, the patient may present
phoma. Other tumors, which have been reported on with a mass in the inguinal area. This could be due to the
very rare occasions, include rhabdomyosarcoma, 71 lymph node enlargement secondary to inflammatory
epithelial sarcoma, malignant schwannoma, myxosar- response, or metastases. This mass may become ulcera-
coma, and neurofibrosarcoma. tive, suppurative, or hemorrhagic.
Approximately 50% of SCCs are well differentiated, These lesions are usually painless. In the late stage of
30% are moderately differentiated, and 20% are poorly the disease the patient may experience weakness, weight
differentiated.60 Nine percent of lesions are found on the loss, loss of appetite, fever, and malaise.
glans and prepuce, 21% on the prepuce, 48% on the
glans, and 14% on the prepuce, glans, and shaft. Thus,
DIAGNOSIS
92% of SCCs of the penis involve the glans and/or pre-
puce. Approximately 6% involve the coronal sulcus and Unfortunately, there is often a significant delay in diag-
only 2% of SCCs are found on the shaft with no lesions nosis due to patient and/or physician factors. The
elsewhere. patient’s delay is usually due to embarrassment, guilt,
fear, or ignorance. Delay in seeking medical care may be
as long as 1 year and may include up to 50% of the
NATURAL HISTORY
patients.86
Carcinoma of the penis usually presents as a small papil- Physician’s delay in diagnosis and treatment is usually
lary, exophytic, or flat ulcerative lesion that does not due to prolongation of a conservative approach (long
resolve spontaneously. This ulcer extends gradually and course of antibiotics, antifungal, or topical steroid ther-
may ultimately involve the entire glans or penis. It seems apy), or misdiagnosis.
714 Part VII Urethra and Penis

PHYSICAL EXAMINATION
visible with 7.5 MHz linear array small parts trans-
A thorough physical examination is crucial for the diag- ducer.87–90 In extensive infiltrating tumors, ultrasound’s
nosis and accurate staging. The penile lesion should be ability to delineate corporal invasion is compromised sig-
evaluated with regard to location, appearance, size, and nificantly.91
depth of involvement. Fixation of the lesion to the adja- MRI has been tested in several studies and it appears
cent structures, such as corporal bodies, needs to be doc- that it is the most sensitive method for determining cor-
umented. The scrotum, base of the penis, and perineum pus cavernosal infiltration but at the cost of lower speci-
should be examined for any possible tumor extension. ficity.91–95 Lont et al.91 have recently compared the
Rectal examination complements the examination and accuracy of physical examination to MRI or ultrasound in
rules out gross involvement of the perineal body or the the evaluation of primary tumor extension and concluded
presence of a pelvic mass. that physical examination alone is a reliable method for
The inguinal area needs to be inspected and palpated predicting corporal involvement. MRI and ultrasound
thoroughly for any possible lymph node enlargement. may be reserved only to examine tumors in which the
This is a crucial part of physical examination and we can- physical examination is equivocal in determining tumor
not overemphasize its importance. extension.
CT scan relies on lymph node size for detecting
metastasis. MRI evaluates the lymph node size and its
Biopsy
signal intensity. Unfortunately, neither of these image
Histologic confirmation of penile cancer should be modalities is able to differentiate benign versus malig-
obtained by a biopsy from the penile lesion. The biopsy nant lymph node enlargement. Furthermore, their sensi-
is important to evaluate the depth of invasion, tumor dif- tivity and specificity drop remarkably in normal-sized
ferentiation (grade), and the presence of vascular inva- lymph nodes. Presently, CT scan and MRI scan do not
sion. This information is very helpful to accurately stage add additional information over thorough physical exam-
the tumor and allows the surgeon to discuss the thera- ination, especially in patients with no palpable inguinal
peutic options with the patient. Although it is possible to lymph nodes.
perform the biopsy (with frozen section diagnosis) and Recently, we reported the use of lymphotropic super-
partial or total penectomy in one session, we do not paramagnetic nanoparticles, as an MRI contrast agent, in
advocate this approach due primarily to psychologic rea- evaluating lymph node metastasis in prostate cancer
sons. Loss of the phallus is psychologically devastating to patients.96 Our early experience of applying this nonin-
the patient and most patients need some time to cope vasive technique in a few penile cancer patients is encour-
with the diagnosis. The interval between the biopsy and aging. More studies are required to establish the role of
the definitive radical surgery will allow the patient and this technique in patients with penile cancer.
his physician to establish their relationship and address
the psychologic aspects of the treatment to decrease the
STAGING
enormous tension and stress that ensues following the
ablative surgery. An ideal staging system allows accurate prognosis and
assists in determining an optimal treatment.
Unfortunately, the staging system of carcinoma of the
Imaging
penis is not universally accepted and each system car-
Knowledge of the extent and depth of the primary ries its own flaws. Considering that penile cancer is a
tumor and the involvement of inguinal lymph nodes locoregional disease, accurate evaluation of the
prior to any surgical intervention is crucial in patients regional lymph nodes plays a major role in staging. The
with penile cancer. In practice, this decision is usually original Jackson system (Table 44-1) is not particularly
based on findings from a physical examination. Various helpful clinically in selecting who is most likely to have
imaging modalities have been used for this purpose, groin disease.97 The tumor-nodes-metastasis (TNM)
including ultrasonography, computed tomography system is a bit better (Table 44-2) but again suffers
(CT) scan, and magnetic resonance imaging (MRI) from inability to predict the incidence of positive
scan. regional lymph nodes. With the TNM system it is dif-
Due to the poor soft tissue resolution of CT scan, ficult to assign nodal status before definitive therapy.
ultrasound and MRI are clearly superior to CT scan in Combining the TNM system with tumor differentia-
the evaluation of primary tumor extension. tion (grade) improves the prognostic ability for
Ultrasound cannot precisely detect tumor extension in regional nodal involvement60 (Table 44-3). Considering
the glans penis area. Ultrasound, however, has shown the low incidence of penile cancer, multicenter
adequate resolution to detect corpus cavernosum inva- prospective studies are needed to validate and improve
sion, owing to the thick tunica albuginea that is readily the staging system.
Chapter 44 Invasive Carcinoma of the Penis 715

TREATMENT superficial lesions and is generally not particularly appli-


Local Treatment of the Primary Lesion cable for most cancers of the penis.
Advocates of cryotherapy101 and laser therapy suggest
For lesions that are small and involve only the dermis (<1
that the local lesions can be destroyed with preservation
cm in diameter), Mohs’ micrographic surgery may be
of the part. For lesions involving only the dermis, they
appropriate.98–100 This involves serial local excision of
may be extremely successful. For lesions involving the
the primary tumor in thin layers, with thorough micro-
subcutaneous tissue and/or corpora, successful eradica-
scopic examination of each layer. This technique has the
tion is less likely and the local recurrence rate is signifi-
advantage of preserving the part but is limited to very
cant. Laser therapy as a minimally invasive treatment has
the following outcomes: When the Nd:YAG is used as
Table 44-1 Jackson Staging System primary therapy for patients with CIS, the local recur-
Stage I Tumor confined to glans or prepuce rence rate is 6%; for lesions that invade the subcutaneous
tissue, the recurrence rate is between 10% and 20%; for
Stage II Tumor invasive into the shaft or corpora. lesions that invade the corpora, the recurrence rate
No palpable adenopathy ranges between 50% and 100%.102
External beam radiation therapy carries with it a 61%
Stage III Palpable metastases to the groin, which are
recurrence rate103 with an unacceptably high stricture
resectable
rate. Local lesions may be treated with brachytherapy
Stage IV Inoperable groin nodes or distant metastases with considerable success, particularly in patients with
T1 to T2 penile cancer, who insist on preserving the

Table 44-2 American Joint Committee on Cancer (AJCC) Staging System for Penile Cancer
Primary tumor (T)

Tx Primary tumor cannot be assessed

To No evidence of primary tumor

TIS CIS

Ta Noninvasive verrucous carcinoma

T-1 Tumor invades subepithelial connective tissue

T-2 Tumor invades corpus spongiosum or cavernosum

T-3 Tumor invades urethra or prostate

T-4 Tumor invades other adjacent structures

Regional Lymph nodes (N)

Nx Regional lymph nodes cannot be assessed

N0 No regional node metastasis

N1 Metastasis in a single superficial, inguinal lymph node

N2 Metastasis in multiple or bilateral superficial inguinal lymph nodes

N3 Metastasis in deep inguinal or pelvic lymph node(s), unilateral or bilateral

Distant metastasis (M)

Mx Distant metastasis cannot be assessed

M0 No distant metastasis

M1 Distant metastasis
716 Part VII Urethra and Penis

Table 44-3 Depth of Invasive/Grade Staging System


T Stage

Stage I The tumor is superficial with no extension into the subcutaneous tissue To, TIS, Ta, NO

Stage IIA Locally invasive tumor without involvement of the corpora, well or moderately differentiated T1, NO

Stage IIB The tumor invades the corpora or it is poorly differentiated T1, T2, NO

Stage III Persistent palpable inguinal nodes N-1, N-2

Stage IV Bulky groin nodes with invasion extending outside the node, pelvic node involvement, N-3, N-4
distant metastases

part.104,105 Crook et al.105 showed that 76% of patients ● Rarely, deeply invasive tumors, particularly those at
with T1 to T2 diseases were free of tumor locally for the base of the penis, may bypass the groin and drain
5 years. directly to the pelvic nodes.
Still, the most effective local therapy is surgical exci-
sion, although the cosmetic defects can be major. If the There are multiple cross-communications so those
lesion is located on the foreskin only, circumcision is lesions on one side of the penis may in fact metastasize to
appropriate. However, this approach is associated with a the contralateral groin. On physical examination approx-
30% local recurrence rate. Partial penectomy is the most imately 50% of patients have palpable nodes. Often these
effective method of dealing with the disorder provided nodes are inflammatory due to infection of the primary
one can establish a 2-cm proximal margin. This carries lesion. Therefore, a patient should be restaged after the
with it a 6% recurrence rate. Total penectomy has the primary lesion has been eradicated, the wound has been
least risk of local recurrence and is only employed when closed, and the patient is infection free. This generally
the tumor replaces the entire penis or when it is located requires 6 weeks of antibiotics following closure of the
at the base of the shaft. It results in a significant cosmetic penile wound. If patients are reevaluated, those with pal-
defect and many patients have considerable psychologic pably negative groin nodes will have a 20% incidence of
issues during the postoperative period.81,106–109 metastatic disease if all patients are subjected to a groin
dissection.
Tumor grade and depth of invasion (stage) have sig-
Treatment of the Inguinal Lymph Nodes nificant prognostic value in predicting lymph node
The status of the inguinal lymph nodes is the most involvement.60,81,111,113 While almost 30% of patients
important prognostic factor in men with invasive SCC of with grade I penile SCC have inguinal lymph node
the penis.72,80–83 SCC of the penis tends to spread locally involvement, about 80% of grade III tumors have posi-
to regional lymph nodes and distant metastasis is rare. tive inguinal lymph nodes.60,114 In patients whose pri-
Therefore, even in patients with local lymph node metas- mary lesion involves the corpora (T2) and the tumor is
tases, regional lymphadenectomy alone can be curative poorly differentiated, approximately 80% of such
and should be performed.76,81,110,111 Studies suggest that patients will in fact have positive groin nodes.60,111 If
inguinal lymphadenectomy offers 30% to 60% cure rate there are discrete palpable lymph nodes, approximately
to patients with inguinal node metastases. If the tumor 86% of patients with high-grade tumors will have patho-
extends to the pelvic lymph nodes the success rate drops logically positive nodes on dissection.
to <10%. Unfortunately, there is currently no effective The issue of controversy involves whether preemptive
chemotherapeutic and/or radiation therapeutic option lymphadenectomy should be performed. Because lym-
available for patients with disease extending beyond the phadenectomy carries with it some morbidity and
inguinal lymph nodes. Most of these patients will suc- because there is a defined number of patients who would
cumb to disease within 1 to 2 years.78,112 undergo lymphadenectomy needlessly if all patients were
The lymphatic drainage of the penis is as follows: subjected to regional groin dissection, a watch-and-wait
approach has been adopted over the years.
● The prepuce drains with the shaft skin to the Unfortunately, this relegates patients with nonpalpable
superficial inguinal nodes. microscopic disease to a much worse survival. Patients
● The glans drains with the corporal bodies to the who have groin dissections and have nonpalpable micro-
deep inguinal nodes. scopic disease have a markedly improved survival over
● The deep inguinal nodes drain to the pelvic nodes. those in whom the microscopic disease is allowed to
Chapter 44 Invasive Carcinoma of the Penis 717

develop into palpable disease at which point a groin dis- and their survival in our experience is close to 100%.
section performed.60,78,80,81,115 Patients with IIB disease have a 78% to 88% incidence of
It has been suggested that a sentinel lymph node groin metastases. In this group in whom watchful waiting
biopsy would be predictive of the status of the groin so is employed there is a 17% 5-year survival; in those who
that an unnecessary groin dissection could be undergo an immediate lymphadenectomy there is a 92%
avoided.116–118 Unfortunately, the location of the sentinel survival; those who have a delayed lymphadenectomy
node, which is most often located between the superficial when nodes become palpable have a 33% survival.
epigastric and the superficial external pudendal vein, is Patients with stage III disease who undergo an immedi-
variable. Therefore, its clinical use has not been found to ate lymphadenectomy have a 75% survival, whereas if no
be reliable by others.119–122 lymphadenectomy is performed there is a 33% survival.60
To overcome the anatomic variability of the sentinel Thus, it is clear that patients who have nonpalpable
node and based on the experience with breast and microscopic metastases to the groin and have a groin dis-
melanoma cancers,123 intraoperative lymph node map- section have a much better prognosis than do those in
ping (IOLM) has been proposed recently. Injection of a whom the lymphadenectomy is performed when nodal
vital blue dye and/or technetium-labeled colloid around disease becomes palpable. The problem, of course, is to
the primary lesion allows the surgeon to follow its determine who is most likely to have microscopic metas-
drainage to a single or a few lymph nodes in the inguinal tases without subjecting a large group of patients to an
region.124–126 Selective biopsies of these nodes assist to unnecessary operation since the morbidity of the opera-
outline the extent of the lymph node dissection. In the tion on occasion is not inconsequential.112,113,128–130
absence of any sentinel node involvement, some would Inguinal lymphadenectomy is not without significant
argue that there is no need for radical inguinal lymph complications. Series from the literature report: skin
node dissection (high negative predictive value). In a edge necrosis, 8% to 50%; major flap necrosis, 5%;
recent report by Horenblas et al.,127 55 patients were wound infection, 10% to 15%; lymphedema, 25% to
scanned and biopsied. One-third were found not to have 50%; seroma formation, 6% to 16%; death, 0% to
surgical findings that correlated with scintigraphy. 1%.128,130–132 A second operation is required in approxi-
Twenty percent had positive nodes and 6% of patients, mately 15% of patients.132
who were found to have negative nodes within a 3-year If the lymph node dissection is performed for micro-
period, were found to develop evidence of positive groin scopic nonpalpable disease, the complication rate is much
nodes. Thus, this methodology does eliminate a number less. In our experience the complications are as follows:
of patients who would needlessly undergo a groin dissec- small wound seroma, 15%; minor skin edge necrosis
tion at the expense of subjecting all patients to groin sen- not requiring a secondary procedure, 20%; minor, self-
tinel node biopsies. The number of false-negatives is at limited lymphedema, 20%; prolonged lymphedema, 5%.
least 6% as the follow-up in the current contemporary None have required secondary operations. The group at
series is too short to determine the true false-negative MD Anderson has also reported similar results.132
rate. In our opinion, currently, the most predictive method
New modalities for lymph node imaging, particularly of determining the probability of microscopic nodal
with lymphotropic contrast agents used with MRI, are metastases is grade of primary tumor combined with
extremely exciting. Based on our early experience with depth of invasion. Sentinel node biopsy as employed with
this technology at Massachusetts General Hospital, MR blue dye and technetium may be helpful in selected cases.
lymphangiogram, in all likelihood will have a diagnostic For the present, patients who have clinically negative
efficacy sufficient to predict who should and who should groins and have a grade III lesion invasive to the corpora
not receive a groin dissection. Unfortunately, at this time should have bilateral groin dissections. If the groin dis-
tumor differentiation and stage of tumor combined are as section is positive, a pelvic lymphadenectomy should
predictive as the modalities, such as sentinel node biop- subsequently be performed on the ipsilateral side. It is
sies either with or without blue dye. Thus, poorly differ- our preference to do the bilateral groin dissection in
entiated tumors have a 80% to 100% incidence of patients with any grade III tumor or any tumor whose
metastatic disease, moderately differentiated tumors a primary lesion invades the corpora. Perhaps the sentinel
46% incidence, and well-differentiated tumors a 24% node biopsy may be useful in these latter patients, as only
incidence of groin metastasis. In tumors that have not about 60% of them will harbor microscopic metastatic
invaded into corpora there is only a 5% to 11% incidence disease. Of course patients with persistently palpable
of metastatic disease, whereas if the corpora are invaded, groin nodes should undergo a groin dissection. In this
there is a 61% to 75% incidence of groin metastases. group, approximately 86% of patients will have metasta-
Thus, we have proposed a modified staging system60 (see tic disease. It is our preference to perform the bilateral
Table 44-3). Stages I and IIA diseases have an extremely superficial and deep node dissections in one setting.
low likelihood of metastatic groin disease (0% to 12%) When the permanent pathology has returned, a pelvic
718 Part VII Urethra and Penis

lymphadenectomy is performed on the positive side. Our and chemotherapy protocols, needs to be developed to
technique is described in Glenn’s Urologic Surgery133 optimize disease-free status.
Although some would suggest that a pelvic lym-
phadenectomy is not likely to impact survival, on occa-
sion we have found an isolated positive node in the pelvis REFERENCES
that resulted in long-term survival. Besides, the knowl-
edge of pelvic node status has prognostic significance and 1. Schoen EJ: The status of circumcision of newborns.
allows us to consider adjuvant chemotherapy in positive N Engl J Med 1990; 322(18):1308–1312.
cases. 2. Brinton LA, Li JY, Rong SD, et al: Risk factors for penile
On occasion a groin dissection must be performed for cancer: results from a case-control study in China. Int J
Cancer 1991; 47(4):504–509.
palliative reasons. Under these circumstances large tissue
3. Melmed EP, Pyne JR: Carcinoma of the penis in a Jew
defects may be created in the process of removing all vis- circumcised in infancy. Br J Surg 1967; 54(8):729–731.
ible tumor. These defects can be closed with an abdomi- 4. Kochen M, McCurdy S: Circumcision and the risk of
nal advancement flap as we have previously described.134 cancer of the penis. A life-table analysis. Am J Dis Child
Following closure of the wound, radiation therapy may 1980; 134(5):484–486.
be given to the pelvis for palliation; however, invariably 5. Dillner J, von Krogh G, Horenblas S, et al: Etiology of
these patients will suffer significant external genitalia squamous cell carcinoma of the penis. Scand J Urol
lymphedema, which can be quite disabling in the final Nephrol Suppl 2000; 205:189–193.
stages of life. 6. Maden C, Sherman KJ, Beckmann AM, et al: History of
circumcision, medical conditions, and sexual activity and
risk of penile cancer. J Natl Cancer Inst 1993;
The Role of Radiation and Chemotherapy in 85(1):19–24.
Squamous Cell Carcinoma of the Penis 7. Tsen HF, Morgenstern H, Mack T, et al: Risk factors for
penile cancer: results of a population-based case-control
The role of radiation and topical chemotherapy for the study in Los Angeles County (United States). Cancer
primary lesion has been discussed previously. Several Causes Control 2001; 12(3):267–277.
nonrandomized studies have reviewed the role of radia- 8. Hellberg D, Valentin J, Eklund T, et al: Penile cancer: Is
tion therapy for the treatment of metastatic inguinal there an epidemiological role for smoking and sexual
nodes.79,135 The results are overall dismal and indicate behaviour? Br Med J (Clin Res Ed) 1987;
the role of radiation therapy, merely as a palliative meas- 295(6609):1306–1308.
9. Reddy CR, Devendranath V, Pratap S: Carcinoma of
ure at the present time.136
penis-role of phimosis. Urology 1984; 24(1):85–88.
Due to the limited number of cases and lack of 10. Bain L, Geronemus R: The association of lichen planus of
prospective studies, an optimal chemotherapy protocol the penis with squamous cell carcinoma in situ and with
has not yet been developed. Most of the chemotherapy verrucous squamous carcinoma. J Dermatol Surg Oncol
regimens are based on the results of chemotherapy tri- 1989; 15(4):413–417.
als in SCC of the head and neck. Cisplatin, methotrex- 11. Powell JJ, Wojnarowska F: Lichen sclerosus. Lancet 1999;
ate, bleomycin, and vincristine have been used alone or 353(9166):1777–1783.
in combination. It appears that adjuvant or neoadjuvant 12. Nasca MR, Innocenzi D, Micali G: Penile cancer among
use of chemotherapy agents may be beneficial and patients with genital lichen sclerosus. J Am Acad
result in partial response in many cases.137–140 The Dermatol 1999; 41(6):911–914.
optimal chemotherapy regimen remains to be deter- 13. Bissada NK, Morcos RR, el-Senoussi M: Post-
circumcision carcinoma of the penis. I. Clinical aspects.
mined.
J Urol 1986; 135(2):283–285.
14. Daling JR, Sherman KJ, Hislop TG, et al: Cigarette
smoking and the risk of anogenital cancer. Am J
FUTURE CHALLENGE
Epidemiol 1992; 135(2):180–189.
In the future, the optimum treatment of penile cancer 15. Harish K, Ravi R: The role of tobacco in penile
needs to be developed. This requires more knowledge of carcinoma. Br J Urol 1995; 75(3):375–377.
the tumor biology. Furthermore, a staging system needs 16. Stern RS: Genital tumors among men with psoriasis
to be revised to allow more accurate prediction of tumor exposed to psoralens and ultraviolet A radiation (PUVA)
and ultraviolet B radiation. The photochemotherapy
extension prior to surgery. New imaging technologies,
follow-up study. N Engl J Med 1990;
including lymphotropic agents, need to be applied to 322(16):1093–1097.
improve the staging accuracy. A multidisciplinary 17. Aubin F, Puzenat E, Arveux P, et al: Genital squamous cell
approach incorporating surgical ablation, applying vari- carcinoma in men treated by photochemotherapy. A
ous energy modalities (i.e., radiation, laser, cryotherapy, cancer registry-based study from 1978 to 1998. Br J
thermotherapy, high intensity focused ultrasound, etc.) Dermatol 2001; 144(6):1204–1206.
Chapter 44 Invasive Carcinoma of the Penis 719

18. Martinez I: Relationship of squamous cell carcinoma of 36. Wade TR, Kopf AW, Ackerman AB: Bowenoid papulosis
the cervix uteri to squamous cell carcinoma of the penis of the penis. Cancer 1978; 42(4):1890–1903.
among Puerto Rican women married to men with penile 37. Peters MS, Perry HO: Bowenoid papules of the penis.
carcinoma. Cancer 1969; 24(4):777–780. J Urol 1981; 126(4):482–484.
19. Graham S, Priore R, Graham M, et al: Genital cancer in 38. Wade TR, Ackerman AB: Bowenoid papulosis. J Am Acad
wives of penile cancer patients. Cancer 1979; Dermatol 1981; 4(2):235.
44(5):1870–1874. 39. Bhojwani A, Biyani CS, Nicol A, et al: Bowenoid
20. Cocks PS, Peel KR, Cartwright RA, et al: Carcinoma of papulosis of the penis. Br J Urol 1997; 80(3):508.
penis and cervix. Lancet 1980; 2(8199):855–856. 40. Dore B, Grange P, Irani J, et al: Atrophicus sclerosis
21. Cocks PS, Adib RS, Hunt KM: Concurrent carcinoma of lichen and cancer of the glans. J Urol 1989;
penis and carcinoma-in situ of the cervix in a married 95(7):415–418.
couple. Case report. Br J Obstet Gynaecol 1982; 41. Campus GV, Alia F, Bosincu L: Squamous cell carcinoma
89(5):408–409. and lichen sclerosus et atrophicus of the prepuce. Plast
22. Hellberg D, Nilsson S: Genital cancer among wives of Reconstr Surg 1992; 89(5):962–964.
men with penile cancer. A study between 1958 and 1982. 42. Pride HB, Miller OF III, Tyler WB: Penile
Br J Obstet Gynaecol 1989; 96(2):221–225. squamous cell carcinoma arising from balanitis
23. Maiche AG, Pyrhonen S: Risk of cervical cancer among xerotica obliterans. J Am Acad Dermatol 1993;
wives of men with carcinoma of the penis. Acta Oncol 29(3):469–473.
1990; 29(5):569–571. 43. Simonart T, Noel JC, De Dobbeleer G, et al: Carcinoma
24. Iversen T, Tretli S, Johansen A, et al: Squamous cell of the glans penis arising 20 years after lichen sclerosus.
carcinoma of the penis and of the cervix, vulva and vagina Dermatology 1998; 196(3):337–338.
in spouses: is there any relationship? An epidemiological 44. Lowe FC, McCullough AR: Cutaneous horns of the
study from Norway, 1960–1992. Br J Cancer 1997; penis: an approach to management. Case report and
76(5):658–660. review of the literature. J Am Acad Dermatol 1985; 13(2
25. Hemminki K, Dong C: Cancer in husbands of cervical Pt 2):369–373.
cancer patients. Epidemiology 2000; 11(3):347–349. 45. Raghavaiah NV, Soloway MS, Murphy WM: Malignant
26. Zderic SA, Carpiniello VL, Malloy TR, et al: Urological penile horn. J Urol 1977; 118(6):1068–1069.
applications of human papillomavirus typing using 46. Fields T, Drylie D, Wilson J: Malignant evolution of
deoxyribonucleic acid probes for the diagnosis and penile horn. Urology 1987; 30(1):65–66.
treatment of genital condyloma. J Urol 1989; 47. Goldstein HH: Cutaneous horns of the penis. J Urol
141(1):63–65. 1933; 30:367–374.
27. Mandell GL, Bennett JE, Dolin R: Mandell, Douglas, and 48. Winterhoff E, Sparks JA: Penile horn. J Urol 1951;
Bennett’s Principles and Practice of Infectious Diseases. 66(5):704–707.
Edinburgh, Churchill Livingstone, 2000. 49. Mikhail GR: Cancers, precancers, and pseudocancers on
28. Schneider A, Kirchmayr R, De Villiers EM, et al: the male genitalia. A review of clinical appearances,
Subclinical human papillomavirus infections in male histopathology, and management. J Dermatol Surg Oncol
sexual partners of female carriers. J Urol 1988; 1980; 6(12):1027–1035.
140(6):1431–1434. 50. Grabstald H, Kelley CD: Radiation therapy of penile
29. Mullokandov MR, Kholodilov NG, Atkin NB, et al: cancer: six to ten-year follow-up. Urology 1980;
Genomic alterations in cervical carcinoma: losses of 15(6):575–576.
chromosome heterozygosity and human papilloma virus 51. Goette DK, Elgart M, DeVillez RL: Erythroplasia of
tumor status. Cancer Res 1996; 56(1):197–205. Queyrat. Treatment with topically applied fluorouracil.
30. Lazo PA, Gallego MI, Ballester S, et al: Genetic JAMA 1975; 232(9):934–937.
alterations by human papillomaviruses in oncogenesis. 52. Goette DK, Carson TE: Erythroplasia of Queyrat:
FEBS Lett 1992; 300(2):109–113. treatment with topical 5-fluorouracil. Cancer 1976;
31. Rosl F, Arab A, Klevenz B, et al: The effect of DNA 38(4):1498–1502.
methylation on gene regulation of human 53. Rosemberg SK, Fuller TA: Carbon dioxide rapid
papillomaviruses. J Gen Virol 1993; 74(Pt 5): superpulsed laser treatment of erythroplasia of Queyrat.
791–801. Urology 1980; 16(2):181–182.
32. Anderson S, Shera K, Ihle J, et al: Telomerase activation 54. Madej G, Meyza J: Cryosurgery of penile carcinoma.
in cervical cancer. Am J Pathol 1997; 151(1):25–31. Short report on preliminary results. Oncology 1982;
33. Patterson JW, Kao GF, Graham JH, et al: Bowenoid 39(6):350–352.
papulosis. A clinicopathologic study with ultrastructural 55. Rosemberg SK: Carbon dioxide laser treatment
observations. Cancer 1986; 57(4):823–836. of external genital lesions. Urology 1985; 25(6):
34. Su CK, Shipley WU: Bowenoid papulosis: a benign lesion 555–558.
of the shaft of the penis misdiagnosed as squamous 56. Landthaler M, Haina D, Brunner R, et al: Laser therapy
carcinoma. J Urol 1997; 157(4):1361–1362. of bowenoid papulosis and Bowen’s disease. J Dermatol
35. Zelickson AS, Prawer SE: Bowenoid papulosis of the Surg Oncol 1986; 12(12):1253–1257.
penis. Demonstration of intranuclear viral like particles. 57. Broders AC: Squamous cell epithelium of the skin. Ann
Am J Dermatopathol 1980; 2(4):305–308. Surg 1921; 73:141–143.
720 Part VII Urethra and Penis

58. Maiche AG, Pyrhonen S, Karkinen M: Histological 78. Kossow JH, Hotchkiss RS, Morales PA: Carcinoma of
grading of squamous cell carcinoma of the penis: a new penis treated surgically. Analysis of 100 cases. Urology
scoring system. Br J Urol 1991; 67(5):522–526. 1973; 2(2):169–172.
59. Cubilla AL, Barreto J, Caballero C, et al: Pathologic 79. Ekstrom T, Edsmyr F: Cancer of the penis: a clinical
features of epidermoid carcinoma of the penis. study of 229 cases. Acta Chir Scand 1958; 115:25–29.
A prospective study of 66 cases. Am J Surg Pathol 1993; 80. Johnson DE, Lo RK: Management of regional lymph
17(8):753–763. nodes in penile carcinoma. Five-year results following
60. McDougal WS: Carcinoma of the penis: improved therapeutic groin dissections. Urology 1984;
survival by early regional lymphadenectomy based on the 24(4):308–311.
histological grade and depth of invasion of the primary 81. McDougal WS, Kirchner FK Jr, Edwards RH, et al:
lesion. J Urol 1995; 154(4):1364–1366. Treatment of carcinoma of the penis: the case for primary
61. Theodorescu D, Russo P, Zhang ZF, et al: Outcomes of lymphadenectomy. J Urol, 1986; 136(1):38–41.
initial surveillance of invasive squamous cell carcinoma of 82. Ravi R: Prophylactic lymphadenectomy vs observation vs
the penis and negative nodes. J Urol 1996; inguinal biopsy in node-negative patients with invasive
155(5):1626–1631. carcinoma of the penis. Jpn J Clin Oncol 1993;
62. Heyns CF, van Vollenhoven P, Steenkamp JW, et al: 23(1):53–58.
Carcinoma of the penis–appraisal of a modified 83. Ornellas AA, Seixas AL, Marota A, et al: Surgical
tumour-staging system. Br J Urol 1997; 80(2): treatment of invasive squamous cell carcinoma of the
307–312. penis: retrospective analysis of 350 cases. J Urol 1994;
63. Moore SW, Wheeler JE, Hefter LG: Epitheloid sarcoma 151(5):1244–1249.
masquerading as Peyronie’s disease. Cancer 1975; 84. Staubitz WJ, Lent MH, Mack FG, et al: Carcinoma of the
35(6):1706–1710. Penis. Cancer 1955; 8:371.
64. Mabogunje O: Kaposi sarcoma of glans penis. Urology 85. Johnson DE, Fuerst DE, Ayala AG: Carcinoma of the
1981; 17(5):476–478. penis. Experience with 153 cases. Urology 1973;
65. Parsons MA, Fox M: Malignant fibrous histiocytoma of 1(5):404–408.
the penis. Eur Urol 1988; 14(1):75–76. 86. Gursel EO, Georgountzos C, Uson AC, et al: Penile
66. Rasbridge SA, Parry JR: Angiosarcoma of the penis. cancer. Urology 1973; 1(6):569–578.
Br J Urol 1989; 63(4):440–441. 87. Yamashita T, Ogawa A: Ultrasound in penile cancer. Urol
67. Manivel JC, Fraley EE: Malignant melanoma of the penis Radiol 1989; 11(3):174–177.
and male urethra: 4 case reports and literature review. 88. Dorak AC, Ozkan GA, Tamac NI, et al: Ultrasonography
J Urol 1988; 139(4):813–816. in the recognition of penile cancer. J Clin Ultrasound
68. de Bree E, Sanidas E, Tzardi M, et al: Malignant 1992; 20(9):624–626.
melanoma of the penis. Eur J Surg Oncol 1997; 89. Herbener TE, Seftel AD, Nehra A, et al: Penile
23(3):277–279. ultrasound. Semin Urol 1994; 12(4):320–332.
69. Goldminz D, Scott G, Klaus S: Penile basal cell 90. Horenblas S, Kroger R, Gallee MP, et al: Ultrasound in
carcinoma. Report of a case and review of the literature. squamous cell carcinoma of the penis; a useful addition to
J Am Acad Dermatol 1989; 20(6):1094–1097. clinical staging? A comparison of ultrasound with
70. Ladocsi LT, Siebert CF Jr, Rickert RR, et al: Basal cell histopathology. Urology 1994; 43(5):702–707.
carcinoma of the penis. Cutis 1998; 61(1):25–27. 91. Lont AP, Besnard AP, Gallee MP, et al: A comparison of
71. Dalkin B, Zaontz MR: Rhabdomyosarcoma of the penis in physical examination and imaging in determining the
children. J Urol 1989; 141(4):908–909. extent of primary penile carcinoma. BJU Int 2003;
72. Srinivas V, Morse MJ, Herr HW, et al: Penile cancer: 91(6):493–495.
relation of extent of nodal metastasis to survival. J Urol 92. Hricak H, Marotti M, Gilbert TJ, et al: Normal penile
1987; 137(5):880–882. anatomy and abnormal penile conditions: evaluation with
73. Solsona E, Iborra I, Ricos JV, et al: Corpus cavernosum MR imaging. Radiology 1988; 169(3):683–690.
invasion and tumor grade in the prediction of lymph node 93. Kawada T, Hashimoto K, Tokunaga T, et al: Two cases of
condition in penile carcinoma. Eur Urol 1992; penile cancer: magnetic resonance imaging in the
22(2):115–118. evaluation of tumor extension. J Urol 1994;
74. Ayyappan K, Ananthakrishnan N, Sankaran V: Can 152(3):963–965.
regional lymph node involvement be predicted in patients 94. de Kerviler E, Ollier P, Desgrandchamps F, et al:
with carcinoma of the penis? Br J Urol 1994; Magnetic resonance imaging in patients with penile
73(5):549–553. carcinoma. Br J Radiol 1995; 68(811):704–711.
75. Lopes A, Rossi BM, Fonseca FP, et al: Unreliability of 95. Kageyama S, Ueda T, Kushima R, et al: Primary
modified inguinal lymphadenectomy for clinical staging of adenosquamous cell carcinoma of the male distal urethra:
penile carcinoma. Cancer 1996; 77(10):2099–2102. magnetic resonance imaging using a circular surface coil.
76. Beggs JH, Spratt JS Jr: Epidermoid carcinoma of the J Urol 1997; 158(5):1913–1914.
Penis. J Urol 1964; 91(2):166–172. 96. Harisinghani MG, Barentsz J, Hahn PF, et al:
77. Hardner GJ, Bhanalaph T, Murphy GP, et al: Carcinoma Noninvasive detection of clinically occult lymph-node
of the penis: analysis of therapy in 100 consecutive cases. metastases in prostate cancer. N Engl J Med 2003;
J Urol 1972; 108(3):428–430. 348(25):2491–2499 [comment].
Chapter 44 Invasive Carcinoma of the Penis 721

97. Jackson SM: The treatment of carcinoma of the penis. 116. Cabanas RM: an approach for the treatment of penile
Br J Surg 1966; 53(1):33–35. carcinoma. Cancer 1977; 39(2):456–466.
98. Mohs FE: Chemosurgery: microscopically controlled 117. Cabanas RM: Anatomy and biopsy of sentinel lymph
surgery for skin cancer—past, present and future. nodes. Urol Clin North Am 1992; 19(2):267–276.
J Dermatol Surg Oncol 1978; 4(1):41–54. 118. Cabanas RM, Application of the sentinel node concept in
99. Mohs FE, Snow SN, Messing EM, et al: Microscopically urogenital cancer. Recent Results Cancer Res 2000;
controlled surgery in the treatment of carcinoma of the 157:141–149.
penis. J Urol 1985; 133(6):961–966. 119. Perinetti E, Crane DB, Catalona WJ: Unreliability of
100. Mohs FE, Snow SN, Larson PO: Mohs micrographic sentinel lymph node biopsy for staging penile carcinoma.
surgery for penile tumors. Urol Clin North Am 1992; J Urol 1980; 124(5):734–735.
19(2):291–304. 120. Wespes E, Simon J, Schulman CC: Cabanas approach: is
101. Farrell AM, Dawber RP: Squamous cell carcinoma of the sentinel node biopsy reliable for staging penile
penis. J Am Acad Dermatol 1998; 38(3):504–505. carcinoma? Urology 1986; 28(4):278–279.
102. Frimberger D, Hungerhuber E, Zaak D, et al: Penile 121. Srinivas V, Joshi A, Agarwal B, et al: Penile cancer–the
carcinoma. Is Nd:YAG laser therapy radical enough? sentinel lymph node controversy. Urol Int 1991;
J Urol 2002; 168(6):2418–2421 [Discussion 2421]. 47(2):108–109.
103. Zouhair A, Coucke PA, Jeanneret W, et al: Radiation 122. Pettaway CA, Pisters LL, Dinney CP, et al: Sentinel
therapy alone or combined surgery and radiation therapy lymph node dissection for penile carcinoma: the M.D.
in squamous-cell carcinoma of the penis? Eur J Cancer Anderson Cancer Center experience. J Urol 1995;
2001; 37(2):198–203. 154(6):1999–2003.
104. Kiltie AE, Elwell C, Close HJ, et al: Iridium-192 123. Morton DL, Wen DR, Wong JH, et al: Technical details
implantation for node-negative carcinoma of the penis: of intraoperative lymphatic mapping for early stage
the Cookridge Hospital experience. Clin Oncol 2000; melanoma. Arch Surg 1992; 127(4):392–399.
12(1):25–31. 124. Han KR, Brogle BN, Goydos J, et al: Lymphatic
105. Crook J, Grimard L, Tsihlias J, et al: Interstitial mapping and intraoperative lymphoscintigraphy for
brachytherapy for penile cancer: an alternative to identifying the sentinel node in penile tumors. Urology
amputation. J Urol 2002; 167(2 Pt 1):506–511. 2000; 55(4):582–585.
106. Hanash KA, Furlow WL, Utz DC, et al: Carcinoma of 125. Valdes Olmos RA, Tanis PJ, Hoefnagel CA, et al: Penile
the penis: a clinicopathologic study. J Urol 1970; lymphoscintigraphy for sentinel node identification. Eur
104(2):291–297. J Nucl Med 2001; 28(5):581–585.
107. Jensen MO: Cancer of the penis in Denmark 1942 to 126. Tanis PJ, Lont AP, Meinhardt W, et al: Dynamic sentinel
1962 (511 cases). Dan Med Bull 1977; 24(2):66–72. node biopsy for penile cancer: reliability of a staging
108. Opjordsmoen S, Fossa SD: Quality of life in patients technique. J Urol 2002; 168(1):76–80.
treated for penile cancer. A follow-up study. Br J Urol 127. Horenblas S, Jansen L, Meinhardt W, et al: Detection of
1994; 74(5):652–657. occult metastasis in squamous cell carcinoma of the penis
109. Opjordsmoen S, Waehre H, Aass N, et al: Sexuality in using a dynamic sentinel node procedure. J Urol 2000;
patients treated for penile cancer: patients’ experience 163(1):100–104.
and doctors’ judgement. Br J Urol 1994; 128. Ornellas AA, Seixas AL, de Moraes JR: Analyses of 200
73(5):554–560. lymphadenectomies in patients with penile carcinoma.
110. Fraley EE, Zhang G, Manivel C, et al: The role of J Urol 1991; 146(2):330–332.
ilioinguinal lymphadenectomy and significance of 129. Kamat MR, Kulkarni JN, Tongaonkar HB: Carcinoma of
histological differentiation in treatment of carcinoma of the penis: the Indian experience. J Surg Oncol 1993;
the penis. J Urol 1989; 142(6):1478–1482. 52(1):50–55.
111. Horenblas S, van Tinteren H: Squamous cell carcinoma 130. Ravi R: Morbidity following groin dissection for penile
of the penis. IV. Prognostic factors of survival: analysis of carcinoma. Br J Urol 1993; 72(6):941–945.
tumor, nodes and metastasis classification system. J Urol 131. Johnson DE, Lo RK: Complications of groin dissection
1994; 151(5):1239–1243. in penile cancer. Experience with 101
112. Skinner DG, Leadbetter WF, Kelley SB: The surgical lymphadenectomies. Urology 1984; 24(4):312–314.
management of squamous cell carcinoma of the penis. 132. Bevan-Thomas R, Slaton JW, Pettaway CA:
J Urol 1972; 107(2):273–277. Contemporary morbidity from lymphadenectomy for
113. Lopes A, Hidalgo GS, Kowalski LP, et al: Prognostic penile squamous cell carcinoma: the M.D. Anderson
factors in carcinoma of the penis: multivariate analysis of Cancer Center Experience. J Urol 2002;
145 patients treated with amputation and 167(4):1638–1642.
lymphadenectomy. J Urol 1996; 156(5):1637–1642. 133. Graham SD, Kean TE, Glenn JF: Glenn’s Urologic
114. Horenblas S, van Tinteren H, Delemarre JF, et al: Surgery. Philadelphia, Lippincott Williams & Wilkins, in
Squamous cell carcinoma of the penis. III. Treatment press.
of regional lymph nodes. J Urol 1993; 149(3):492–497. 134. Tabatabaei S, McDougal WS: Primary skin closure of
115. Fegen P, Persky L: Squamous cell carcinoma of the large groin defects after inguinal lymphadenectomy for
penis: its treatment, with special reference to radical penile cancer using an abdominal cutaneous
node dissection. Arch Surg 1969; 99(1):117–120. advancement flap. J Urol 2003; 169(1):118–120.
722 Part VII Urethra and Penis

135. el-Demiry MI, Oliver RT, Hope-Stone HF, et al: 138. Hussein AM, Benedetto P, Sridhar KS: Chemotherapy
Reappraisal of the role of radiotherapy and surgery in with cisplatin and 5-fluorouracil for penile and urethral
the management of carcinoma of the penis. Br J Urol squamous cell carcinomas. Cancer 1990; 65(3):433–438.
1984; 56(6):724–728. 139. Dexeus FH, Logothetis CJ, Sella A, et al: Combination
136. Vaeth JM, Green JP, Lowy RO: Radiation therapy of chemotherapy with methotrexate, bleomycin and
carcinoma of the penis. Am J Roentgenol Radium Ther cisplatin for advanced squamous cell carcinoma of the
Nucl Med 1970; 108(1):130–135. male genital tract. J Urol 1991; 146(5):1284–1287.
137. Pizzocaro G, Piva L: Adjuvant and neoadjuvant 140. Shammas FV, Ous S, Fossa SD: Cisplatin and
vincristine, bleomycin, and methotrexate for inguinal 5-fluorouracil in advanced cancer of the penis. J Urol
metastases from squamous cell carcinoma of the penis. 1992; 147(3):630–632.
Acta Oncol 1988; 27(6b):823–824.
C H A P T E R

45 Penectomy and Ilioinguinal


Lymphadenectomy
Donald F. Lynch, Jr., MD

In North America, cancer of the penis is an uncommon at wide excision or who recur subsequent to laser therapy
disease, but it is a substantial health problem in parts of may also require penectomy.
Africa, South America, and Asia. Squamous cell carci-
noma (SCC), the most common penile cancer, is a malig-
SURGICAL PROCEDURES FOR THE PRIMARY
nancy, like testicular cancer, where the behavior of the
LESION
tumor is reasonably predictable, and where regional dis-
Biopsy
ease may be curable by timely surgical intervention.
Patients with Buschke-Löwenstein tumors (verrucous Histology of any penile tumor must be confirmed with
carcinoma of the penis) are usually managed with local an adequate biopsy. Most penile cancers are SCCs, which
excision but on occasion may require partial or total spread by local invasion early in their course. Such
penectomy. Similarly, patients with Kaposi’s sarcoma of tumors metastasize to regional lymph nodes as they
the penis may occasionally require surgical excision or progress in size and depth to involve the highly vascular
partial penectomy. These tumor types do not metasta- structures of the penis. Histologic confirmation of tumor
size, and further management generally involves only depth is important in assessing the need for inguinal
interval follow-up. lymphadenectomy, as well as in determining whether
Squamous carcinoma is a far more ominous diagnosis; adequate margins of resection are possible so as to permit
these tumors have a propensity for metastasis to the partial penectomy. Several recent studies suggest that
inguinal lymph nodes and beyond. With squamous carci- tumor grade also is an important determinant in progno-
noma, the size of the primary tumor, its location on the sis and is an important consideration in assessing the risk
penis, the grade of the tumor, and the depth of invasion of inguinal metastases.2,3 Penile biopsy may be done as a
determine the extent of surgical treatment that will be separate procedure, or may done in conjunction with
required.1–3 This information comes from biopsy of the frozen-section confirmation as a prelude to definitive
lesion and from careful physical examination. partial or total penile amputation. Full informed consent
The goal of surgery for localized carcinoma of the must be obtained prior to such a procedure.
penis is complete excision of the tumor with adequate tis- Biopsy is performed by excising a 1- to 1.5-cm wedge of
sue margins. Some superficial lesions of the glans or shaft tissue, which includes the margin of the tumor and provides
may be amenable to wide excision or to treatment using normal tissue adjacent to the lesion to be examined for
laser surgery or Mohs’ micrographic surgery, while tumor infiltration (Figure 45-1). The incision may be closed
superficial tumors limited to the prepuce may sometimes with interrupted 2-0 or 3-0 chromic gut after that a sterile
be managed with circumcision.4,5 In the case of large compressive dressing is applied. There have been no reports
tumors (>2.0 cm), lesions that are located so that an ade- of tumor dissemination from biopsy of penile cancers.6
quate margin of resection cannot be achieved, or cancers
that invade the tunica albuginea, corpora, or urethra,
Partial Penectomy
limited resection may not be possible, and partial or total
penectomy will be required. Additionally, patients who Partial penectomy is the surgical treatment of choice for
develop recurrent tumor following unsuccessful attempts tumors of the glans and distal penile shaft where a margin

723
724 Part VII Urethra and Penis

If the patient has not been receiving antibiotic


therapy prior to surgery, intravenous antibiotics
are administered preoperatively. A broad-spectrum
cephalosporin, which can be converted to oral therapy
48 to 72 hours following surgery, is preferred. With the
patient in the supine position, the entire penis and scro-
tum are scrubbed with povidone-iodine solution, and
the lesion is then covered with a sterile glove or sponge
secured with a sterile rubber band. A 7/8-inch Penrose
drain is used as a tourniquet about the base of the penis
(Figure 45-2A).
A transverse incision is made over the dorsum of the
penis 2.0 cm proximal to the most proximal tumor mar-
gin and carried circumferentially around the shaft. The
superficial dorsal vessels are fulgurated, and the corpora
are then sharply divided down to the urethra, taking care
to ligate the deep dorsal vessel complex (Figure 45-2B).
The distal urethra is dissected free approximately 1 cm
proximally and distally, maintaining the 2-cm margin, to
allow for spatulation and creation of a neomeatus. The
amputation is then completed.
The corporal stumps are closed with interrupted
mattress sutures of 2-0 Vicryl (Figure 45-2B and C).
Figure 45-1 Biopsy of penile tumor, including normal tissue
for comparison. The tourniquet is then removed and further hemostasis
obtained. The urethra is spatulated and sutured to the
skin of the shaft with 3-0 or 4-0 Vicryl or Dexon. The
remaining shaft skin is closed using 3-0 absorbable
of at least 2 cm of normal tissue can be achieved. In the suture (Figure 45-2D). An 18Fr Foley catheter is placed
appropriately selected patient, this procedure should to straight drainage for 24 to 48 hours, and the wound
provide a penile stump of sufficient length to allow the is dressed.
patient to void while standing. If an adequate stump can- Alternatively, the penile shaft skin can be excised to
not be assured, total penectomy with creation of a per- create a flap dorsally. This can be folded ventrally to
ineal urethrostomy is preferable. cover the stump of the penis, and the urethra spatulated
The incidence of recurrent tumor following partial and brought through a buttonhole in the flap. The flap
penectomy has been reported as ranging from 0% to 6%. edges are sutured with 3-0 Vicryl, and excess flap skin is
Five-year survival rates of 70% to 80% following a prop- then excised as needed.
erly performed partial penectomy have been noted when Additional stump length can sometimes be gained by
inguinal nodes are free of metastatic disease.6,7 dividing the suspensory ligament of the penis, dividing

Figure 45-2 Partial penectomy.


Chapter 45 Penectomy and Ilioinguinal Lymphadenectomy 725

the ischiocavernosus muscle, and mobilizing the crura down through Buck’s fascia, and the deep dorsal venous
from the inferior pubis. The scrotum is incised along the complex is identified and traced to the symphysis pubis
raphe and reconstructed superior to the transposed phal- (Figure 45-3D). The dorsal vein complex is clamped,
lus.8 These maneuvers may provide 1 or 2 cm of addi- ligated with 2-0 silk suture and divided at this point.
tional length and avoid the need for a total penectomy in The suspensory ligament of the penis is divided, and
selected cases. with downward traction on the penile shaft, the cor-
pora cavernosa are identified and dissected from their
attachments to the inferior pubis. The corpora are
Total Penectomy
individually ligated with 0 Vicryl suture and divided
Total penectomy is the treatment of choice in patients behind the pubis.
where the lesion is located too proximally on the shaft to Following amputation of the penis, a 1-cm ellipse of
permit a partial amputation. The patient is placed in skin is excised from the region of the perineal body
standard lithotomy position, and the penis, scrotum, midway between the rectum and scrotum. Using blunt
lower abdomen, and perineum prepared with povidone- dissection, a tunnel is developed in the perineal subcuta-
iodine solution. The tumor is covered with a glove or neous tissue, and the urethra grasped and directed
dressing. through this passage, taking care not to create excessive
An elliptical incision is begun over the pubis and angulation. Excess urethra is excised and the urethral
carried around the base of the penis to the midportion stump spatulated and secured to the perineal skin with 3-
of the scrotal raphe (Figure 45-3A). The corpus spon- 0 chromic or Vicryl sutures (Figure 45-3E). After 3/4-
giosum is exposed ventrally and mobilized to the uro- inch Penrose drains are placed in the subcutaneous space,
genital diaphragm. It is transected distal to the bulb, the primary incision is sutured with interrupted 3-0
taking care to assure a 2-cm margin from the lesion Vicryl transversely, which closes the suprapubic wound
(Figure 45-3B and C ). Dorsally, the incision is carried and serves to elevate the scrotum off of the perineum

Figure 45-3 Total penectomy. (See text for details.)


726 Part VII Urethra and Penis

(Figure 45-3E and F ). The wound is dressed and an 18Fr


Foley catheter is placed to straight drainage for 48 to 72
hours. The patient is maintained on broad-spectrum
antibiotic therapy for 7 to 10 days.

MANAGEMENT OF REGIONAL DISEASE


A thorough understanding of the lymphatic drainage of
the penis is essential to stage the patient, to monitor his
progress following initial surgical intervention, and to
deal with regional disease if it is present at diagnosis or
develops subsequent to primary treatment. While penile
cancer, like testis cancer, remains one of the few malig-
nancies for which regional lymphadenectomy can be cur-
ative, controversy exists regarding the timing and
indications for inguinal and pelvic lymphadenectomy, as
well as the extent of the surgery required.

Anatomic Considerations
For the surgeon performing inguinal lymphadenectomy,
a thorough knowledge of the anatomy of the groin region
is essential. Inguinal anatomy has been meticulously
described by Daseler, who performed a series of 450
inguinal lymphadenectomies for a variety of tumors, and
whose technique has been viewed as the standard for
penile cancer.7 The boundaries of the classical lym-
phadenectomy and their relationship to significant
anatomic landmarks are illustrated in Figure 45-4. Figure 45-4 Limits of dissection for both the classical
(Daseler) lymphadenectomy (solid line) and the modified
(Catalona) lymphadenectomy (dashed line). Relation of the
Fascial Planes
femoral vein, saphenous vein, and sentinel node group to the
The lower abdomen has two superficial fascial compart- dissections is shown.
ments, Scarpa’s and Camper’s fasciae. Camper’s fascia is a
fibrofatty layer that is continuous from the lower
abdomen onto the thigh. At the scrotum, it fuses with the the fascia lata except for the fossa ovalis, an opening
deeper Scarpa’s fascia to form the tunica dartos. Scarpa’s through which the greater saphenous vein passes to empty
fascia is comprised of a distinct fibrous sheath. From the into the femoral vein below. Passing through the femoral
abdomen, it crosses the inguinal ligament and fuses with triangle are the femoral vessels and nerve. The vessels are
the fascia lata of the thigh about 1 cm inferior to invested within the femoral sheath that is formed by
Poupart’s ligament, forming Holden’s line. extensions of the transversalis fascia, which passes below
Within the pelvis, the iliac fascia invests the iliopsoas the inguinal ligament. This sheath is comprised of three
muscle and also covers the femoral nerve. The iliac fascia compartments: the medial containing the lymph nodes
fuses with the transversalis fascia at the inguinal ligament. and lymphatic trunks, the middle the femoral vein, and
The inguinal ligament is formed by the aponeurosis of the the lateral the femoral artery. Lateral to the artery within
external oblique muscle and runs from the anterior supe- the iliac fascia is the femoral nerve (Figure 45-5).
rior iliac spine to the pubic tubercle. The internal oblique
muscle attaches to the lateral half of the inguinal liga-
Veins
ment, and the transversus abdominis to its outer third.
The femoral vein enters the femoral triangle at its apex,
medial to the femoral artery. About 4 cm below Poupart’s
Femoral Triangle (of Scarpa)
ligament, it receives the greater saphenous vein anteri-
The femoral triangle is formed by the inguinal ligament, orly and the profunda femoris vein posteriorly. The
the medial margin of the sartorius muscle, and the medial greater saphenous vein originates at the dorsal venous
border of the adductor longus muscle. It is covered by arch of the foot and ascends medially, passing posterior
Chapter 45 Penectomy and Ilioinguinal Lymphadenectomy 727

Figure 45-5 Relationship of femoral anatomy—nerve, artery, vein, lymph nodes—at


the level of the fossa ovalis.

to the medial epicondyle at the knee. It ascends in the and circumflex iliac veins laterally (Figure 45-6). The
superficial fascia of the thigh and empties into the femoral femoral vein passes below the inguinal ligament to
vein after passing through the fossa ovalis. Just before the become the external iliac vein, which joins with the inter-
fossa ovalis, it receives the superficial epigastric and exter- nal iliac (hypogastric) vein at the level of the sacroiliac
nal pudendal veins medially and the accessory saphenous joint to form the common iliac vein. The paired common
iliac veins empty into the inferior vena cava at the level of
the fifth lumbar vertebra.

Arteries
The common iliac arteries arise from the terminal aorta
and divide at the level of the sacral promontory into the
internal and external iliac arteries. The internal iliac
passes posteromedially and divides to supply the pelvic
musculature and pelvic organs. The external iliac artery
passes below the inguinal ligament to become the
femoral artery, which supplies the lower limb. Just prior
to passing beneath the inguinal ligament, the external
iliac artery gives off the medial inferior epigastric artery
and the lateral circumflex iliac artery. Just after passing
beneath the inguinal ligament, the femoral artery gives
off the superficial epigastric artery, the superficial cir-
cumflex iliac artery, the superficial and deep external
pudendal arteries, and the profunda femoris artery
(Figure 45-7). The femoral artery continues past the apex
of the femoral triangle and terminates in the lower third
of the adductor magnus, where it becomes the popliteal
artery.

Nerves
The femoral nerve lies beneath the iliac fascia lateral to
the femoral vessels and outside of the femoral sheath. It
is usually not visualized in the inguinal dissection, as the
Figure 45-6 Superficial inguinal nodes, inguinal ligament, and femoral nodal tissue lies medial to the vessels within the
major venous structures. femoral sheath. Other nerves that may be encountered in
728 Part VII Urethra and Penis

Figure 45-7 Surgical approaches to inguinal and pelvic node dissections.

the groin are the ilioinguinal and lateral cutaneous nerve superficial inguinal nodes. From the glans penis, lym-
of the thigh. The obturator nerve and genitofemoral phatic drainage coalesces at the frenulum and passes lat-
nerves are encountered in the course of the pelvic lym- erally to the dorsal penis, deep to Buck’s fascia paralleling
phadenectomy. the deep dorsal vein. One pathway is to the deep femoral
node chain, which includes the femoral node of Cloquet
or Rosenmüller. An additional lymphatic pathway may
Lymphatics
course along the inguinal canal to the lateral retro-
Lymphatic drainage of the penis is to the superficial and femoral node, the most superficial of the external iliac
deep inguinal lymph nodes and to the pelvic nodes. The chain.
superficial inguinal nodes are comprised of from 4 to 25 The drainage of the corporal bodies also accompanies
nodes lying within Camper’s fascia, superficial to the the deep dorsal vein of the penis, anastomosing freely
deep fascia of the thigh, and closely associated with the with the presymphyseal lymphatic plexus at the base of
saphenous vein (see Figure 45-6). Rouviere’s classic study the penis and draining into both superficial and deep
of inguinal anatomy divides the superficial inguinal nodes inguinal nodes. Because of this network, metastatic
into five zones with the sentinel nodes lying within zone 2, involvement of both inguinal regions is possible. Further
just medial to the femoral vein around the superficial epi- drainage from the base of the penis occurs via channels
gastric and superficial external pudendal veins.7,8 The deep following the femoral canals proximally into the external
inguinal nodes—usually 1 to 3 on a side—lie below the fas- iliac and pelvic nodes.7–10
cia lata medial to the femoral vein (Figure 45-8E and F). The pelvic nodes include the external iliac group, the
Lymphatic drainage of the prepuce and penile skin internal iliac or hypogastric nodes, and the common iliac
commences in the ventral penile skin, drains superficial chain. The external iliac group—usually 8 to 10 nodes—
to Buck’s fascia in a dorsolateral direction and then lies anterior and medial to the external iliac vein. The
courses proximally along the dorsum of the penis to the hypogastric nodes—4 to 6 in number—are located
Chapter 45 Penectomy and Ilioinguinal Lymphadenectomy 729

Figure 45-8 Modified (Catalona) technique for inguinal lymphadenectomy. A, Inguinal


incisions. B, Fascia lata exposed. C, Mobilization of lymphatic tissues around saphenous
vein and femoral vessels. D, Superficial inguinal dissection has been completed. Note that
the saphenous vein has been preserved. E, Limits of the modified (solid lines) and full or
classical (Daseler) inguinal lymphadenectomy (dashed line) demonstrating the deep inguinal
nodes and the lower extent of the external iliac nodes (node of Cloquet or Rosenmüller). F,
Dissection of the deep inguinal nodes.
730 Part VII Urethra and Penis

around the hypogastric vein, and the common iliac least 2 years can be assured.1,2,12 Unreliable patients or
chain—4 to 10 nodes—is located medial to or sometimes others who cannot be provided adequate follow-up are
posterior to the common iliac vessels. Metastasis to the best served by prompt inguinal lymphadenectomy.
pelvic lymph nodes without involvement of the inguinal Considerable debate has raged over the years regarding
nodes is an extremely rare event and appears to be based immediate or delayed lymphadenectomy in patients with
on a single report by Schreiner.11 Such metastatic spread clinically negative groins. There has been support for
has not been observed in many recent studies.1,12–14 expectant management of patients with invasive (T2 to
Therefore, in the setting of negative superficial and deep T4) lesions and no palpable inguinal adenopathy, with
inguinal node dissections and a negative pelvic computed surgical intervention undertaken only when lym-
tomography (CT) scan, resection of the pelvic nodes is phadenopathy develops. This avoids the substantial mor-
not required. bidity associated with groin dissection. In those patients
who later develop clinically positive nodes, delayed lym-
Sentinel Node Biopsy phadenectomy can provide cure in 30% to 50%.1,21–23
However, recent series support prophylactic inguinal
The concept of the sentinel node was proposed by lymphadenectomy in these cases, citing ultimate mortal-
Cabanas, and postulates that there is a node or group of ity rates of up to 20% in patients who are followed until
nodes—lying between the superficial external pudendal they develop clinically positive nodes before undergoing
vein and the superficial epigastric vein—where the earli- inguinal exploration.1,2,6,12,21,23,24,25,26 It would appear
est metastasis from a penile tumor will occur consistently that in these patients, the window of opportunity to pro-
(see Figure 45-6).10,14 It was postulated that a negative vide a cure has been lost.
sentinel node biopsy in the presence of clinically negative In a patient with low-grade, low-stage tumor where
groins indicated that further inguinal dissection was unilateral groin metastases develop some time after surgi-
unnecessary. Five-year survivals of 90% have been cal treatment of the primary, inguinal lymphadenectomy
reported in this setting. However, subsequent reports of of only the involved groin is required.6 While this would
the development of metastases following a negative sen- seem to contradict recommendations outlined above,
tinel node biopsy have suggested that sentinel node bilateral occult metastases present from diagnosis that
biopsy may not be consistently reliable.15–17 Most sur- become clinically apparent later should manifest them-
geons treating penile cancer no longer use the sentinel selves at about the same time. Consequently, the lack of
node biopsy technique.18,19 In a patient where the sen- clinically detectable nodes on the side contralateral to a
tinel node biopsy has been performed, the original biopsy late-developing groin metastasis suggests that the clini-
incision should be excised with the lymphadenectomy cally negative nodes were not involved from the outset.
specimen. Preoperative staging of the pelvic, common iliac, and
paraaortic nodes with CT or magnetic resonance imag-
Considerations in Inguinal and Ilioinguinal ing (MRI) scan and, where indicated, with needle biopsy,
Lymphadenectomy is essential. While survival has followed resection of
tumor limited to the external iliac nodes, no patient with
Because lymph node enlargement and lymphangiitis due common iliac or paraaortic metastases has survived.12,27
to infection of the primary tumor is common at the time Fine needle aspiration of suspicious nodes in these areas
of diagnosis, all patients with penile cancer should may confirm metastatic tumor and thus avoid unneces-
receive 4 to 6 weeks of antibiotic therapy before under- sary surgery. The most important predictor of ultimate
going inguinal lymphadenectomy. At the completion of survival continues to be primary tumor stage—the pres-
this course of treatment, the groins are carefully exam- ence or absence of nodal disease.2,28
ined for the presence of abnormal nodes. Thirty percent Occasionally, palliative groin dissection may be
to 60% of patients who present with penile cancer have required, even in the face of extensive regional or distant
palpable inguinal nodes, and following antibiotic therapy, metastases. Lymphadenopathy, which threatens to erode
half of these nodes will harbor metastases.19,20 When through the inguinal skin or invade into the femoral ves-
metastases are detected clinically in one groin, contralat- sels, may need to be removed to avoid pain, inguinal
eral metastases will be present in 60% of cases due to the infection, or serious hemorrhage.29
crossover of lymphatics at the base of the penis.6
Consequently, in this situation contralateral inguinal
lymphadenectomy should be performed. Avoiding Complications from Inguinal
It is generally accepted that patients with low-grade, Lymphadenectomy
noninvasive tumors and clinically negative inguinal nodes Despite the encouraging prognosis for many patients
may safely be managed expectantly if close follow-up with inguinal metastases who undergo inguinal or ilioin-
with careful groin examinations every 2 months for at guinal lymphadenectomy, there has been a long-standing
Chapter 45 Penectomy and Ilioinguinal Lymphadenectomy 731

reluctance on the part of surgeons to subject patients to ligament to include all iliac and deep inguinal nodes
this procedure because of the 30% to 50% incidence of there. If only one groin is involved, a unilateral pelvic
major morbidity associated with it.22,25,28,30 Complications node dissection may be done through an oblique low
include lymphocele, substantial lower limb lymphedema, abdominal incision (Figure 45-7E) and may provide some
skin loss, and infection. Skin flap necrosis can be mini- diminished patient morbidity.18
mized by selecting the appropriate incision, by careful
tissue handling, by careful attention to skin flap thickness
Modified Inguinal Lymphadenectomy
with excision of ischemic flap margins, and by transposing
the head of the sartorius muscle to cover the defect left over While some patients with clinically negative groins will
the femoral vessels. Use of intravenous fluorescein dye and have impalpable metastatic disease, many will not. This
a Woods’ lamp intraoperatively to assess viability of wound coupled with the morbidity associated with the classical
edges is a useful adjunct.31 Lower limb lymphedema can be inguinal lymphadenectomies described by Daseler and
reduced by careful attention to intraoperative ligation of Baranofsky has made surgeons reluctant to subject
lymphatics, by immobilization of the limb or limbs in the patients to these procedures, even when tumor charac-
postoperative period, and by suction drainage of the lym- teristics indicate that lymphadenectomy would be advis-
phadenectomy site. Elastic support hose should be used in able.7,34 Catalona33 and Puras and Rivera35 have
the immediate postoperative period and may be required described modifications of the classical dissection that are
long term in many patients. Wound infection can be mini- aimed at reducing many of the unpleasant sequelae of the
mized by intensive preoperative antibiotic therapy to classical operation.33,35
reduce infection and inflammation from the primary and All patients are treated with 4 to 6 weeks of oral
by the use of prophylactic antibiotics.30,32 Thrombotic broad-spectrum antibiotics following excision of the pri-
problems may be avoided through the use of subcutaneous mary tumor. A clear liquid diet the day prior to surgery is
heparin in the perioperative period, particularly when the prescribed, and the patient is given cleansing enemas the
full classical inguinal and pelvic dissection is combined with night before surgery. Antiembolism compressive stock-
prolonged bed rest postoperatively. ings are used.
The patient is placed in a supine position after spinal
anesthesia is first administered. A Foley catheter is placed
Technique of Inguinal Lymphadenectomy
to straight drainage. The scrotum and penis are then
Figure 45-7 shows the various incisions described for retracted out of the field and draped off. The legs are
inguinal and ilioinguinal lymphadenectomy. Many sur- then abducted and externally rotated to best expose the
geons prefer the oblique incision below and parallel to anteromedial thigh and groin. A pillow is placed beneath
the inguinal crease because it maintains the integrity of the knees for support. A 6- to 8-cm incision is then made
the inguinal ligament, and because it preserves the cuta- 3 to 4 cm below the inguinal ligament and parallel to it
neous blood supply that runs parallel to the inguinal lig- (see Figure 45-8A). The incision is carried down to
ament (Figures 45-7A and D and 45-8A). Scarpa’s fascia. Gentle sponge traction is used to separate
If the patient has impalpable lymph nodes following the skin edges (Figure 45-8B). The subcutaneous tissue
antibiotic therapy but has histologically confirmed grade in Camper’s layer is carefully preserved, and meticulous
2 or grade 3 invasive disease, a modified, somewhat lim- handling of these tissues and the skin edges must be
ited inguinal lymphadenectomy as described by observed throughout. Stay sutures of 2-0 or 3-0 silk and
Catalona33 is performed. If bilateral palpable inguinal skin hooks are helpful to avoid injuring the wound edges.
nodes persist, a full bilateral ilioinguinal lymphadenec- The saphenous vein and its tributaries are identified,
tomy, using the classical limits of dissection is under- and the superficial areolar and node-bearing tissue is
taken. In the event one groin normalizes following gently dissected off the vein downward to the fascia lata
antibiotic therapy but the other remains abnormal, a (Figure 45-8C). The venous branches emptying into the
superficial inguinal dissection is performed on the nor- saphenous vein are carefully ligated and divided. The
mal side with frozen-section evaluation. If this is nega- saphenous vein is preserved (Figure 45-8D). Large lym-
tive, a full ilioinguinal dissection is then performed on phatics and small venous branches must be carefully and
the contralateral abnormal side. meticulously ligated or fulgurated. The dissection is car-
Generally, pelvic node dissection is done through a ried superiorly to approximately 2 cm above the inguinal
low midline incision giving access to both right and left ligament where it is carried down to the fascia of the
pelvic node groups (see Figure 45-7D). The dissection external oblique muscle (Figure 45-8E).
technique for this part of the operation is familiar to The dissection is carried inferiorly to about 4 cm
urologists, as it is similar to the technique used in blad- below the incision. In Catalona’s operation, the lower
der cancer staging, except that the dissection of the exter- limit of dissection is the lower border of the fossa ovalis
nal iliac nodes is carried more distally under the inguinal (Figure 45-8F ). Other surgeons carry the dissection
732 Part VII Urethra and Penis

slightly lower, although there is not much superficial Scarpa’s fascia, and the saphenous vein is identified. This
nodal tissue in this area. The specimen of superficial is ligated with 2-0 silk sutures and divided. The superfi-
nodal tissue is oriented, labeled, and sent for frozen- cial nodal tissue is systematically mobilized, beginning in
section evaluation. the superomedial quadrant, freeing the tissue towards the
If the nodes are negative, the wound is thoroughly junction of the saphenous vein with the femoral vein at
irrigated with sterile water and closed in layers, taking the fossa ovalis. The sentinel node tissue is identified and
care to eliminate any potential spaces. A closed suction tagged with a suture. Dissection is extended above the
drainage system is placed (Figures 45-9C and 45-10F ) inguinal ligament 2 cm, and the limits of the standard
and remains for 5 to 7 days during that time the patient dissection, as outlined in Figure 45-4, are utilized.
is maintained at complete bed rest. Next, the venous tributaries of the saphenous vein are
meticulously ligated and divided, as well as any large
lymphatics. The saphenous vein is carefully dissected
Classic Inguinal Lymphadenectomy (after Daseler)
down to the fossa ovalis and the junction with the
When known positive nodes are present, or when femoral vein is identified, clamped, divided, and ligated.
metastatic disease is confirmed by frozen section during The fascia lata is opened and excised with all the nodal
a limited inguinal dissection, a classic radical inguinal tissue superficial to it. This exposes the femoral sheath
lymphadenectomy is performed.9 An incision of 6 to 8 and Scarpa’s triangle. The lateral aspect of this dissec-
cm is made parallel to the inguinal ligament and about 3 tion is the medial border of the sartorius, and the medial
to 4 cm inferior to it. The incision is carried down to border of the adductor longus comprises the medial side.

Figure 45-9 Completed full classical lymphadenectomy. Note that the fascia lata
has been more widely excised and that the saphenous vein has been divided.
The proximal head of the sartorius muscle is transposed to cover the femoral
vessels. Meticulous closure with suction drainage and complete bed rest for 4 or
5 days is essential to avoid wound complications. (See text for details.)
Chapter 45 Penectomy and Ilioinguinal Lymphadenectomy 733

The femoral sheath is opened from the inguinal liga- Rosenmüller), the most proximal of the deep inguinal
ment to the apex of the femoral triangle. Within the chain. One to 5 nodes are usually encountered. About 5
sheath are the femoral vein, the femoral artery lateral to to 8 cm below the inguinal ligament, the profunda
it, and fatty areolar tissue containing the deep inguinal femoris artery arises on the lateral aspect of the femoral
nodes medially (see Figure 45-5). The femoral nerve is artery. This must be carefully dissected and preserved,
lateral to the artery and is not encountered in the usual especially if myocutaneous flap coverage may later be
dissection. The vein is gently retracted laterally to allow required.
dissection of tissue from the lateral and anterolateral The wound is then irrigated carefully and careful
aspect of the vein. Nodal and areolar tissue is then dis- attention is given to all bleeders and lymphatics. The sar-
sected from anterior to the artery and between the artery torius muscle is separated sharply from its origin on the
and vein (Figure 45-9A). The nodes are dissected to anterior superior iliac spine and mobilized medially to
below the inguinal ligament to the node of Cloquet (or cover the now exposed femoral vessels (Figure 45-9B).34,36

Figure 45-10 Management of the ulcerated groin with vascular involvement. If required, a
Gore-tex vascular graft is placed. Transposition of a myocutaneous flap based on the
gracilis muscle is used to cover the inguinal defect. (See text for details.)
734 Part VII Urethra and Penis

Drainage using a closed suction catheter apparatus is 4. Mohs FE, Snow SN, Larson PO: Mohs micrographic
placed, and the wound is carefully closed in layers. surgery for penile tumors. Urol Clin North Am 1992;
Several sutures are used to secure Camper’s fascia to the 19:291.
anterior aspect of the muscles to close potential space and 5. Bissada NK: Conservative extirpative treatment of cancer
of the penis. Urol Clin North Am 1992; 19:283.
discourage lymphocele formation (Figure 45-9C).35 The
6. Ekstrom T, Edsmyr F: Cancer of the penis: a clinical
skin edges are carefully assessed, using intravenous fluo-
study of 229 cases. Acta Chir Scand 1958; 115:25.
rescein and the Woods’ lamp if necessary, and any ques- 7. Daseler E, Anson B, Riemann A: Radical excision of the
tionably viable skin is excised. The wound is then inguinal and iliac lymph nodes. Surg Gynec Obstet 1948;
meticulously closed in layers. The patient is maintained 87:679.
at complete bed rest with compressive stockings in place 8. Rouviere HA: Anatomy of the Human Lymphatic
for 5 to 7 days. Subcutaneous heparin is given. The Foley System. Ann Arbor, MI, Edward Bros., 1938 [Tobias MJ
catheter is discontinued after 24 to 48 hours. Closed suc- (trans)].
tion is maintained for 5 days. 9. Dewire D, Lepor H: Anatomic considerations of the penis
and its lymphatic drainage. Urol Clin North Am 1992;
19:211.
Management of Enlarged or Ulcerated Nodes and 10. Riveros M, Garcia R, Cabanas RM: Lymphangiography of
Femoral Vessel Involvement the dorsal lymphatics of the penis: technique and results.
Cancer 1967; 20:2026.
SCC is characterized by aggressive local invasion into 11. Schreiner BF: Treatment of epithelioma of the penis
surrounding tissues. Large inguinal nodes, particularly based on a study of 60 cases. Radiology 1929; 13:353.
when the patient presents for treatment late or after an 12. Srinivas V, Morse MJ, Herr HW, et al: Penile cancer:
extended period of neglect, will have invaded the overly- relation of extent of nodal metastases to survival. J Urol
ing skin of the groin and may ulcerate through it in 1987; 137:880.
extreme cases (Figure 45-10A). Induration of the skin is 13. Horenblas S, Van Tinteren H, Dellemarre JFM, et al:
suggestive of such local invasion. In this setting, the Squamous cell carcinoma of the penis III. Treatment of
tumor is excised leaving a 2-cm margin of normal skin regional lymph nodes. J Urol 1993; 149:492.
around the indurated or ulcerated area (Figure 45-10B). 14. Cabanas RM: An approach for the treatment of penile
carcinoma. Cancer 1977; 39:456.
A radical ilioinguinal lymphadenectomy is performed,
15. Perinetti EP, Crane DC, Catalona WJ: Unreliability of
and all tumor is completely resected.
sentinel node biopsy for staging penile carcinoma. J Urol
In some instances, there may be involvement of the 1980; 124:734.
femoral vessels, particularly the vein. Resection of the 16. Wespes E, Simon J, Schulman CC: Cabanas approach: Is
anterior wall of the vein with reconstruction using a sentinel lymph node biopsy reliable for staging penile
saphenous vein graft or Gore-tex patch may be required carcinoma? Urology 1986; 28:278.
(Figure 45-10C and D).36 In some instances of severe 17. Fowler JE Jr: Sentinel node biopsy for staging penile
involvement, the vein may be ligated and excised. cancer. Urology 1984; 23:352.
Large skin defects may be closed with a tensor fascia 18. Fowler JE: Mastery of Surgery: Urology. Boston, MA,
lata or gracilis myocutaneous graft (Figure 45-10E).33,37,38 Little Brown Co., 1991.
Such a graft may be based over the hip and swung medi- 19. Pettaway CA, Pisters LL, Colin PN, et al: Sentinel lymph
node dissection for penile carcinoma: the MD Anderson
ally and superiorly to cover a groin defect (Figure 45-10F).
Cancer Center experience. J Urol 1995; 154:1999.
Again, all postoperative precautions recommended for
20. Johnson DE, Lo RK: Management of regional lymph
the classical radical lymphadenectomy should be nodes in penile carcinoma: five-year results following
employed for patients undergoing this procedure. therapeutic groin dissections. Urology 1986; 28:308.
21. Catalona WJ: Role of lymphadenectomy in carcinoma of
the penis. Urol Clin North Am 1980; 7:85.
REFERENCES 22. Narayana AS, Olney LE, Loening SA, et al: Carcinoma of
the penis: analysis of 229 cases. Cancer 1982; 49:2185.
1. Fraley EE, Zhang G, Manivel C, et al: The role of 23. Lesser JH, Schwarz H II: External genital carcinoma:
ilioinguinal lymphadenectomy and the significance of results of treatment at Ellis Fischel State Cancer Hospital.
histological differentiation in the treatment of carcinoma Cancer 1955; 8:1021.
of the penis. J Urol 1989; 142:1478 24. McDougal WS, Kirchner FK Jr, Edwards RH, et al:
2. McDougal WS: Carcinoma of the penis: improved Treatment of carcinoma of the penis: The case for
survival by early regional lymphadenectomy based on the primary lymphadenectomy. J Urol 1986; 136:38.
histological grade and depth of invasion of the primary 25. Beggs JH, Spratt JS Jr: Epidermoid carcinoma of the
lesion. J Urol 1995; 154:1364. penis. J Urol 1964; 91:166.
3. Theodorescu D, Russo P, Zhang ZF, et al: Outcomes of 26. Fossa SD, Hall KS, Johanssen MB: Carcinoma of the
initial surveillance of squamous cell carcinoma of the penis: experience at the Norwegian Radium Hospital:
penis and negative nodes. J Urol 1996; 155:1626. 1974–1985. Eur Urol 1987; 13:372.
Chapter 45 Penectomy and Ilioinguinal Lymphadenectomy 735

27. Ornellas AA, Seixas AL, Morota A, et al: Surgical Krane RJ, Siroky MB, Fitzpatrick JM (eds): Operative
treatment of invasive squamous cell carcinoma of the Urology. New York, Churchill Livingstone, 2000.
penis: retrospective analysis of 350 cases. J Urol 1994; 33. Catalona WJ: Modified inguinal lymphadenectomy for
151:1244. carcinoma of the penis with preservation of the saphenous
28. deKernion JB, Tynbery P, Persky L, et al: Carcinoma of veins: technique and preliminary results. J Urol 1988;
the penis. Cancer 1973; 32:1256. 140:306.
29. Ferrigni RG, Novicki DE: Complications of 34. Spratt JS Jr, Shieber W, Dillard BM: Anatomy and
lymphadenectomy in urologic surgery. Atlas Urol Clin Surgical Techniques of Groin Dissection. St Louis,
North Am 1995; 3:105. Mosby, 1965.
30. Herr HW: Surgery of penile and urethral cancer. In 35. Puras A, Rivera J: Inguinal and pelvic lymphadenectomy
Walsh PC, Retik AB, Stamey TA, Vaughan ED Jr (eds): for penile cancer. Atlas Urol Clin North Am 1995; 3:81.
Campbell’s Urology, 6th edition. Philadelphia, WB 36. Baranofsky ID: Technique of inguinal node dissection.
Saunders, 1992. Surgery 1948; 24:555.
31. Smith JA, Middleton RG: The use of fluorescein in 37. Hill HL, Nahai F, Vasconez LO: The tensor fascia lata
radical inguinal lymphadenectomy. J Urol 1979; 122:754. myocutaneous free flap. Plast Reconstr Surg 1978; 61:517.
32. Lynch DF, Schellhammer PF: Partial penectomy, total 38. Staubitz WJ, Melbourne HL, Oberkircher OJ: Carcinoma
penectomy, and radical inguinal lymphadenectomy. In of the penis. Cancer 1955; 8:371.
C H A P T E R

46Neuroblastoma
Michael C. Carr, MD, PhD, and Michael E. Mitchell, MD

ETIOLOGY
Neuroblastoma is the second most common solid tumor
in infants and children. Virchow,1 in 1864, was the first to Neuroblastoma develops from neural crest cells in the
speculate that its origin was neural, and he considered it a embryo. These cells give rise to sympathetic neuroblasts,
glioma. Marchand, in 1891,2 noted the histologic similar- which are found in the adrenal medulla, autonomic gan-
ities between neuroblastoma and developing sympathetic glia, and peripheral nerve sheaths. Other tumors that can
ganglia. Following Wright’s use of the term “neuroblas- arise from these cells include ganglioneuroma and gan-
toma” in 1910, there has been little debate with regard to glioneuroblastoma. The former may cause considerable
origin of the tumor. Herxheimer3 demonstrated that fib- morbidity secondary to spread to contiguous organs but
rils in neuroblastoma stained with a specific neural silver does not metastasize. The latter is known to metastasize
stain helping establish the true origin of this tumor. There about 20% of the time and has greater malignant poten-
was little published in the literature earlier this century, tial than ganglioneuroma.
with Blacklock4 noting only 116 cases in the literature, to Knudson and Strong,9 noting the apparent genetic
which he added 18 of his own. At that time, neuroblas- influence, suggested that 20% of cases arise in children
toma was fourth in order of frequency of malignant predisposed to the tumor by a dominant transmittable
tumors in children. The number of cases described mutation. Later work suggested that familial cases result
increased to 623 by 1953,5 and today it remains the fourth from a prezygotic mutation and that nonfamilial cases
most common malignancy in childhood, being less result from a postzygotic somatic mutation.
common than leukemia, brain tumors, and lymphoma. Neuroblastomas are characterized cytogenetically by
Cushing and Wolbach6 reported for the first time a deletion of the short arm of chromosome 1, double-
transformation of neuroblastoma into benign gan- minute chromatin bodies (dmins), and homogeneously
glioneuroma laying the foundation for a greater under- staining regions (HSRs).10,11 The latter two abnormali-
standing of its biologic variability. Later descriptions ties reflect gene amplification, whereas the former may
noted that this event generally occurred in infants <6 represent deletion of a tumor suppressor gene. No other
months of age. Beckwith and Perrin7 noted microscopic specific karyotypic abnormalities have been detected
foci of neuroblastoma cells in the adrenal glands in a thus far.
number of infants under 3 months of age who died from Flow cytometry of DNA content provides a way of
other causes. These were termed “neuroblastoma in measuring total DNA content, which correlates with
situ,” and were estimated to occur approximately 40 the modal chromosome number. Determination of the
times more frequently than the number of neuroblas- DNA index (DI) of neuroblastomas from infants pro-
toma cases clinically diagnosed. Recession of these foci vides important information that may be predictive of
appeared to be complete after 3 months of age. response to particular chemotherapeutic regimens, as
Biochemical activity associated with neuroblastoma was well as outcome.12–14 Tumors with “hyperdiploid” DNA
initially noted due to elevated levels of urinary pressor content (DI = 1) are more likely to have lower stages of
amines.8 It has subsequently been recognized that ele- disease and to respond to cyclophosphamide and dox-
vated levels of norepinephrine, its precursors and orubicin, whereas those with a “diploid” DNA content
metabolites occurs in patients with this tumor. (DI = 1) are more likely to have advanced stages of

739
740 Part VIII Pediatric Malignancies

disease and do not respond to this combination.15 This first year of life and another quarter between 1 and 2
analysis provides no information about specific chromo- years of age. This is a younger age of presentation than
some rearrangements (deletions, translocation, or gene that for Wilms’ tumor. Unlike other childhood tumors, a
amplification) but does correlate with biologic behavior. low incidence of associated congenital anomalies exists.
The discovery of extrachromosomal dmins and chro- However, brain and skull defects have been noted in 2%
mosomally integrated HSRs is a cytogenetic manifesta- of patients with neuroblastoma.26 An association with
tion of gene amplification.16,17 The region amplified is neurofibromatosis and Hirschbrung’s disease has been
derived from the distal short arm of chromosome 2, con- noted.27
taining the proto-oncogene N-myc. Brodeur et al.18 have
demonstrated that N-myc amplification occurs in 25% to PATHOLOGY
30% of primary neuroblastomas from untreated patients,
and amplification is associated primarily with advanced Arising from primitive, pleuripotential sympathetic cells
stages of disease. Subsequent studies have shown that (sympathogonia) derived from neural crest, neuroblas-
N-myc amplification was associated with rapid tumor tomas are found in a variety of locations. The adrenal
progression and a poor prognosis. Amplification was medulla or cells in the adjacent retroperitoneal tissues on
found in only 5% to 10% of patients with low stage of the posterior abdominal wall account for 50% to 80% of
disease or stage IV-S but 30% to 40% of advanced dis- neoplasms in most reported series. The second most
ease patients.19 It is almost always present at the time of common location is within the posterior mediastinum
diagnosis and, thus, appears to be an intrinsic biologic usually in paravertebral sites. The remaining neoplasms
property in a distinct subset with a poor prognosis.20 occur in derivatives of the neural crest within the pelvis,
There is a correlation between N-myc copy number and cervical region, lower abdominal sympathetic chain,
expression, and tumors with amplification expressed N- rarely within the posterior cranial fossa, or other
myc at much higher levels than are seen in tumors with- locations.
out amplification. Finally, there is heterogeneity in the Macroscopically, the growths are lobular, soft in con-
level of expression among the tumors that have a single- sistency, and weigh between 80 and 150 g. The cut sur-
copy of N-myc, but higher expressing single-copy tumors face is red-gray in color, and areas of hemorrhage and
do not appear to be a particularly aggressive subset.21,22 necrosis may be obvious as the tumor increases in size.
Deletions of the short arm of chromosome 1 are found Calcification is not infrequent and this can help in the
in 70% to 80% of the near-diploid tumors that have been radiologic localization (Figure 46-1).
karyotyped. This cytogenetic finding is commonly associ- Histologically, the cells are small and dark like lym-
ated with more advanced stages of disease, whereas tumors phocytes and frequently are arranged in masses without
from patients with lower stages are more likely to be any true pattern. It is one of the “small blue round cell”
hyperdiploid or triploid with very few structural tumors of childhood (e.g., lymphomas, neuroectodermal
rearrangements. The deletions of chromosome 1 are vari- tumor, and rhabdomyosarcoma). In characteristic lesions,
able but generally map to a region (1p36) that may contain
a suppressor gene important in malignant transformation
or progression. Recent molecular studies have shown a
strong correlation between loss of heterozygosity (LOH)
for chromosome 1p and N-myc amplification, suggesting
that these two events may be related.23 An allelic loss on
the long arm of chromosome 14 also has been noted sug-
gesting that there may be another suppressor gene
involved in the pathogenesis of neuroblastomas.

INCIDENCE
Neuroblastoma accounts for 8% to 10% of all childhood
tumors and is the most common malignant tumor of
infancy. In the United States the incidence is 10.5 per
million per year in white children and 8.8 per million per
year in black children <15 years of age.24,25 Male pre-
dominance is noted but is slight, 1.1 to 1. The incidence Figure 46-1 Typical adrenal neuroblastoma with areas of
is underestimated because of the occurrence of sponta- hemorrhage and necrosis. Tumor encircles aorta (straight
neous regression of neuroblastoma in situ. arrow) with kidney seen at left (curved arrow). (Courtesy Dr.
Approximately one-third of cases are diagnosed in the Kathleen Patterson, Seattle, WA.)
Chapter 46 Neuroblastoma 741

rosettes are formed when the tumor cells occupy the Shimada et al.30 The system is formulated around patient
periphery and the young nerve fibrils grow into the cen- age and the following histologic features: the presence or
ter of each rosette (Figure 46-2). absence of schwannian stroma; the degree of differentia-
The fully differentiated, and benign, counterpart of tion; the mitosis-karyorrhexis index (MKI). A retrospec-
neuroblastoma is the ganglioneuroma, composed princi- tive evaluation of the Shimada’s method in 295 patients
pally of mature ganglia, neuropil, and schwannian cells. treated by the Children’s Cancer Study Group (CCSG)
Ganglioneuroblastoma defines a heterogenous group of identified favorable and unfavorable patient subsets. The
tumors with histopathologic features reflecting the spec- histologic patterns were independently predictive of out-
trum of maturation of neuroblastoma and ganglioneu- come whereas stage was prognostically less important
roma. These may be either focal or diffuse, depending on than histologic grade.31,32
the pattern seen, but diffuse ganglioneuroblastoma is A simplified system has been devised, which predicts a
associated with less aggressive behavior. Multiple sec- favorable outlook based on presence of calcification and a
tions should be examined to categorize these tumors low mitotic rate (=10 mitoses/10 high power field).33 A
accurately because viability and histopathologic features grading system was developed for finding tumors with
are variable. both features (grade 1), with the presence of only one of
Hematoxylin–eosin staining and light microscopy is these features (grade 2), or the absence of both features
often not enough to distinguish these tumors from (grade 3). When these grades were combined with age (≤1
other “small round blue cell” tumors of childhood. year or >1 year) and surgicopathologic staging, low- and
Immunohistochemical techniques and electron high-risk groups emerged, which were clearly correlated
microscopy are helpful additions to light microscopy. with the Shimada’s favorable and unfavorable groups.
Monoclonal antibodies recognizing neural filaments, The result of this analysis has led to the formation
synaptophysin, and neuron-specific enolase will stain of the International Neuroblastoma Pathology Classi-
neuroblastoma.28 Electron microscopy visualizes the fication System. A recent study has analyzed the mor-
dense core, membrane-bound neural secretory granules phologic features of neuroblastoma (schwannian
in addition to microfilaments, and parallel arrays of stroma-poor tumors) in clinically favorable and unfavor-
microtubules within the neuropil (Figure 46-3).29 able groups. This study reaffirmed the prognostic impact
A histology-based prognostic classification that has of Shimada’s criteria and demonstrated that additional
gained wide-spread acceptance was developed by morphologic features, such as prominent nucleoli and

Figure 46-2 Histologically, the cells comprising neuroblastoma are small with darkly
staining nuclei and inconspicuous cytoplasm. Rosettes composed of tumor cells
surround neuropil material (arrows). (Courtesy Dr. Kathleen Patterson, Seattle, WA.)
742 Part VIII Pediatric Malignancies

Figure 46-3 Electron microscopy showing dense core neural secretory granules
(straight arrows) with microtubules (curved arrow) characteristic of neuroblastoma.
(Courtesy Dr. Kathleen Patterson, Seattle, WA.)

undifferentiated and poorly differentiated neuroblasts, sion.39 A recent collaborative study from 6 European
cellularity and nuclear size provide additional critical rel- centers identified 17q gain as the most powerful prog-
evance partly independent of the patient’s age at diagno- nostic factor in survival and multivariant analysis with
sis.34 The analysis of these tumors at the cytogenetic and other clinical and tumor genetic parameters, including 1p
molecular level revealed ploidy changes, amplification of deletion and MYCN amplification.40
the oncogene MYCN, deletions of chromosome 1p,
gains of chromosome arm 17q, and deletions of 11q. It
PRESENTATION
was demonstrated that in infants, DNA content was sig-
nificantly linked to tumor stage, with advanced stages Neuroblastoma most often presents as an abdominal mass
seen in diploid tumors. Hyperdiploidy is mainly observed (65% discovered at the time of routine medical examination
in low-stage tumors of younger patients with a favorable or by the parents). Infants tend to have more thoracic and
clinical outcome, whereas diploid tumors are associated cervical primary tumors. The tumor is usually asymmetric
with advanced tumor growth and significantly reduced and located in the supraumbilical region, the hypochon-
survival probability.35 drium, or the flank often extending beyond the midline.
Approximately 35% of all neuroblastomas have 1p Tumors which arise from the adrenal or suprarenal sympa-
deletions of variable lengths36 with poor outcomes seen thetic chain are more lateral whereas the medially located
in patients with large 1p deletions than patients with tumors may arise from the periaortic sympathetic chain.
short or interstitial deletions.37 Amplified MYCN is one These tumors may then grow through the intervertebral
of the most prominent genomic abnormalities of neurob- foramina and produce spinal cord compression.
lastoma, being prototypic for the significance of proto- Neuroblastomas can arise from anywhere along the
oncogene amplification and tumor agenesis. Amplified sympathetic chain; about 14% arise in the chest in infants
MYCN correlates with advanced-stage disease; in the over 1 year of age, but only 0.5% in the cervical region.
absence of amplified MYCN, overall survival is approxi- Tumors arising from the cervical sympathetic ganglion
mately 60% over a 5-year period, but only 10% of may produce Horner’s syndrome. In about 1% of
patients survived a 1-year period when MYCN was patients, a primary tumor cannot be found. The majority
amplified at more than 10 copies.38 of children with neuroblastoma are diagnosed by the age
The most frequent genomic alteration in neuroblas- of 5 years, and it rarely occurs after age 10.
toma is a gain of 17q, seen in about 50% of tumors. Such Metastatic extension of neuroblastoma occurs in two
a gain was significantly associated with tumor progres- patterns, lymphatic and hematogenous. Approximately
Chapter 46 Neuroblastoma 743

70% of all patients will present with metastatic disease at


DIAGNOSTIC EVALUATION
diagnosis. Regional lymph node metastasis will be noted
in 35% of patients with apparently localized disease. Minimum criteria for establishing the diagnosis of neu-
Tumor spread to lymph nodes outside the cavity of ori- roblastoma include43: (1) an unequivocal pathologic diag-
gin is considered to be disseminated disease. These chil- nosis is made from tumor tissue by standard methods,
dren may have a better prognosis if no other metastatic including immunohistology or electron microscopy if
disease is found.41 Hematogenous metastasis occurs most necessary; or (2) bone marrow contains unequivocal
often to bone marrow, bone, liver, and skin. Rarely, dis- tumor cells (i.e., syncytia) and urine contains increased
ease may spread to lung and brain parenchyma, usually as urinary catecholamine metabolites (SD > 3 above the
a manifestation of relapsing- or end-stage disease. The mean, corrected for age). Since neuroblastoma often
proportion of patients presenting with localized, secretes catecholamines, urinary levels of vanillylman-
regional, or metastatic disease is age dependent. The delic acid (VMA) or homovanillic acid (HMA) should be
incidence for localized tumors, regional lymph node elevated.44 A 24-hour urine was previously recom-
spread, and disseminated disease is 39%, 18%, and 25%, mended, but normalizing VMA and HMA excretion to
respectively, in infants (18% with stage IV-S) compared the milligram creatinine in the sample makes a timed col-
to 19%, 13%, and 68%, respectively, in older patients. lection unnecessary and it avoids false negative results
Several classical signs and symptoms have been associ- due to dilution. Table 46-1 outlines the minimum testing
ated with metastatic neuroblastoma. Proptosis and peri- to define the clinical stage of disease.43 The evaluation
orbital ecchymosis are frequent and result from often begins with an abnormal ultrasound, which demon-
retrobulbar and orbital infiltration with tumor. strates the mass, distinguishing a solid mass from a
Hutchinson syndrome describes widespread bone mar- hydronephrotic kidney. It can also demonstrate lymph
row and bone disease, causing bone pain, limping, and node enlargement, tumor extension, and liver metastasis,
irritability in the younger child. Skin involvement is seen although the CT or magnetic resonance imaging provide
exclusively in infants with Evan’s stage IV-S tumors42 and greater sensitivity (Figures 46-4 to 46-6).
is characterized by nontender, bluish subcutaneous nod- Most centers in the U.S. rely on 99mTc-diphosphonate
ules. Disseminated disease may manifest as failure to scintigraphy for the evaluation of bone disease. Because of
thrive and fever, the latter observed most often in the the biochemical activity of neuroblastoma, meta-iodoben-
presence of bone metastasis. These are usually seen as zylguinidine (MIBG) scintigraphy becomes an attractive
lytic lesions on skeletal radiographs. Rarely, patients may tool.45 The compound is taken up by catecholaminergic
present with the complication of catecholamine secretion cells, which includes most neuroblastomas. Thus, it
by the neuroblastoma, which may include headache, becomes a very specific and sensitive method of assessing
hypertension, palpitations, and diaphoresis. The tumor the primary tumor and focal metastatic disease.
may also lead to watery diarrhea secondary to vasoactive There exists great variability in the number of bone
intestinal polypeptide. marrow aspirates or biopsies done at different institutions.

Table 46-1 Minimum Recommended Tests for Determining Extent of Disease36


Tumor Site Tests

Primary Three-dimensional measurement of tumor by CT scan or MR or ultrasound

Metastases Bilateral posterior iliac bone marrow aspirates and core biopsies (4 adequate specimens necessary to exclude
tumor)

Bone radiographs and either scintigraphy by 99mTc-diphosphonate or 131I-(or 123I-) meta-iodobenzylguanidine


(MIBG) or both

Abdominal and liver imaging by CT scan or MR or ultrasound

Chest radiograph (AP and lateral) and chest CT scan

Markers Quantitative urinary catecholamine metabolites (VMA and HVA)

Note: For evaluation of bone metastases, 99mTc-diphosphonate scintigraphy is recommended for all patients and is essential if MIBG scintigraphy is
negative in bone. (From Brodeur, GM, Castleberry, RP: Neuroblastoma. In Pizzo PA, Poplack DG (eds): Principles and Practice of Pediatric
Oncology, 2nd edition, p 750. Philadelphia, J.B. Lippincott, 1993.)
744 Part VIII Pediatric Malignancies

Recent data support the increased yield of bone marrow


biopsies versus aspirates. The international conferees on
neuroblastoma staging recommended two bone marrow
aspirates and two biopsies, that is one of each from each
of the posterior iliac crests.43 Core biopsy specimens must
contain at least 1 cm of marrow (excluding cartilage).

STAGING
The current, favored staging system is based on clinical,
radiographic, and surgical evaluation of children with
neuroblastoma. The International Neuroblastoma
Staging System (INSS) allows for uniformity in staging
of patients, facilitating clinical trials, and biologic studies
around the world (Table 46-2).43
The Children’s Oncology Group (COG) risk-based Figure 46-4 CT scan of 4-year old with IV contrast showing
schema is based on three factors: patient age at diagnosis, right adrenal neuroblastoma with diffuse calcification, seen
certain biologic characteristics of the patient’s neuroblas- adjacent to liver. A, aorta; K, left kidney. (Courtesy Dr. Edward
toma tumor, and the stage of the tumor as defined by INSS. Weinberger, Seattle, WA.)

PROGNOSTIC CONSIDERATIONS
The COG is investigating a risk-based neuroblastoma
treatment plan that assigns risk-based on patients’ age,
INSS stage, and tumor biology (Table 46-3).46 The treat-
ment plan is based on the risk group, with low-, intermedi-
ate-, and high-risk groups having an overall survival of
>90%, 70% to 90%, and >30%, respectively, 3 years after
diagnosis. The overall prognosis of patients with stages I,
II, and IV-S is between 75% and 90%, while those with
stage III and IV have only a 10% to 30% 2-year disease-
free survival. Age is a sufficient prognostic variable, with
the outcome of infants under 1 year of age being substan-
tially better than those >1 year. Origin of the tumor is
another important variable, as patients with adrenal pri-
maries have poorer outcomes than those with other origins.
A number of biologic variables have been assessed that
may help predict the ultimate outcome for a patient.
Figure 46-5 CT scan of 1-year old with IV contrast showing
Hyperdiploid tumor DNA is associated with a favorable extensive right adrenal neuroblastoma invading into right
prognosis,47 while N-myc amplification is associated with kidney, encircling aorta (A) and extending beyond the midline.
a poor prognosis regardless of patient age.48–50 A high (Courtesy Dr. Edward Weinberger, Seattle, WA.)
proportion of proliferating tumor cells may independ-
ently predict poor prognosis.51 Expression of the gene
encoding one of the high-affinity neurotrophin receptors
(TRK-A) is associated with good prognosis tumors.52 is also used to make a diagnosis, provide tissue for bio-
Increased levels of telomerase RNA,53 elevated serum logic studies, surgically stage the tumor, and to attempt
ferritin,54 elevated serum lactate dehydrogenase,55 and excision of the tumor. Delayed primary or second-look
the presence of neuroblastoma cells in bone marrow dur- surgery determines the response to therapy.
ing or after chemotherapy are each associated with poor Based on the INSS criteria, the operative protocol
prognosis.55–58 incorporates the following: (1) The resectibility of pri-
mary or metastatic tumor should be determined in light
of tumor location, mobility, relationship to major vessels,
TREATMENT
ability to control blood supply, and overall prognosis of
As with most solid malignant neoplasms, complete surgi- patient. Modern chemotherapy effectively consolidates
cal removal is the most effective form of therapy. Surgery and decreases the size of primary tumors and large lymph
Chapter 46 Neuroblastoma 745

Figure 46-6 A, Chest x-ray with subtle right paravertebral curvilinear stripe (arrowheads)
raising possibility of right paravertebral mass. B and C, Coronal T1-weighted MRI images.
B represents more posterior view showing large mass between right kidney and vertebral
bodies. C demonstrates multilobulated right paravertebral mass extending through three
contiguous neural foramina into spinal canal with effacement of spinal cord. (Courtesy Dr.
Edward Weinberger, Seattle, WA.)
746 Part VIII Pediatric Malignancies

Table 46-2 International Neuroblastoma Staging System (INSS)


Stage I Localized tumor confined to the area of origin; complete gross excision, with or without microscopic residual
disease; identifiable ipsilateral and contralateral lymph nodes negative microscopically

Stage IIA Unilateral tumor with incomplete gross excision; identifiable ipsilateral and contralateral lymph nodes negative
microscopically

Stage IIB Unilateral tumor with complete or incomplete gross excision; with positive ipsilateral regional lymph nodes;
identifiable contralateral lymph nodes negative microscopically

Stage III Tumor infiltrating across the midline with or without regional lymph node involvement; or, unilateral tumor
with contralateral regional lymph node involvement; or, midline tumor with bilateral lymph node involvement

Stage IV Dissemination of tumor to distant lymph nodes, bone, bone marrow, liver, and/or other organs (except as
defined in stage IV-S)

Stage IV-S Localized primary tumor as defined for stage 1 or stage 2 with dissemination limited to liver, skin, and/or bone
marrow (limited to infants <1 year of age). Marrow involvement should be minimal (<10% of total nucleated
cells identified as malignant by bone biopsy or dry bone marrow aspirate). More extensive bone marrow
involvement would be considered stage 4 disease

node metastasis. Therefore, there is little place for the ally, followed by mobilizing of spleen, stomach, and pan-
less predictable surgical assaults of previous years. (2) creas. This is best accomplished by maintaining the plane
Nonadherent, intracavitary lymph nodes should be sam- between the mesocolon and Gerota’s fascia. The viscera
pled. Gross examination during surgery may be inaccu- are reflected as far as the midline and placed in an intes-
rate for detecting or ruling out lymph node metastasis in tinal bag, so that the tumor is exposed.
up to 25% of cases.59 Lymph nodes adherent to and The first stage of the operation involves exposure of
removed en bloc with the primary tumor do not alter the the arterial wall from below the distal limit of the tumor
outcome of the patient.60 Ideally, lymph nodes superior to just above the proximal limit of the tumor. The origin
and inferior to the primary tumor should be sought and of the main visceral arteries are often noted at this time.
sampled, and location documented. Lymph node sam- Further dissection focuses on the distal extent of the
pling in patients with high thoracic, low cervical, or large tumor, along the external or common iliac artery.
abdominal primary tumors that are unresectable may be Surgeon and assistant each pick up the adventitia, allow-
problematic. Other prognostic factors may be utilized in ing longitudinal dissection along the middle of the vessel.
these situations, and thus, the value of additional infor- The subadventitial plane has been entered, establishing
mation regarding lymph node involvement is question- the plane for the remainder of the operation. Proximal
able. (3) Routine biopsy of the liver in situations dissection proceeds to the tumor, incising along the mid-
involving an abdominal neuroblastoma without evidence dle of the vessel down to tunica media. Bipolar cautery is
of metastatic disease has been advocated. This practice of considerable benefit.
has come into question, particularly due to the sensitivity Sympathetic nerves on the left will be transected, and
of clinical staging by CT or MR. Until the INSS criteria an attempt should be made to preserve the nerves on the
for staging are prospectively studied in all age groups, right. The aortic bifurcation is encountered and the dis-
routine biopsy of liver during initial surgery in patients section proceeds to the inferior mesenteric artery. If
with primary abdominal tumors remains appropriate. there is a large bulk of tumor, retrograde dissection may
The surgical approach to neuroblastoma is best be helpful. Further dissection cephalad leads to the ori-
regarded as a vascular-type procedure. Neuroblastoma gin of the gonadal arteries and left renal vein crossing the
rarely invades the tunica media of large blood vessels but aorta at the same level. Considerable tumor bulk can be
can involve the tunica adventitia. The appropriate plane encountered in this area. It is usually possible to identify
of dissection usually exists beneath the tunica adventitia. the vein before entering the lumen. Dissection of 4 to 5
Following partial exposure of the circumference of each cm of vein will allow for retraction and adequate dissec-
vessel, the tumor is segmentally removed as complete tion beneath the aorta. Aortic dissection may be difficult
vessel dissection occurs.60 at this level due to tumor adherence and the two gonadal
The approach to an extensive left-sided abdominal arteries. Dividing the gonadal arteries and maintenance
tumor or preaortic tumor is through a long, supraumbil- of the appropriate plane of dissection allow for safe dis-
ical, transverse incision. The left colon is reflected medi- section. Cephalad to the vein is the origin of the left renal
Chapter 46 Neuroblastoma 747

Table 46-3 Children’s Oncology Group Neuroblastoma Risk Group Assignment Schema
INSS Stage Age MYCN Shimada’s Histology DNA Ploidy Risk Group

1 0–21 years Any Any Any Low

2A/2B* <365 days Any Any Any Low

≥365 days to 21 years NonAmp Any — Low

≥365 days to 21 years Amp Fav — Low

≥365 days to 21 years Amp Unfav — High

3† <365 days NonAmp Any Any Intermediate

<365 days Amp Any Any High

≥365 days to 21 years NonAmp Fav — Intermediate

≥365 days to 21 years NonAmp Unfav — High

≥365 days to 21 years Amp Any — High

4† <365 days NonAmp Any Any Intermediate

<365 days Amp Any Any High

≥365 days to 21 years Any Any — High

IV-S‡ <365 days NonAmp Fav >1 Low

<365 days NonAmp Any =1 Intermediate

<365 days NonAmp Unfav Any Intermediate

<365 days Amp Any Any High

Biology defined by MYCN status: amplified (Amp) versus nonamplified (NonAmp)

Shimada’s histopathology: favorable (Fav) versus unfavorable (Unfav)

DNA Ploidy: DI ≥ 1; hypodiploid tumors (with DI <1) will be treated as a tumor with a DI > 1
(DI < 1 [hypodiploid] to be considered favorable ploidy)

*INSS 2A/2B symptomatic patients with spinal cord compression, neurologic deficits or other symptoms will be treated on the LOW RISK NB
Study with immediate chemotherapy for 4 cycles (Course 1).
†INSS 3 or 4 patients with clinical symptoms as above (or in the investigator’s opinion it is in the best interest of the patient) will receive immediate

chemotherapy.
‡INSS IV-S infants with favorable biology and clinical symptoms will be treated on the LOW RISK NB study with immediate chemotherapy until

asymptomatic (2 to 4 cycles). Clinical symptoms defined as: respiratory distress with or without hepatomegaly or cord compression and neurologic
deficit or IVC compression and renal ischemia; or genitourinary obstruction; or gastrointestinal obstruction and vomiting; or coagulopathy with
significant clinical hemorrhage unresponsive to replacement therapy.

artery and just proximal to this is the superior mesenteric SMA usually being the easier vessel to dissect free. Once
artery (SMA). Thus, at the level of the renal artery, the this artery is mobilized, the celiac artery and its branches
plane of dissection should become perpendicular to the are given attention. This area can be difficult but by dis-
anterolateral wall of the artery rather than directly ante- secting as before and maintaining the subadventitial
riorly. This plane is then maintained as far as the plane, success is achieved. Frequently, two diaphragmatic
diaphragm, where normal tissue is usually encountered. branches arise from the trunk of the celiac artery or just
With clearing of the anterior surface of the proximal proximally from the aorta, and these are divided. The left
aorta, the origins of the celiac artery and SMA are iden- gastric artery may also be sacrified without concern.
tified. These vessels are exposed and cleared, with the Once the two main intestinal arteries are immobilized,
748 Part VIII Pediatric Malignancies

the right side of the aorta may be exposed with the right partial response rates ranging from 34% to 45%. A com-
renal artery next being encountered. The tumor at this bination of agents are used that take advantage of drug
location may be attached posteriorly to the crus of the synergism, mechanisms of cytotoxicity, and differences in
hemidiaphragm. Once its main attachments are divided, side effects. Cyclophosphamide and cisplatin are noncell
the tumor is removed. The tumor that has been attached cycle-specific agents often used in combination with cell
between the celiac artery and SMA usually adheres to the cycle-dependent drugs (doxorubicin, teniposide). Using
posterior surface of the pancreas as well. In dissecting these combinations has resulted in improved response
this from the pancreas, the termination of the splenic rates in children with advanced neuroblastoma. The
vein and its junction with superior mesenteric vein are chemotherapy is given in phases, which include induc-
exposed. It is uncommon to see dense adherence in this tion, consolidation, and maintenance phases.
region.60 Following the removal of the preaortic tumor The COG risk stratification schema is currently being
proximally, the left renal artery should be exposed and utilized in the treatment of most patients with neuroblas-
dissected free. This can be fairly tedious but is possible. toma in North America. This risk-based neuroblastoma
When tumor is deep within the renal hilum, nephrec- treatment plan assigns each patient to a low-, intermedi-
tomy may be necessary. Once the renal artery is dis- ate-, or high-risk group. Thus, in patients without
played, the primary tumor can be mobilized and excised. metastatic disease, initial surgery is performed to estab-
The remaining areas to be cleared lie below the level of lish the diagnosis, to resect as much of the primary tumor
the renal vessels and posterior to the length of the aorta. as safely possible, to accurately stage the disease via sam-
These areas do not present any unusual difficulty. As pling of regional lymph nodes that are not adherent to
many as possible of the lumbar arteries should be pre- the tumor, and to obtain adequate tissue for biologic
served, but no untoward consequences have followed studies.
division of up to 5 arteries.60 Finally, if there is consider-
able tumor bulk posterior to the inferior venae cava,
Treatment of Low Risk Disease
reflection of the ascending colon may be necessary to
achieve complete tumor clearance. Treatment for patients categorized as low risk (Table 46-3)
In general, thoracic procedures become more an exer- consists most commonly of surgery alone but in some
cise of rib clearance rather than a vascular-type opera- cases surgery combined with 6 to 12 weeks of
tion. Pelvic tumors may provide some difficulty with chemotherapy. Chemotherapy consists of carboplatin,
regard to access. An extended Pfannenstiel-type incision, cyclophosphamide, doxorubicin, and etoposide. Clearly
altered in that the recti are detached from the pubis, pro- the dose of each agent is kept low in order to minimize
vides optimal exposure, but access may still be difficult. permanent injury from the chemotherapy regimen.63
The anatomy is complicated by the presence of nerves of
the sacral plexus, particularly with formal dissection of
Treatment of Intermediate Risk Disease
the internal iliac vessels. Once again, formal systematic
and planned exposure of the important structures before Patients categorized as intermediate risk (Table 46-3) are
tumor removal is of great value.60 treated with surgery and 12 to 24 weeks of the same
Surgical complication rates in neuroblastoma range chemotherapy regimen as described above.64
from 5% to 25%.61,62 The incidence increases in
instances of aggressive abdominal resection of tumors at
Treatment of High Risk Disease
diagnosis. Reported complications include nephrectomy,
hemorrhage, postoperative intussusception or adhesions, In contrast, patients categorized as high risk (Table 46-3)
injury to renal vessels with subsequent renal failure, and are generally treated with aggressive multiagent
neurologic defects, such as Horner’s syndrome. chemotherapy comprising very high doses of carboplatin,
Avoidance of surgical risk in infants who have better sur- cyclophosphamide, doxorubicin, etoposide, and ifos-
vival is recommended. Downstaging of the tumor with phamide with high-dose cisplatin. After a response to
chemotherapy followed by surgical resection is the chemotherapy, resection of the primary tumor should be
favored approach. attempted, followed by myeloablative chemotherapy,
sometimes total-body irradiation, and autologous stem
cell transplantation. Irradiation of residual tumor and
Chemotherapy
original sites of metastasis is often performed before,
The mainstay of management in neuroblastoma is during or after myeloablative therapy. After recovery,
chemotherapy. A number of effective chemotherapeutic patients are treated with oral 13-cis-retinoic acid for 6
agents have been identified that include cyclophos- months. Both myeloablative therapy and retinoic acid
phamide, cisplatin, doxorubicin, vincristine, etoposide, improve outcome in patients categorized as high risk.65
and teniposide, which alone yield complete response or See Table 46-4 for a summary.
Chapter 46 Neuroblastoma 749

Table 46-4 Disease-Free Survival (2 years) Based on Risk Category and Age
Risk Category Patient Age (years) INSS Stage Two-Years Disease-Free Survival (%)

All 1 >90

All 2A 85

Low <1 2B/3 87/89

<1 4S 57–90

Intermediate >1 2B/3 59

<1 4 75

High >1 4 40/15†


*Personal observation, RP Castleberry.
†Difference relates to complete versus partial surgical resection, respectively. Modified from Brodeur, 1993.

without obtaining a definitive histologic diagnosis and


Radiation Therapy
without surgical intervention, thus avoiding potential
Despite the radiosensitive nature of neuroblastoma in complications of surgery in the newborn.69,70 The expe-
culture models, clinical response has been variable.66 rience with tumors detected by mass urinary cate-
Historically, radiation has been used in the multimodal- cholamine metabolite screening in Japan appears to be
ity management of residual neuroblastoma, bulky unre- applicable to tumors detected by prenatal or perinatal
sectable tumors, and disseminated disease. Radiation ultrasound in the United States. Twenty-six infants with
therapy is reserved for patients with symptomatic life or presumed Evans stages I, II, or IV-S by imaging, urinary
organ threatening tumor that does not respond rapidly VMA, and HVA levels less than 50 mcg/mg of creatinine,
enough to chemotherapy, or for intermediate-risk no tumor involvement of great vessels or invasion into
patients whose tumor has responded incompletely to the spinal canal and tumor size <5 cm were observed with
both chemotherapy and attempted resection and also frequent imaging. Biopsy and tissue diagnosis were not
have unfavorable biologic characteristics. Radiation obtained initially. Progression of tumor size was noted in
therapy to the primary site is recommended for high-risk one-third of the infants with resection without any
patients even in cases of complete resection. apparent increase in stage being accomplished. All had
favorable biologic features. In the remainder, following
observation for 6 to 73 months, no surgery had been per-
Urgent Chemotherapy
formed, the VMA and HVA had normalized, and in sev-
Symptomatic spinal cord compression necessitates eral cases the tumors have become undetectable by
immediate treatment with chemotherapy. Neurologic imaging.69 The COG is currently investigating system-
recovery is more likely the less the severity of compro- atic observation without surgery for infants with pre-
mise and the shorter the duration of symptoms. sumed small Evans stage I adrenal neuroblastoma
Neurologic outcome appears to be similar whether cord detected by prenatal or perinatal ultrasound.
compression is treated with chemotherapy, radiation
therapy, or surgical laminectomy. However, laminectomy
FUTURE CONSIDERATIONS
may result in later scoliosis, and chemotherapy is often
needed whether or not surgery or radiation is used.67,68 The identification of various prognostic markers of neu-
The COG neuroblastoma treatment plan recommends roblastoma allow for a more precise determination of
immediate chemotherapy for cord compression in biologic potential. A statistically significant association
patients classified as low risk or intermediate risk. has been noted with tumors of low histologic grade along
with DI of more than 1 (hyperdiploid), single copy of N-
myc gene per haploid genome, and a serum lactic dehy-
Observation Without Surgery of Localized,
drogenase of <1500 IU/l. High histologic grade was
Presumed Adrenal Neuroblastoma in Infants
associated with a DI of 1, amplified N-myc gene, and an
Studies suggest that selected presumed neuroblastomas LDH of 1500 or more, factors that are associated with
detected in infants by screening may safely be observed aggressive behavior.71 The use of these prognostic factors
750 Part VIII Pediatric Malignancies

allow for appropriate risk-specific therapy and the devel- 7. Beckwith J, Perrin E: In situ neuroblastoma: A
opment of optimal treatment protocols for childhood contribution to the natural history of neural crest tumors.
neuroblastoma. Am J Pathol 1963; 43:1089.
Identification of individuals who are predisposed to 8. Mason GH, Hart-Nercer J, Miller EJ, et al: Adrenaline-
secreting neuroblastoma in an infant. Lancet 1957; 2:322.
develop the tumor has become increasingly important.
9. Knudson AGJ, Strong LC: Mutation and cancer:
Cytogenetic studies have demonstrated alterations of
statistical study of retinoblastoma. J Natl Cancer Inst
chromosome 1p, reaching a frequency of more than 1976; 57:675.
70%.72 Alterations of 1p are also seen in significant fre- 10. Brodeur GM, Fong CT: Molecular biology and genetics
quency in other types of cancers, including colon cancer, of human neuroblastoma. Cancer Genet Cytogenet 1989;
alveolar rhabdomyosarcoma, hepatoblastoma, and ductal 41:153.
carcinoma of the breast.73 Progress is being made that 11. Balaban-Malenbaum G, Gilbert F: Relationship between
should reveal whether specific genetic information is homogeneously staining regions and double minute
commonly altered in neuroblastomas and, if so, what its chromosomes in human neuroblastoma cell lines. Prog
significance is for tumorigenesis. Cancer Res Ther 1980; 12:97.
Urinary catecholamine metabolites are commonly ele- 12. Look AT, Hayes FA, Nitschke R, et al: Cellular DNA
content as a predictor of response to chemotherapy in
vated in neuroblastoma but unfortunately are not as reli-
infants with unrectable neuroblastoma. N Engl J Med
able tumor markers as alpha fetoprotein or beta-human
1984; 311:231.
chorionic gonadotropin for following germ cell tumors. 13. Gansler T, Chatten J, Varello M, et al: Flow cytometric
With greater understanding of the tumor biology, new DNA analysis of neuroblastoma. Correlation with
markers, such as telomerase RNA, serum ferritin, and histology and clinical outcome. Cancer 1986; 58:2453.
neurotrophin receptor, may prove useful for following 14. Taylor SR, Locker J: A comparative analysis of nuclear
response to therapy, as well as early relapse. DNA content and N-myc gene amplification in
Treatment strategies continue to evolve as clinical neuroblastoma. Cancer 1990; 65:1360.
response improves. The goals now are to maximize the 15. Look AT, Hayes FA, Schuster JJ, et al: Clinical relevance
response and yet minimize the overall morbidity of treat- of tumor cell ploidy and N-myc gene amplification in
ment. Newer modalities currently under investigation childhood neuroblastoma. A Pediatric Oncology Group
Study 1997; 9:581.
include monoclonal antibody therapy with or without
16. Biedler JL, Ross RA, Shanske S, Spengler BA: Human
GM-CSF following chemotherapy,74 targeted radiation
neuroblastoma cytogenetics: search for significance of
therapy with 131I MIBG,75 tandem myeloablation and homogeneously staining regions and double minute
stem cell transplantation,76,77 and inclusion of myelobla- chromosomes. Prog Cancer Res Ther 1980; 12:81.
tive doses of 131I MIBG prior to stem cell transplanta- 17. Brodeur GM: Neuroblastoma – clinical applications of
tion.78 The knowledge gained from the ongoing biologic molecular parameters. Brain Pathol 1990; 1:47.
studies will continue to be applied to the clinical arena as 18. Brodeur GM, Seeger RC, Schwab M, et al:
mechanisms of neuroblastoma transformation and pro- Amplification of N-myc in untreated human
gression are elucidated. neuroblastomas correlates with advanced disease stage.
Science 1984; 224:1121.
19. Brodeur GM: Molecular Biology and Genetics of Human
Neuroblastoma. Boca Raton, CRC Press, 1990.
REFERENCES 20. Brodeur GM, Hayes FA, Green AA, et al: Consistent
N-myc copy number in simultaneous or consecutive
1. Virchow R: Die Krankenhaften Geschwulste, Vol 11, neuroblastoma samples from sixty individual patients.
p 149. Berlin, Germany, Hirschwald, 1864. Cancer Res 1987; 47:4248.
2. Marchand F: Beitrage zur Kenntniss der normalen und 21. Nisen PD, Waber PG, Rich MA, et al: N-myc oncogene
pathologischen Anatomic der Glandula carotica und der RNA expression in neuroblastoma. J Natl Cancer Inst
Nebennieren Festschriff fur Rudolph. Virchows Arch 1988; 80:1633.
1891; 5:578. 22. Slavc I, Ellenbogen R, Jung W-H, et al: N-myc gene
3. Herxheimer G: Uebur Tumoren des Nebennierenmarkes, amplification and expression in primary human
insbesondere das Neuroblastoma sympaticum. Beitr neuroblastoma. Cancer Res 1990; 50:1459.
Pathol Anat 1914; 57:112. 23. Fong C, White PS, Peterson K, et al: Loss of
4. Blacklock JWS: Neurogenic tumours of the heterozygosity for chromosomes 1 to 14 defines subsets of
sympathetic system in children. J Pathol Bacteriol advanced neuroblastomas. Cancer Res 1992; 52:1780.
1934; 39:27–48. 24. Young JLJ, Ries LG, Silverberg E, et al: Cancer
5. Phillips R: Neuroblastoma. Ann R Coll Surg Engl 1953; incidence, survival and mortality for children younger
12:29–47. than 15 years. Cancer 1986; 58:598.
6. Cushing H, Wolbach SB: The transformation of a 25. Voute PA: Neuroblastoma. In Sutow WW, Fernbach DJ,
malignant paravertebral sympathicoblastoma into a benign Vietti TJ (eds): Clinical Pediatric Oncology, p 559. St.
ganglioneuroma. Am J Pathol 1927; 3:203. Louis, Mosby, 1984.
Chapter 46 Neuroblastoma 751

26. Miller RW, Fraumeni JF, Hill JA: Neuroblastoma: 42. Evans AE, D’Angio GJ, Randolph JA: A proposed staging
Epidemiologic approach to its origin. Am J Dis Child for children with neuroblastoma. Children’s Cancer Study
1968; 115:253. Group A. Cancer 1971; 27:374.
27. Hope JW, Borns PF, Berg PK: Roentgenologic 43. Brodeur GM, Seeger RC, Barrett A, et al: International
manifestations of Hirschbrung’s disease in infancy. Am criteria for diagnosis, staging and response to treatment in
J Roentgenal Radium Ther Nucl Med 1965; 95:217. patients with neuroblastoma. J Clin Oncol 1988; 6:1874.
28. Dehner LP: Pathologic anatomy of classic neuroblastoma: 44. LaBrosse EH, Com-Nougue C, Zucker JM, et al: Urinary
including prognostic features and differential diagnosis. In excretion of 3-methoxy-4-hydroxymandelic acid and 3-
Pochedly C (ed): Neuroblastoma: Tumor Biology and methoxy-4-hydroxyphenylacetic acid by 288 patients with
Therapy, p 111. Boca Raton, CRC Press, 1990. neuroblastoma and related neural crest tumors. Cancer
29. Triche TJ, Askin FB, Kissane JM: Neuroblastoma, Res 1980; 40:1995.
Ewing’s sarcoma and the differential diagnosis of small-, 45. Voute PA, Hoefnagel CA, Marcuse HR, de Kraker
round-, blue-cell tumors. In Finegold M (ed): Pathology J: Detection of neuroblastoma with 131I-meta-
of Neoplasia in Children and Adolescents, p 145. iodobenzylguanidine. Prog Clin Biol Res 1985; 175:389.
Philadelphia, PA, WB Saunders, 1986. 46. Neuroblastoma Treatment, www.cancer.gov.
30. Shimada H, Chatten J, Newton WA Jr, et al: 47. Ladenstein R, Ambros IM, Pötschger U, et al: Successful
Histopathologic prognostic factors in neuroblastic tumors: treatment of stage III neuroblastoma based on prospective
definition of subtypes of ganglioneuroblastoma and an biologic staging: a Children’s Cancer Group study. J Clin
age-linked classification of neuroblastomas. J Natl Cancer Oncol 1998; 16(4):1256–1264.
Inst 1984; 73:405. 48. Brodeur GM, Azar C, Brother M, et al: Neuroblastoma.
31. O’Neill JA, Littman P, Blitzer P, et al: The role of surgery Effect of genetic factors on prognosis and treatment.
in localized neuroblastoma. J Pediatr Surg 1985; 20:708. Cancer 1992; 70(6 Suppl):1685–1694.
32. Evans AE, D’Angio GJ, Propert K, Anderson J, Hann 49. Look AT, Hayes FA, Shuster JJ, et al: Clinical relevance of
H-WL: Prognostic factors in neuroblastoma. Cancer tumor cell ploidy and N-myc gene amplification in
1987; 59:1853. childhood neuroblastoma: a Pediatric Oncology Group
33. Joshi V, Canto A, Altshuler G, et al: Prognostic study. J Clin Oncol 1991; 9(4):581–591.
significance of histopathologic features of neuroblastoma: 50. Tonini GP, Boni L, Pession A, et al: MYCN oncogene
a grading system based on the review of 211 cases from amplification in neuroblastoma is associated with worse
the Pediatric Oncology Group. Proc Am Soc Clin Oncol prognosis, except in stage 4s: the Italian experience with
1991; 10:311 (abstract). 295 children. J Clin Oncol 1997; 15(1):85–93.
34. Ambros IM, Hata J, Joshi VV, et al: Morphologic features 51. Krams M, Hero B, Berthold F, et al: Proliferation marker
of neuroblastoma (schwannian stroma-poor tumors) in K1-S5 discriminates between favorable and adverse
clinically favorable and unfavorable groups. Cancer 2002; prognosis in advanced stages of neuroblastoma with and
94(5):1574–1583. without MYCN amplification. Cancer 2002;
35. Cohn SL, Rademaker HR, Salwen WA, et al: Analysis of 94(3):854–861.
DNA ploidy and proliferative activity in relation to 52. Nakagawara A, Arima-Nakagawara M, Scavarda NJ, et al:
histology and N-myc amplification in neuroblastoma. Am Association between high levels of expression of the TRK
J Pathol 1990; 136(5):1043–1052. gene and favorable outcome in human neuroblastoma.
36. Takayama H, Suzuki T, Mugishima H, et al: Deletion N Engl J Med 1993; 328(12):847–854.
mapping of chromosomes 14q and 1p in human 53. Poremba C, Hero B, Goertz HG, et al: Traditional and
neuroblastoma. Oncogene 1992; 7(6):1185–1189. emerging molecular markers in neuroblastoma prognosis:
37. Takeda O, Homma C, Maseki N, et al: There may be two the good, the bad and the ugly. Klin Padiatr 2001;
tumor suppressor genes on chromosome arm 1p closely 213(4):186–190.
associated with biologically distinct subtypes of 54. Hann HW, Evans AE, Siegel SE, et al: Prognostic
neuroblastoma. Genes Chrom Cancer 1994; 10(1):30–39. importance of serum ferritin in patients with stages III
38. Seeger RC, Brodeur H, Sather A, et al: Association of and IV neuroblastoma: the Children’s Cancer Study
multiple copies of the N-myc oncogene with rapid Group experience. Cancer Res 1985; 45(6):2843–2848.
progression of neuroblastomas. N Engl J Med 1985; 55. Shuster JJ, McWilliams NB, Castleberry R, et al: Serum
313(18):1111–1116. lactate dehydrogenase in childhood neuroblastoma.
39. Caron H: Allelic loss of chromosome 1 and additional A Pediatric Oncology Group recursive partitioning study.
chromosome 17 material are both unfavorable prognostic Am J Clin Oncol 1992; 15(4):295–303.
markers in neuroblastoma. Med Pediatr Oncol 1995; 56. Berthold F, Trechow R, Utsch S, et al: Prognostic factors
24(4):215–221. in metastatic neuroblastoma. A multivariate analysis of
40. Brown N, Cotterill S, Lastowska M: Gain of chromosome 182 cases. Am J Pediatr Hematol 1992; 14(3):207–215.
arm 17q and adverse outcome in patients with 57. Burchill SA, Lewis IJ, Abrams KR, et al: Circulating
neuroblastoma. N Engl J Med 1999; 340(5):1954–1961. neuroblastoma cells detected by reverse transcriptase
41. Rosen EM, Cassady JR, Frantz CN, et al: Neuroblastoma: polymerase chain reaction for tyrosine hydroxylase
The Joint Center for Radiation Therapy/Dana-Farber mRNA ae an independent poor prognostic indicator in
Cancer Institute/Children’s Hospital experience. J Clin stage 4 neuroblastoma in children over 1 year. J Clin
Oncol 1984; 2:714. Oncol 2001; 19(6):1795–1801.
752 Part VIII Pediatric Malignancies

58. Seeger RC, Reynolds CP, Gallego R, et al: Quantitative 69. Nishihira H, Toyoda Y, Tanaka T, et al: Natural course of
tumor cell content of bone marrow and blood as a neuroblastoma detected by mass screening: a 5-year
predictor of outcome in stage IV neuroblastoma: a prospective study at a single institution. J Clin Oncol
Children’s Cancer Group study. J Clin Oncol 2000; 2000; 18(16):3012–3017.
18(24):4067–4076. 70. Holgersen LO, Subramanian S, Kirpeker M, et al:
59. Smith EI, Nitschke R, Shochat S, et al: Lack of Spontaneous resolution of antenatally diagnosed
significance of involved lymph nodes attached to localized adrenal masses. J Pediatr Surg 1996;
neuroblastoma. A Pediatric Oncology Group study. Proc 31(1):153–155.
Am Soc Clin Oncol 1987; 6:219 (abstract). 71. Joshi VV, Cantor AB, Brodeur GM, et al: Correlation
60. Kiely EM: The surgical challenge of neuroblastoma. between morphologic and other prognostic markers of
J Pediatr Surg 1994; 29:128. neuroblastoma. Cancer 1993; 71:3173.
61. Castleberry RP, Kun L, Shuster JJ, et al: Radiotherapy 72. Brodeur GM, Green AA, Hayes FA, et al: Cytogenetic
improves the outlook for children older than one year features of human neuroblastomas and cell lines. Cancer
with POG stage C neuroblastoma. J Clin Oncol 1991; Res 1981; 41:4678.
9:789. 73. Schwab M: Amplification of N-myc as a prognostic marker
62. Azizkjan RG, Shaw A, Chandler JG: Surgical for patients with neuroblastoma. Semin Cancer Biol 1993;
complications of neuroblastoma resection. Surgery 4:13.
1985; 97:514. 74. Kushner BH, Kramer K, Cheung NK: Phase II trial of
63. Strother DR: Children’s Oncology Group: Phase III the anti-G(D2) monoclonal antibody 3F8 and
Study of Primary Surgical Therapy in Children With granulocyte-macrophage colony-stimulating factor
Low-Risk Neuroblastoma, COG-P9641, Clinical trial, for neuroblastoma. J Clin Oncol 2001; 19(22):
Active. 4189–4194.
64. Baker D: Children’s Oncology Group: Phase III Study of 75. Garaventa A, Bellagamba O, Lo Piccolo MS, et al:
Combination Chemotherapy in Children with 131I-metaiodobenzylguanidine (131I-MIBG) therapy
Intermediate-Risk Neuroblastoma, COG-A3961, Clinical for residual neuroblastoma: a mono-institutional
trial, Active. experience with 43 patients. Br J Cancer 1999;
65. Matthay KK, Villablanca JG, Seeger RC, et al: Treatment 81(8):1378–1384.
of high-risk neuroblastoma with intensive chemotherapy, 76. Frappaz D, Michon J, Coze C, et al: LMCE3 treatment
radiotherapy, autologous bone marrow transplantation, strategy: results in 99 consecutively diagnosed stage 4
and 13-cis-retinoic acid. Children’s Cancer Group. neuroblastomas in children older than 1 year at diagnosis.
N Engl J Med 1999; 341(16):1165–1173. J Clin Oncol 2000; 18(3):468–476.
66. Weichselbaum RR, Epstein J, Little JB: In vitro cellular 77. Kletzel M, Katzenstein HM, Haut PR, et al: Treatment of
radiosensitivity of human malignant tumors. Eur J Cancer high-risk neuroblastoma with triple-tandem high-dose
1976; 36:47. therapy and stem-cell rescue: results of the Chicago Pilot
67. Katzenstein HM, Kent PM, London WB, et al: II Study. J Clin Oncol 2002; 20(9):2284–2292.
Treatment and outcome of 83 children with intraspinal 78. Miano M, Garaventa A, Pizzitola MR, et al: Megatherapy
neuroblastoma: the Pediatric Oncology Group experience. combinint 131I-metaiodobenzylguanidine and high-dose
J Clin Oncol 2001; 19(4):1047–1055. chemotherapy with hematopoietic progenitor cell rescue
68. De Bernardi B, Pianca C, Pistamiglio P, et al: for neuroblastoma. Bone Marrow Transplant 2001;
Neuroblastoma with symptomatic spinal cord 27(6):571–574.
compression at diagnosis: treatment and results with 76
cases. J Clin Oncol 2001; 19(1):183–190.
C H A P T E R

47 Wilms’ Tumor
Michael L. Ritchey, MD, and Max J. Coppes, MD, PhD, MBA

The management of Wilms’ tumor has evolved consider- embryogenesis, have provided important information
ably since its description nearly a century ago by Max regarding cancer biology and development. Of particular
Wilms. Wilms, a German surgeon,1 was the first to pro- interest is Wilms’ tumor, which comprises up to one-
pose that all the various elements of the tumor were third of pediatric embryonal tumors. In North America,
derived from the same cell. His careful pathologic the annual incidence in children under the age of 15
description of this tumor, also referred to as nephroblas- years is 7 per million.6 Therefore, approximately 450
toma, led to the association of his name with this tumor. new cases should be expected each year. The overall
Initially, surgery held the only hope of cure. The first median age at diagnosis is 3.5 years, although for boys it
attempt at a nephrectomy in a child was probably per- is 6 months lower than for girls. The disease occurs
formed by Hueter in 1876.2 Unfortunately, the patient died nearly equally in girls and boys worldwide.7 The inci-
during the surgical procedure. Ten years later, Kocher, in dence shows some ethnic variation with slightly higher
Bern, Switzerland, performed the first successful nephrec- rates reported for the black populations and a lower inci-
tomy in a child. At first the ultimate outcome for patients dence in Asian children. Environmental factors are not
with Wilms’ tumor remained poor. By the late 1930s, that important a role in the etiology of Wilms’ tumor.8
improvements in surgical technique provided a survival of Of greater interest is the genetic epidemiology of
30%. The introduction of radiation therapy as an effective Wilms’ tumor. For many years Wilms’ tumor has been
adjuvant treatment resulted in improved survival,3 associated with a number of congenital abnormalities,
although it was not until the introduction of effective including sporadic aniridia, hemihypertrophy, and geni-
chemotherapy in the 1950s that a dramatic improvement in tourinary malformations.9,10 A number of recognizable
survival was achieved.4 First, Farber from Boston reported syndromes are associated with an increased incidence of
the effectiveness of dactinomycin (AMD), then workers at Wilms’ tumor (Table 47-1). These may be divided into
M.D. Anderson Cancer Center introduced vincristine syndromes characterized by overgrowth and those lack-
(VCR) for the treatment of this disease.5 Due to the rarity ing overgrowth.
of this tumor it was recognized in the early 1970s that mul- Genitourinary abnormalities, such as hypospadias,
ticenter collaboration was necessary to conduct the needed cryptorchidism, and renal fusion anomalies, are seen in
clinical research to improve outcome. The cooperative association with Wilms’ tumor in 4.5% of patients with
group studies conducted by the National Wilms’ Tumor Wilms’ tumor.11 Of particular interest is the recognition
Study Group (NWTSG) in North America and the of the Denys-Drash syndrome12,13 in children presenting
International Society of Paediatric Oncology (SIOP) in with ambiguous genitalia. The Denys-Drash syndrome is
Europe have ultimately provided excellent survival rates for characterized by ambiguous genitalia and renal mesangial
children with this malignant disorder. This chapter reviews sclerosis, usually resulting in end-stage renal failure and
the major contributions of these clinical trials, some of the Wilms’ tumor.14 Genetic studies indicate that the Denys-
molecular genetic changes associated with this tumor, and Drash syndrome is associated with specific mutations of
outlines the current treatment strategies. WT1, one of the Wilms’ tumor genes on the short arm
of chromosome 11.15,16 It should be noted that the phe-
notype can vary considerably. Any patient with Wilms’
EPIDEMIOLOGY
tumor and renal dysfunction, with or without a geni-
Embryonal neoplasms of childhood, named because their tourinary abnormality, should be evaluated for a WT1
morphologic appearance resembles that observed during mutation.

753
754 Part VIII Pediatric Malignancies

Table 47-1 Incidence of Congenital Anomalies months is the appropriate screening interval. Tumors
Associated with Wilms Tumor Patients Reported to detected by screening will generally be a lower stage.22,23
the National Wilms’ Tumor Study Retrospective reviews have been unable to determine if
Anomaly Rate (per 1000) early detection had an impact on patient survival.
Children with BWS can also develop nonmalignant renal
lesions.24 Recognition of these benign lesions is impor-
Aniridia 7.6 tant to avoid unnecessary nephrectomy when new lesions
BWS 8.4
are identified on screening ultrasound.

Hemihypertrophy 33.8
GENETICS
Genitourinary anomalies Our knowledge of the genetic alterations involved in the
Hypospadias 13.4
development of Wilms’ tumor has grown substantially
over the past two decades. Initially, the genetics of
Cryptorchidism 37.3 Wilms’ tumor was thought to be very similar to that of
retinoblastoma (a childhood tumor of the retina), since
Hypospadias and both tumors can occur bilaterally and/or affect several
cryptorchidism 12.0 members of the same family, although the latter is more
common in retinoblastoma than it is in Wilms’ tumor.
Also, as in retinoblastoma, when Wilms’ tumor occurs
The incidence of aniridia in patients with Wilms’ bilaterally, the age at presentation is younger than in
tumor is 1.1%, much higher than the incidence in the cases where the disease is limited to one kidney, suggest-
general population, which is estimated at 1:50,000 to ing a constitutional defect that predisposes that child to
1:100,000. Wilms’ tumor will develop in >30% of chil- tumor formation. The younger age at diagnosis is also
dren with the aniridia, genitourinary malformations and found in children with specific, Wilms’ tumor predispos-
mental retardation (AGR syndrome). In the presence of ing, congenital anomalies, such as aniridia.11 However, it
Wilms’ tumor, the term WAGR syndrome is used is now clear that the genetic alterations leading to the
(Wilms’ tumor and AGR). Most children with the development of Wilms’ tumor are more complex than
WAGR syndrome suffer from a constitutional deletion those of retinoblastoma and in fact involves multiple
on band 13 of the short arm of chromosome 11.17 genes.25
Syndromes with overgrowth features at risk for the Analysis of the different median ages at presentation
development of Wilms’ tumor include hemihypertrophy, between groups of patients led Alfred Knudson in the
which may occur alone or as part of the Beckwith- early 1970s to propose a “two-hit” model for the devel-
Wiedemann syndrome (BWS), Perlman, Soto, and the opment of retinoblastoma and Wilms’ tumor.26 His
Simpson-Golabi-Behmel syndromes.11,18,19 BWS is a hypothesis predicts that two rate-limiting events are nec-
rare congenital overgrowth disorder characterized by vis- essary for tumor development. He predicted that chil-
ceromegaly (splenomegaly, hepatomegaly), macroglossia, dren with a genetic susceptibility to cancer already have
and hyperinsulinemic hypoglycemia.18,19 Most cases of a constitutional (prezygotic) genetic alteration, either
BWS are sporadic, but 15% exhibit heritable character- inherited from one of the parents or resulting from a
istics with apparent autosomal dominant inheritance. spontaneous (de novo) mutation. In these children, only
Patients with BWS are predisposed for certain cancers, in one additional (postzygotic) genetic alteration would suf-
particular embryonal neoplasms (adrenocortical neo- fice for Wilms’ tumor to develop. By contrast, children
plasms and hepatoblastoma). The incidence of Wilms’ that do not have a constitutional genetic alteration, and
tumor in this patient population is <5%. Children with therefore are not susceptible to cancer, require two
BWS found to have nephromegaly (kidneys greater than postzygotic (or somatic) genetic events for Wilms’ tumor
or equal to the 95th percentile of age adjusted renal to develop. Since the probability of one somatic event
length) are at the greatest risk for the development of occurring is higher than that of two independent ones,
Wilms’ tumor.20 Children with BWS are also at an children with a constitutional genetic alteration have a
increased risk to develop bilateral tumors.21 A review of greatly increased likelihood of developing cancer as com-
NWTS-3 and NWTS-4 patients found that 21% of the pared to children that do not carry a constitutional
BWS patients had either synchronous or metachronous genetic defect.
bilateral tumors. Two genetic regions, both on the short arm of chro-
Screening with serial renal ultrasounds has been rec- mosome 11, have been linked to Wilms’ tumor develop-
ommended in children with aniridia, hemihypertrophy, ment. The first evidence of band 13 of the short arm of
and BWS. Review of most studies suggests that 3 to 4 chromosome 11 (11p13) being involved in the development
Chapter 47 Wilms’ Tumor 755

of Wilms’ tumor was the detection of cytogenetically vis- short arm of chromosome 1, shown in ~11% of tumors39,
ible deletions of this region in patients with the WAGR are clinically of greatest importance. In a preliminary
syndrome. The deletion in these patients is not restricted study, tumor-specific loss of either region (16q or 1p) was
to their Wilms’ tumor cells but is present in all body cells correlated with poor outcome, independently from stage
(i.e., constitutional deletions).27 According to Knudson’s or histology at initial presentation.39 A larger group of
two-hit hypothesis, the constitutional 11p13 deletion was patients has been studied to confirm this observation.
proposed to represent the first of two events, the second
event being somatic in these patients. Similarly, the loss
of heterozygosity (LOH) for 11p13 DNA markers CLINICAL PRESENTATION
demonstrated in approximately one-third of sporadic The typical presentation of Wilms’ tumor is that of a
Wilms’ tumors26,28 also suggested that chromosome large asymptomatic abdominal mass, often found inci-
11p13 was involved in the development of Wilms’ tumor. dentally. Other presentations include acute abdominal
In fact, both observations implied the presence of a pain mimicking appendicitis or some other intraabdomi-
tumor suppressor gene at 11p13, a notion that was con- nal event. This is often the result of tumor rupture with
firmed in 1990.29–31 Once cloned, alterations of the hemorrhage into the retroperitoneum or the peritoneal
Wilms’ tumor suppressor gene WT1 at 11p13 were cavity. Hematuria is found in approximately one-fourth
shown in tumor DNA obtained from patients with spo- of children at diagnosis, but only gross hematuria war-
radic Wilms’ tumors, as well as in the constitutional rants further evaluation to exclude tumor extension into
DNA of patients with a genetic predisposition to Wilms’ the renal pelvis and ureter. Hypertension is also present
tumor.32 WT1, which is expressed transiently in the in about 25% of cases.40
developing kidney and also in specific cells of the gonads, Vascular tumor extension into the renal vein and infe-
is required for normal genitourinary development and rior vena cava (IVC) can result in atypical presenta-
specifically of importance for the differentiation of the tions.41,42 For example, a varicocele, particularly one that
renal blastema.33 Constitutional point mutations in the persists when the child is supine, may be secondary to
zinc finger DNA-binding region of WT1 give rise to the obstruction of the spermatic vein due to tumor throm-
Denys-Drash syndrome, characterized by pseudoher- bus. However, varicocele, hepatomegaly due to hepatic
maphroditism, early renal failure with diffuse mesangial vein obstruction, ascites, or congestive heart failure are
sclerosis, and Wilms’ tumor.16 only found in less than 10% of patients with intracaval or
A second Wilms’ tumor locus (already designated atrial tumor extension.41 Very rarely the tumor can
WT2) has been identified on chromosome 11p15.5.34 embolize to the pulmonary artery with catastrophic
This same chromosomal region has also been linked to the consequences.
BWS.35 Although this region has revealed at least 10
imprinted (expressed preferentially from one of the parent
alleles) genes, for now, it is not known whether a single PREOPERATIVE EVALUATION
gene is responsible for both disorders or adjacent genes Laboratory evaluation should include a complete blood
independently for each disease. The presence of imprinted count, liver function tests, and renal function tests.
genes and the observation that in Wilms’ tumors showing Serum calcium should also be checked. Hypercalcemia
LOH for 11p15 DNA markers, it is invariably the mater- should alert the physician for the possible presence of
nal allele that is lost36, suggest that the paternal and mater- either congenital mesoblastic nephroma (CMN) or rhab-
nal genes in this region are functionally unequal. In doid tumor of the kidney (RTK), rather then Wilms’
keeping with this, some BWS patients have been shown to tumor.40 A coagulation screen should be obtained to
have two paternal copies of chromosome 11p in addition exclude acquired von Willebrand’s disease. The coagula-
to the maternal copy (trisomy 11p15), while in some tion defect underlying the acquired von Willebrand’s dis-
patients with BWS who seem to have grossly normal kary- ease in patients with Wilms’ tumor can be corrected
otypes (i.e., no trisomy), both copies of chromosome 11 preoperatively with the administration of DDAVP.
are in fact found to be inherited from the father, a condi- Modern imaging modalities provide detailed informa-
tion termed uniparental isodisomy.37 Apparently in BWS, tion regarding origin and extent of intra-abdominal
expression is required from two paternal 11p15 alleles, tumors. There has been considerable debate regarding
while in Wilms’ tumor the first of two events almost the role of imaging studies in the staging and manage-
always occurs on the paternal 11p15 allele. ment of children with Wilms’ tumor.43,44 Imaging studies
Other molecular abnormalities studied in Wilms’ can often establish the correct diagnosis before surgery
tumor patients seem to be related to outcome rather than (Figure 47-1). In addition, imaging studies can help the
development. Of these, loss of chromosomal material, as surgeon to plan for a major surgical procedure. There is
determined by LOH, on the long arm of chromosome an increased incidence of surgical complications in
16, demonstrated in ~20% of tumors36,38, and on the Wilms’ tumor patients with an incorrect preoperative
756 Part VIII Pediatric Malignancies

dered by unopacified blood due to an inadequate bolus


injection of contrast. If the ultrasound is inconclusive,
magnetic resonance imaging (MRI) is recommended to
establish IVC patency.42,46,47
One of the unanswered questions regarding imaging is
the role of CT and MRI scans in determining the extent of
disease, i.e., stage.43,44 Since survival is correlated with
stage (and histology), NWTSG investigators keep
stressing the need for accurate staging.48 The current
NWTSG staging system utilizes surgery and pathology
findings only. There have been no studies to compare data
obtained by imaging to those obtained by the surgeon dur-
ing nephrectomy in assessing extrarenal extent.
Occasionally, preoperative CT can suggest extracapsular
tumor extension when there is irregularity of the tumor
margin or evidence of regional adenopathy when lymph
nodes are demonstrated physically separate from the
Figure 47-1 CT scan of a large left Wilms’ tumor with a
tumor mass. However, enlarged retroperitoneal lymph
small rim of functioning renal parenchyma. nodes are common in children and often reactive.
Moreover, even intraoperative lymph node evaluation by
the surgeon does not correlate well with histopathologic
diagnosis.45 Preoperative imaging will establish the pres- findings49 and one would therefore expect similar difficul-
ence of a functioning contralateral kidney prior to per- ties when correlating histopathology with imaging studies.
forming a nephrectomy. Preoperative imaging can be useful to identify groups
Ultrasonography is the initial investigation performed of patients that are best treated with chemotherapy
in a child with a palpable abdominal mass. This study can prior to attempts at surgical excision.50,51 One important
distinguish between solid and cystic lesions, and also group is children with bilateral Wilms’ tumors (BWTs).
determine if the mass is of intrarenal or extrarenal in ori- The recommended treatment of these children is preop-
gin. During ultrasonography, the patency of the IVC erative chemotherapy to shrink the tumors rather than
should be assessed to exclude extension of tumor throm- upfront surgery. This approach facilitates preservation of
bus into the IVC (Figure 47-2) that occurs in 4% of renal parenchyma.50,52 CT and MRI will detect most
Wilms’ tumor patients.41,42 Computed tomography (CT) bilateral lesions but can miss some lesions <1 cm in
may be useful, but visualization of the IVC is often hin- diameter.53 Primary nephrectomy has been shown to be

Figure 47-2 Ultrasound that depicts intracaval thrombus (arrow).


Chapter 47 Wilms’ Tumor 757

associated with an increased incidence of surgical com- life.66,67 Neuroblastoma is the most common solid
plications in patients with either extensive intravascular abdominal malignancy in childhood but in most cases can
tumor involvement or tumors that require en bloc resec- be distinguished by its extrarenal location and excretion
tion of other organs to achieve complete excision.45 As of catecholamines and its metabolites in the urine.
noted above, IVC extension can and should be identified Rarely, neuroblastoma will arise within the kidney mak-
during the preoperative imaging evaluation. ing the distinction difficult. Renal angiomyolipoma is
Identifying visceral involvement by CT is much more only rarely seen in childhood and is associated with
difficult. The majority of children identified as having tuberous sclerosis.68,69 Demonstration of fat within a
possible invasion of the liver on CT later prove to be renal lesion by CT scan preoperatively should alert the
negative at surgery.54 CT may be able to detect deep surgeon of the presence of this tumor.
seated liver metastases but is much more predictive for Another tumor commonly seen in infancy is CMN.
absence of liver invasion.55 This is generally a benign lesion found in approximately
The most common site of distant metastases is within 2% of childhood renal tumors. Most patients are under
the lungs, present at diagnosis in 8% of patients. The the age of 6 months at the time of diagnosis.70 In fact
majority of these lesions can be identified on routine the typical presentation is that of a newborn with an
chest x-ray films. However, some children are only iden- abdominal mass. A number of cases have been diag-
tified to have pulmonary metastases by chest CT.56,57 nosed prenatally (Figure 47-3). However, FH Wilms’
There continues to be some debate regarding the man- tumor and RTK can also present in the first few months
agement of these patients.58,59 Not all small lesions seen of life.63,71 Nephrectomy is curative for most patients
on CT are due to metastatic disease. Some lesions iden- with CMN,70 although there have been reports of local
tified on CT only represent benign lesions, e.g., histo- recurrence, more common in those with cellular histol-
plasmosis, granuloma or round atelectasis. An important ogy and occasionally metastases.72–74 Adequacy of surgi-
question is whether chest x-ray-negative, CT-positive cal resection and age at diagnosis in these patients
patients require the same adjuvant treatment as those appear to be more important predictors of relapse than
with a positive x-ray.59,60 Meisel et al.60 reviewed 53 chil- histology.75
dren enrolled in NWTS-3 and NWTS-4 who had a neg- In addition to age at presentation, other clinical
ative chest roentgenogram but were identified with parameters can aid the clinician in narrowing the diag-
metastatic lesions on a chest CT. There was no difference nostic possibilities. For example, a renal mass developing
in survival in those patients treated according to the local in a child with hemihypertrophy, BWS, or aniridia is
extent of the abdominal disease alone. They did note most likely to be a Wilms’ tumor. Bilateral or multicen-
fewer pulmonary relapses in patients that did not receive tric renal tumors are also highly suggestive of Wilms’
pulmonary irradiation, but more deaths are attributable tumor.
to lung toxicity. Other investigators have also noted an
increased risk of recurrent disease in favorable histology
(FH) patients with pulmonary densities detected on CT PATHOLOGY
alone when pulmonary irradiation was omitted.56,61 On gross examination, Wilms’ tumor is usually a soft,
Uncommon sites of metastases can occur in renal lobulated and tan or light gray in color. Hemorrhagic or
tumors other then Wilms’ tumor. A radionuclide bone necrotic areas are frequently noted. The microscopic fea-
scan and skeletal survey are both recommended if a his- tures of Wilms’ tumor classically consist of three compo-
tologic diagnosis of clear cell sarcoma of the kidney nents in varying proportions: blastema, stroma, and
(CCSK) or renal cell carcinoma (RCC) is confirmed. epithelium. Tumors that consist predominately of one of
Both of these have a propensity to metastasize to the these elements are encountered in 60% of cases and still
skeleton.44,48,62,63 RTK and CCSK are associated with considered Wilms’ tumor.76 Blastemal predominant
brain metastases, and MRI of the brain should be tumors are important in that they behave more aggres-
obtained in the early postoperative period. sively with early metastatic spread and more advanced
disease at presentation.77 The stromal component can
DIFFERENTIAL DIAGNOSIS differentiate into striated muscle, cartilage, or fat. Wilms’
tumors with predominant rhabdomyomatous elements
RTK, CCSK, and RCC do not have any distinguishing were once thought to behave less aggressively, but recent
radiographic features that will allow a preoperative diag- evidence suggests that they are no different from the typ-
nosis unless unusual sites of metastases (bone, brain) are ical, triphasic, Wilms’ tumor.76
evident.62–67 RTK is more typically seen in infants and The most important determinants of outcome are
very young children with a mean age of 13 months. RCC histopathology and tumor stage. Analysis of early
generally presents after the age of 5 years, and it is the NWTS patients identified a group of patients with unfa-
most common renal malignancy in the second decade of vorable histopathologic features, associated with
758 Part VIII Pediatric Malignancies

Figure 47-3 CT scan of a large CMN in a 2-day-old infant that was detected on
antenatal ultrasound.

increased rates of relapse and death.78 These “unfavor- are further stratified into focal and diffuse anaplasia,
able histology” tumors are responsible for 50% of tumor based on a topographic principle.82 Focal anaplasia
deaths in the NWTSG studies but account for only 10% requires that the anaplastic nuclear changes be confined
of patients.79 Three distinct tumors were originally to a specified region of the primary tumor and absent
included in this unfavorable subgroup: (a) Wilms’ tumor from the surrounding portions of the lesion. Diffuse
with extreme nuclear atypia (anaplasia); (b) RTK; (c) anaplasia on the other hand is diagnosed when anaplasia
CCSK. The latter two tumor groups have been reclassi- is present in more than one portion of the tumor or if
fied and are now considered to be distinct entities from found in any extrarenal or metastatic site. Confirming
Wilms’ tumors. CCSK accounts for 3% of renal tumors the notion that anaplasia is more a marker of chemore-
reported to the NWTSG. Its recognition is quite impor- sistance than inherent aggressiveness of the tumor, out-
tant in order that appropriate adjuvant therapy be insti- come is generally excellent only if the tumor is
tuted. The use of doxorubicin has resulted in an completely removed, i.e., stage I tumor.83
improved outcome for these children.48,62 RTK is a
highly malignant tumor that accounts for 2% of renal Precursor Lesions
tumors registered to the NWTSG.48,63 RTK is now con-
sidered a sarcoma of the kidney and not of metanephric Lesions apparently representing Wilms’ tumor precur-
origin.49 The prognosis of RTK remains dismal with sors are found in 30% to 40% of kidneys removed for
conventional chemotherapeutic regimens and new treat- Wilms’ tumor.83 Nephrogenic rests are defined as foci of
ment strategies are being developed for the management abnormally persistent nephrogenic cells that can develop
of these children. into Wilms’ tumor.84 Two distinct categories of nephro-
Anaplasia is a feature of Wilms’ tumor that is associ- genic rests have been identified. The most common is
ated with resistance to chemotherapy. It is therefore not the perilobar nephrogenic rest (PLNR) that is found in
surprising that the incidence of anaplasia reported by the the periphery of the kidney. There is a higher prevalence
NWTSG investigators (5%) is very similar to that of PLNR in children with Wilms’ tumor and hemihy-
reported in the SIOP studies (5.3%),76 where all tumors pertrophy or the BWS (Table 47-2). Intralobar nephro-
are pretreated with chemotherapy, but the anaplasia per- genic rests (ILNRs) on the other hand can be found
sists after treatment.80 Anaplastic features are rare in the anywhere within the renal lobe (Figure 47-4). Children
first 2 years of life, but the incidence increases to 13% in with the WAGR syndrome or the Denys-Drash syn-
children age 5 years or older.81 Anaplastic Wilms’ tumors drome are more likely to have ILNRs. In the presence of
Chapter 47 Wilms’ Tumor 759

multiple or diffuse nephrogenic rests the term fies patients at risk for the development of contralateral
“nephroblastomatosis” is used. Wilms’ tumor. This is particularly the case for children
The natural history of nephrogenic rests, occasionally younger than 12 months of age who have PLNRs.86
found in kidneys without tumors,85 is not entirely clear. These and other high-risk children need careful follow-
Some undergo spontaneous regression, others hyperplas- up imaging of the contralateral kidney (Table 47-3).
tic changes, producing relatively large tumors. While the
histopathologic differential diagnosis between hyperplas-
Biologic Parameters
tic nephrogenic rests and Wilms’ tumor can be very dif-
ficult (the exact criteria for distinguishing these two As survival of children with FH Wilms’ tumor has con-
lesions remain to be defined), the surgical management tinued to improve, traditional staging factors, e.g., tumor
of both disorders is similar. size, histology, lymph node metastases, are now less able
Multiple rests in one kidney are highly suggestive of to predict risk for tumor progression or relapse in FH
the presence of nephrogenic rests in the other kidney patients. Stratification of FH Wilms’ tumor patients into
since these lesions usually involve both kidneys. low-risk and high-risk groups for relapse, independent of
Consequently, the presence of ILNRs or PLNRs identi- tumor stage, would allow intensification of treatment for
those with an increased risk of relapse. Research has now
Table 47-2 Prevalence of Nephrogenic Rests focused on molecular or biologic factors. The NWTSG
Patient Population ILNR (%) PLNR (%) has established a central tumor bank to maintain biologic
specimens from all patients entered on the study.
As noted previously, LOH for a portion of the long
Infant autopsies 0.01 1 arm of chromosome 16 has been found in 20% of Wilms’
tumor patients.25,38 A prospective study of 232 patients
Renal dysplasia Unknown 3.5
registered on the NWTS found that patients with 16q
Unilateral Wilms’ tumor 15 25 LOH had a statistically significantly poorer 2-year
relapse-free and overall survival than those without LOH
Synchronous bilateral for chromosome 16q.39 In addition, LOH for chromo-
Wilms’ tumor 34–41 74–79 some 1p markers, occurring in approximately 11% of
Wilms’ tumors,39 is also associated with an increased risk
Metachronous bilateral 63–75 42
Wilms’ tumor of relapse, although of borderline statistical significance.
Another potential marker is telomerase; a reverse
Beckwith-Wiedemann, 47–56 70–77 transcriptase that maintains chromosome ends, compen-
hemihypertrophy sating for the loss of DNA that occurs in replication.
High telomerase activity has been found to be an unfa-
Aniridia 84–100 12/20
vorable prognostic feature for several types of cancers. In
Drash syndrome 78 11 a case-cohort study of 78 patients with FH Wilms’
tumor, telomerase enzyme activity, expression of hTR,

Figure 47-4 A, Illustration of renal lobe showing characteristic locations of ILNR (dark gray) and PLNR (black). (From Beckwith
JB: Med Pediatr Oncol 1993; 21:158–168, with permisssion). B, PLNR composed of blastemal cells just beneath the renal
capsule (H&E 40×).
760 Part VIII Pediatric Malignancies

Table 47-3 Recommended Follow-up Imaging Studies for Children with Renal Neoplasms of Proven Histology
and Free of Metastases at Diagnosis
Tumor Type Study Schedule Following Therapy

Favorable histology Wilms’ tumor; Chest films 6 weeks and 3 months postop; then q. 3
Stage I anaplastic Wilms’ tumor q. 6 months × 3, yearly × 2

Irradiated patients only Irradiated bony structures* Yearly to full growth, then q. 5 years indefinitely†

Without NRs, Stages I and II Abdominal ultrasound Yearly × 3

Without NRs, Stage III Abdominal ultrasound As for chest films

With NRs, any stage‡ Abdominal ultrasound q. 3 months × 10, q. 6 months × 5, yearly × 5

Stage II and III anaplastic Chest films As for favorable histology

Abdominal ultrasound q. 3 months × 4; q. 6 months × 4

RCC Chest films Like favorable histology

Skeletal survey and Like CCSK


bone scan

CCSK Brain MRI and/or opacified When CCSK is established; then q. 6 months × 10
CT skeletal survey and
bone scan

Chest films As for favorable histology

Rhabdoid tumor Brain MRI and/or As for CCSK


opacified CT

Chest films As for favorable histology

Mesoblastic nephroma§ Abdominal ultrasound q. 3 months × 6

Modified from D’Angio GJ, Rosenberg H, Sharples K, Kelalis P, Breslow N, Green DM: Med Pediatr Oncol 1993; 21:205–212, with permission.
*To include any irradiated osseous structures.
†To detect second neoplasms, benign (osteochondromas) or malignant.
‡The panelists at the first International Conference on Molecular and Clinical Genetics of Childhood Renal Tumors; Albuquerque, New Mexico,

May 1992, recommended a variation: q. 3 months for 5 years or until age 7, whichever comes first.
§Data from the files of Dr. J.B. Beckwith reveal that 20 of 293 MN patients (7%) relapsed or had metastases at diagnosis; 4 of the 20 in the lungs,

one of the 4 at diagnosis. All but 1 of the 19 relapses occurred within 1 year. Chest films for MN patients may be elected on a schedule, such as
q. 3 months × 4, q. 6 months × 2.

the RNA component of telomerase, and mRNA expres- otypic abnormalities in number are labeled aneuploid.
sion of high telomerase reverse transcriptase (hTERT), Aneuploid DNA histograms are found more commonly
the gene that encodes the catalytic component of the in anaplastic Wilms’ tumors,88 although it is not neces-
enzyme, were measured.87 All had detectable expression sarily associated with poor prognosis.89 While tetraploid
of hTR and 97% had detectable hTERT transcript. The histograms in stage III and stage IV patients are indica-
hTERT mRNA levels correlated with the risk of recur- tive of poor outcome, it remains to be determined
rence even after adjustment for tumor stage. A larger whether DNA ploidy is a more accurate predictor of sur-
study is underway to determine whether this is an inde- vival than histology and stage.90 More recently, investiga-
pendent prognostic indicator. tors have started using cDNA array technology to
The proliferative rate of tumor cells can be estimated develop tumor-specific molecular profiles in the hope to
by the measurement of DNA content. Several studies distinguish bad from good actors among the low-stage
have correlated cell DNA content, measured by flow FH tumors. While a promising approach, solid data have
cytometry, and prognosis in Wilms’ tumor patients. yet to be published.
Normal somatic cells have a diploid DNA content, cells The growth of solid tumors is critically dependent on
in mitosis are tetraploid and tumor cells with gross kary- the induction of neovascularity by angiogenic cytokines.
Chapter 47 Wilms’ Tumor 761

Table 47-4 Staging System of the National Wilms Tumor Study


Stage I Tumor limited to the kidney and completely excised. The renal capsule is intact and the tumor was not ruptured
prior to removal. There is no residual tumor

Stage II Tumor extends through the perirenal capsule, but is completely excised. There may be local spillage of tumor
confined to the flank or the tumor may have been biopsied. Extrarenal vessels may contain tumor thrombus or be
infiltrated by tumor

Stage III Residual non-hematogenous tumor confined to the abdomen: lymph node involvement, diffuse peritoneal spillage,
peritoneal implants, tumor beyond surgical margin either grossly or microscopically, or tumor not completely
removed

Stage IV Hematogenous metastases to lung, liver, bone, brain, etc.

Stage V Bilateral renal involvement at diagnosis

Vascular endocrine growth factor (VEGF) is an angio- patients from NWTS-3 those developed tumor
genic cytokine detected with increased frequency and relapse.95 Tumor extension into the soft tissues of the
quantity in experimental and clinical specimens of sinus or the presence of tumor cells in blood or lymphatic
Wilms’ tumor.91 In experimental animals, lung metas- vessels of the renal sinus is now considered stage II.
tases were far more likely to occur in animals with VEGF Tumors that penetrate the renal capsule or the presence
positive tumors. The potential of anti-VEGF therapy to of tumor cells in the perirenal fat are also classified as
suppress tumor growth has also been assessed.92,93 stage II tumors. Tumor spillage during removal of the
Tumors were induced in kidneys of nude mice by the tumor increases the stage. Local spills confined to the
injection of tumor cells. After 1 week, administration of renal bed are considered stage II and do lead to an
anti-VEGF antibody resulted in a >95% reduction in increased risk of abdominal relapse.96 Diffuse spill into
tumor weight and abolished lung metastases. the peritoneal cavity is considered stage III.
In Wilms’ tumor there are no biologic markers avail- Wilms’ tumor usually metastasizes to regional lymph
able yet that could mimic the function of alpha fetoprotein nodes, the lungs, and the liver. Patients with lymph node
in the management of germ cell tumors. Nevertheless, metastases are classified as stage III and require local radi-
several biologic markers have been studied, including ation therapy following surgery. Since the presence of
serum renin and prorennin, serum erythropoietin, neu- lymph node metastases mandates more intensive treatment
ron-specific enolase (NSE), and hyaluronic acid (HA), HA and is associated with poorer outcome, it is important for
stimulating activity (HASA), and hyaluronidase.40 the surgeon to obtain adequate lymph node sampling dur-
ing nephrectomy in order to allow appropriate staging. In
patients that do not have lymph nodes examined patholog-
PROGNOSTIC CONSIDERATIONS
ically, there is an increased incidence of abdominal relapse,
Histopathology and tumor stage are the most important most notable for stage I patients.96 Approximately 85% of
predictors of survival in Wilms’ tumor patients. The cur- hematogenous metastases at presentation are reported in
rent staging system is summarized in Table 47-4. For the lungs, while about 15% present with liver involve-
NWTS-4, the distribution by stage of FH tumors was ment.97 Other sites of metastases, such as bone, medi-
stage I 40%, stage II 27%, stage III 22%, and stage IV astinum, and spinal cord, are uncommon.
11%.94 Both the surgeon and pathologist have responsi-
bility for determining local tumor stage. Surgical and
SURGICAL MANAGEMENT
pathologic features that are associated with an increased
risk for relapse lead to a higher-stage designation. Although most of the improvement in survival of chil-
Stage I tumors are limited to the kidney and com- dren with Wilms’ tumor is attributable to introduction of
pletely resected. NWTSG pathologists have identified effective chemotherapy and radiation therapy, surgery
several pathologic variables predictive of tumor relapse in continues to play a very important role in its successful
patients with stage I FH tumors.95 These are invasion of treatment. The preoperative evaluation of a child with an
the tumor capsule, presence of an inflammatory pseudo- abdominal mass can generally be completed in 48 hours
capsule, renal sinus invasion, and tumor in the intrarenal in most medical centers. Emergent operation is not nec-
vessels. Analysis of these variables found that one or essary unless there is evidence of active bleeding. The
more of these features were present in 24 stage I FH surgeon is responsible for safely removing the tumor and
762 Part VIII Pediatric Malignancies

assessing the local tumor stage. This requires a thorough Occasionally, patients will present with massive
exploration of the abdominal cavity via a generous tumors that are surgically unresectable. Heroic surgery
transperitoneal incision. The flank approach should not with radical en bloc resection of the tumor and adjacent
be used, as regional staging and examination of the con- organs is not justified. Such operations are associated
tralateral kidney are not possible. The liver is palpated with increased surgical morbidity.45 In addition, the gross
and regional lymph nodes are examined for evidence of appearance of the tumor at the time of surgery can be
tumor spread. misleading in interpreting tumor extent. These tumors
The colon and its mesentery are then reflected off the often compress and adhere to adjacent structures without
tumor and a radical nephrectomy is performed with sam- frank invasion; and in the majority of cases, tumor inva-
pling of regional lymph nodes. Formal lymph node dis- sion is not confirmed after the adjacent visceral organs
section is not required.49 Ligation of the renal vessels is are removed.45 The appropriate management of tumors
performed prior to mobilization of the tumor, but only if that are inoperable at presentation is tumor biopsy
exposure is adequate. More importantly, the surgeon followed by chemotherapy. This approach almost
should be certain that the contralateral renal vessels, always reduces the bulk of the tumor and renders it
aorta, iliac, or superior mesenteric arteries have not been resectable.104,105 Usually, there is adequate size reduction
mistakenly ligated.98 Gentle handling of the tumor within 6 weeks. Serial imaging studies are helpful in
throughout the procedure is mandatory to avoid tumor assessing response. Patients who fail to respond to
spillage as these patients have an increased risk for chemotherapy can subsequently be considered for preop-
abdominal relapse.48,96 Additionally, patients with diffuse erative irradiation that may produce enough shrinkage to
tumor spill are considered stage III and require more facilitate nephrectomy. One disadvantage of using preop-
intensive therapy with whole abdominal irradiation and erative therapy is that there is loss of important staging
the use of doxorubicin. information. Patients with unresectable tumors treated
Prior to ligation of the renal vein, palpation of the with preoperative chemotherapy with or without addi-
vein and IVC should be performed to exclude intravas- tional radiation therapy should be considered stage III
cular tumor extension. The majority of patients with and treated accordingly.105 Therefore, it is very impor-
IVC extension will fortunately be identified by imaging tant that tumors are not determined to be unresectable
studies obtained prior to surgery.41,42,99 Extension of on the basis of preoperative imaging alone.
tumor into the extrarenal vessels does not adversely The morbidity of the surgical procedure should not be
affect prognosis if it is completely excised. For vena caval overlooked in the management of children with Wilms’
involvement below the level of the hepatic veins, the tumor. NWTS-4 patients undergoing primary nephrec-
caval thrombus can be removed via cavotomy after prox- tomy had an 11% incidence of surgical complications.106
imal and distal vascular control is obtained. Generally, The most common complications encountered are hem-
the thrombus will be free-floating, but if there is adher- orrhage and small bowel obstruction45,106,107 SIOP
ence of the thrombus to the caval wall, the thrombus can investigators have reported a lower rate of complications
often be delivered with the passage of a Fogarty or Foley when nephrectomy is performed after preoperative
balloon catheter. Patients with atrial extension may chemotherapy.108 A prospective study comparing the
require cardiopulmonary bypass for thrombus incidence of surgical complications between NWTSG
removal.100 Certain operative findings may suggest and SIOP is underway.
intravascular extension when it has not been correctly
diagnosed preoperatively. For example, excessive bleed-
COOPERATIVE GROUP TRIALS
ing from dilated superficial and retroperitoneal collater-
National Wilms’ Tumor Study Group
als is a clue to obstruction of the vena cava. More
ominous is the finding of sudden unexplained hypoten- Many early accomplishments in the treatment of children
sion, which can result from embolization of the tumor with Wilms’ tumor were made by individuals and large
thrombus.101 single institutions, but as survival improved larger num-
Primary surgical resection of inferior vena caval or bers of patients were needed to conduct prospective ran-
atrial tumor extension is associated with increased surgi- domized trials to answer therapeutic questions. Thus, in
cal morbidity.45 Several reports have demonstrated that North America, both pediatric cooperative groups, the
these patients can best be managed by shrinkage of tumor Children’s Cancer Group (CCG), and the Pediatric
and thrombus with preoperative chemotherapy.42,51,102 Oncology Group (POG) initially initiated clinical trials
This approach facilitates complete removal of the tumor within their own group but subsequently decided to col-
with decreased morbidity. While there has been one laborate within the NWTSG. Meanwhile, investigators
report of tumor embolus during chemotherapy,103 this in Europe formed the Société International d’Oncologie
complication can also occur prior to or during surgical Pédiatrique (SIOP) in an attempt to improve the out-
removal of the tumor.101 come of children with Wilms’ tumor.
Chapter 47 Wilms’ Tumor 763

NWTS-1 and NWTS-2 NWTS-4


There were many important findings as a result of the The goals of the NWTS-4 (1986 to 1994) were to fur-
early clinical trials NWTS-1 (1969 to 1973) and NWTS- ther decrease treatment intensity for patients with favor-
2 (1974 to 1978).109,110 For example, postoperative local able prognosis, while trying to maintain their excellent
irradiation was shown to be unnecessary for group survival. In addition, this was the first clinical pediatric
I patients. Also, combination chemotherapy of vin- cancer trial to evaluate the economic impact of two dif-
cristine (VCR) and AMD was found to be more effec- ferent treatment approaches. Pulse-intensive regimens
tive than the use of either drug alone. The addition of utilized simultaneous administration of agents at more
doxorubicin (DOX) was shown to improve survival of frequent intervals to decrease the number of clinic visits
higher-stage patients. However, even more important and hence the cost of cancer treatment. NWTS-4
findings were the identification of unfavorable histo- demonstrated that while the administered drug dose-
logic features of Wilms’ tumor, and of prognostic fac- intensity was greater on pulse-intensive regimens, these
tors that allowed refinement of the staging system regimens, which indeed are cost effective, produce less
stratifying patients into high-risk and low-risk treat- hematologic toxicity than the standard regimens.113
ment groups.111 After the completion of NWTS-1 and Patients treated with pulse-intensive regimens achieved
NWTS-2, it was recognized that the presence of lymph equivalent survival compared to those treated with stan-
node metastases had an adverse outcome on survival. dard chemotherapy regimens.94 Treatment durations of
Children with lymph node metastases, as well as those approximately 6 and 15 months were found to be equally
with diffuse tumor spill, were found to be at increased effective in patients with stages II to IV/FH tumors.114
risk of abdominal relapse. Therefore, such patients were Children with anaplastic Wilms’ tumors were ran-
considered stage III and given whole abdominal irradi- domized in NWTS-3 and NWTS-4 to receive VCR,
ation. These findings were incorporated into the design AMD, DOX, or those three drugs with the addition of
of NWTS-3 to try and decrease the intensity of therapy cyclophosphamide. There was no difference in outcome
for the majority of low-risk patients while maintaining between the regimens for children with focal anaplasia
overall survival. who had a prognosis similar to that for FH patients.115
For stages II to IV diffuse anaplasia, the addition of
cyclophosphamide to the three-drug regimen improved
NWTS-3
the 4-year relapse-free survival (27.2% versus 54.8%).
In NWTS-3, patients with stage I, FH Wilms’ tumor
were treated successfully with a 10-week regimen of
NWTS-5
VCR and AMD, considerably decreasing the amount of
chemotherapy administered and, as a consequence, the The recently closed intergroup study, NWTS-5, was
total duration of treatment.48 The 4-year relapse-free a single-arm therapeutic trial. Patients were not
survival was 89%, and the overall survival was 95.6%. randomized for therapy, but instead biologic features of
Stage II FH patients treated with AMD and VCR with- the tumors were prospectively assessed. The goal of this
out postoperative radiation therapy (XRT) had an equiv- study was to verify the preliminary findings that LOH for
alent survival, 4-year overall survival of 91.1%, to chromosomes 16q and 1p are useful in identifying
patients that received the same treatment plus DOX, patients at increased risk for relapse.39 However, many
demonstrating the cardiotoxic drug DOX is not neces- other biologic factors can be studied with the use of
sary for the successful treatment of this group of patients. banked tumor specimens collected on all enrolled
The dosage of abdominal irradiation for stage III FH patients.87 This tumor bank is available to all investiga-
patients was reduced to 1080 cGy. This was shown to be tors and will be useful to evaluate new prognostic factors
as effective as 2000 cGy in preventing abdominal relapse that may be identified in the future. If molecular genetic
if DOX was added to VCR and AMD. The 4-year markers are predictive of clinical behavior, they may be
relapse-free survival for stage III patients was 82% in used in subsequent clinical trials to further stratify patients
NWTS-3, the 4-year overall survival was 90.9%. for therapy. Accrual of patients for the NWTS-5 study was
Patients with stage IV FH tumors received abdominal completed in late 2001. At this time, results from NWTS-
(local) irradiation based on the local tumor stage. In addi- 5 have not been published, as data need further matura-
tion, they all received 1200 cGy to both lungs. In combi- tion. Current recommendations for treatment are outlined
nation with VCR, AMD, and DOX, the 4-year in Table 47-5. These represent the treatment regimens used
relapse-free survival was 79% and the overall survival was in NWTS-5. Treatment for patients with stage I or stage II
80.9%.112 There was no statistically significant improve- FH and stage I anaplastic Wilms’ tumor is the same.
ment in survival when cyclophosphamide was added to Patients with stage III FH and stage II and stage III
the three-drug regimen. focal anaplasia are treated with AMD, VCR, and DOX
764 Part VIII Pediatric Malignancies

Table 47-5 Treatment Protocol for National Wilms Tumor Study-5


Stage/Histology Radiation Therapy Chemotherapy

Stages I and II FH None EE-4A; pulse-intensive AMD plus VCR (18 weeks)

Stage I anaplasia

Stages III and IV FH 1080 cGy* DD-4A; pulse-intensive AMD, VCR and DOX (24 weeks)

Stages II to IV focal anaplasia

Stages II to IV diffuse anaplasia Yes† Regimen I: AMD, VCR, DOX, CPM, and etoposide

Stages I to IV CCSK

Stages I to IV rhabdoid Yes† Regimen RTK: carboplatin, etoposide, and CPM tumor of the
kidney

*Stage IV/FH patients are given radiation based on the local tumor stage.
†Radiation therapy is given to all CCSK and RTK patients. Consult protocol for specific treatment.

AMD, dactinomycin; VCR, vincristine; DOX, doxorubicin; CPM, cyclophosphamide; FH, favorable histology.

and 1080 cGy abdominal irradiation. Patients with stage rationale for the SIOP approach is that preoperative
IV FH tumors receive abdominal irradiation based on chemotherapy will make the tumor less prone to intraop-
the local tumor stage and 1200 cGy to both lungs. erative rupture or spill.119–120 The European investigators
Children with stages II to IV diffuse anaplasia and stages also indicate that preoperative therapy leads to a more
I to IV CCSK are treated with a regimen combining favorable stage distribution at the time of surgery,122 as a
VCR, DOX, cyclophosphamide, and etoposide. Children result of which fewer patients will require postoperative
with all stages of RTK were treated with carboplatin, radiation therapy. Preoperative treatment can indeed pro-
cyclophosphamide, and etoposide. All these patients duce dramatic reduction in the size of the primary tumor
receive irradiation to the tumor bed. facilitating surgical excision (Figure 47-5).
One portion of the NWTS-5 study examined the role The SIOP investigators use tumor extent as found at
of surgery alone for children under 2 years of age with the time of delayed (postchemotherapy) surgery to deter-
stage I FH tumors weighing under 550 g has been pub- mine local tumor stage. However, this postchemotherapy
lished.116 This study was based on preliminary observa- stage may inadequately define the risk of intra-abdominal
tion of favorable outcomes on small numbers of such recurrence in nonirradiated patients.121 The SIOP stag-
patients when postoperative adjuvant therapy had been ing system separates stage II into “node-negative” and
omitted.117,118 It was suspended when the number of “node-positive” groups. In SIOP-6, patients with “post-
tumor relapses exceeded the limit allowed by the design chemotherapy stage II” were randomized to receive or
of the study. Seventy-five patients were enrolled. The not receive abdominal irradiation. An unacceptable
sites of relapse were the lung in 5 children and the renal increase in the number of intra-abdominal recurrences in
bed in 3 patients. The 2-year disease-free survival rate nonirradiated patients was noted, although survival rates
was 86.5%. The 2-year survival rate of this cohort of for both groups were not significantly different.121 As a
patients with small tumors is 100% with a median follow- result, stage II node-negative patients are now given an
up of 2.84 years,116 and extended follow-up continues. anthracycline as part of the chemotherapy regimen.
Observation of untreated children may yield interesting Consequently, a larger percentage of SIOP patients are
information on the role of chemotherapy in decreasing now receiving a cardiotoxic drug than do patients regis-
the incidence of contralateral relapse in patients with tered to NWTSG studies. The significance of this is dis-
NRs. Three of these children (4%) have developed cussed later in the section on late effects. Of interest,
metachronous contralateral tumors.116 when one compares the numbers of patients receiving
abdominal irradiation, the incidence has decreased for
both groups. The overall survival figures are comparable
International Society of Pediatric Oncology
between the SIOP and NWTSG.114,123,124
The randomized trials conducted by SIOP differ from The SIOP group has studied the pathologic changes
those of the NWTSG in that patients receive therapy in the tumor after preoperative therapy to help guide
prior to surgery. Also, no routine histopathologic diagno- treatment decisions. This approach features the impor-
sis is obtained prior to commencing treatment. The tance of Wilms’ tumor responsiveness as distinguished
Chapter 47 Wilms’ Tumor 765

Figure 47-5 A, MRI scan of a large inoperable Wilms’ tumor. B, After 6 weeks
of chemotherapy the same tumor has dramatically decreased in size.

from its aggressiveness; the latter is more important in highly differentiated epithelial type. In the latest SIOP
determining tumor stage at diagnosis. In SIOP-9, patho- Wilms’ study, stage I low-risk tumors did not receive
logic review of the resected specimens after chemother- chemotherapy after nephrectomy.
apy revealed completely necrotic (defined as <1% of Intermediate-risk tumors were defined as mixed
viable tumor) Wilms’ tumor in approximately 10% of type nephroblastoma, focal anaplasia, regressive type
specimens.125 Of these patients, 98% had no evidence nephroblastoma, epithelial type nephroblastoma, and
of disease at 5 years (1 nontumor death). These patients stromal type nephroblastoma.126 High-risk tumors are
are now considered to be “low-risk tumors” by SIOP. diffuse anaplasia and nephroblastoma-blastemal type.
The other low-risk tumors are CMN, cystic partially dif- The latter is defined as viable tumor in more than 1/3 of
ferentiated nephroblastoma, nephroblastoma with the mass and 2/3 of the viable tumor consists of blastema.
fibroadenomatous features, and nephroblastoma of Although the proportion of patients with blastemal
766 Part VIII Pediatric Malignancies

predominant tumors is decreased after chemotherapy, patients treated with preoperative chemotherapy com-
these patients had a 31% relapse rate in the SIOP-9 pared to 1.6% of children undergoing primary nephrec-
study.125 These patients require more intensive treatment tomy in NWTS-4. Epithelial predominant tumors also
after nephrectomy. were noted with increased frequency. The latter two
types are often associated with a poor clinical response to
therapy as determined by preoperative imaging. There is
BILATERAL WILMS’ TUMORS
a less marked reduction in tumor volume after
Synchronous BWT occurs in about 5% of children with chemotherapy but lower rates of relapse.125,130 The prog-
metachronous lesions developing in only 1%.50,52,127 nosis for patients with these tumor types is excellent if
Therapy for patients with BWT is focused on sparing the tumors are completely excised. These tumors do
renal parenchyma to decrease the risk for renal failure seem to be more resistant to therapy when seen at higher
noted in these children.50,52,127,128 Review of NWTSG stages. Another histologic subtype that often fails to
patients found that 9.1% of children with synchronous shrink with chemotherapy is fetal rhabdomyomatous
BWT and 18.8% of metachronous BWT developed nephroblastoma.131,132 Some tumors with complete
renal failure.129 The most common cause of renal failure necrosis and predominantly regressive changes can
was bilateral nephrectomy for persistent or recurrent increase in size during therapy.
tumor in the remaining kidney after initial nephrectomy. There is a 10% incidence of anaplasia in BWT.50,128
Therefore, avoiding total nephrectomy at initial surgery The incidence of anaplasia is higher in girls than
is advised. The NWTSG recommends that all children boys.50,128 There can be discordance in the pathology
with BWT should receive chemotherapy prior to surgi- between the two kidneys. Tumor size or other gross fea-
cal resection. tures does not correlate with histology, and careful exam-
At initial surgical exploration, bilateral biopsies should ination of both specimens is required.
be obtained for histopathologic confirmation of disease This information demonstrates the importance of
in both kidneys and definition of the histologic subtype. obtaining biopsy specimens in patients those are not
Examination for extrarenal spread is performed. If suspi- responding to chemotherapy. In some of these children,
cious lymph nodes or other disease is noted, this should further chemotherapy will not lead to tumor shrinkage.
be biopsied and a surgical stage assigned. In NWTS-5, Therefore, if the patient has not responded, reexplo-
patients with FH and stage I or stage II disease were ration with biopsy of the tumor should be performed in
given AMD and VCR, while those with stage III or stage 12 weeks. At the time of the second look procedure, par-
IV FH also received DOX. tial nephrectomy or wedge excision of the tumor is per-
The patient is given 6 weeks of chemotherapy and formed whereever possible.
then reassessed with imaging studies. Experience from If extensive tumor involvement precluding partial
SIOP has shown that most of the reduction in tumor vol- resection is still present after preoperative therapy,
ume will occur early. In SIOP-9, the reduction in tumor complete excision of tumor from the least involved kid-
volume was 48% after 4 weeks and increased to 62% ney is performed. If this leaves a viable kidney, nephrec-
after 8 weeks of chemotherapy.123,124 Although this dif- tomy of the other kidney is carried out. Occasionally,
ference was not of significance for unilateral tumors, fur- bilateral nephrectomy is indicated. This will result in
ther shrinkage may facilitate a parenchymal sparing the need for dialysis. Indeed the most common cause of
approach. renal failure in NWTS patients is bilateral nephrec-
It is important to recognize that response of the tumor tomy for persistent tumor.129 If transplantation is later
by imaging does not always correlate with the histologic considered, a waiting period of 2 years is recommended
response. A clinically good response (by imaging) is usu- to ensure that the patient does not develop metastatic
ally associated with a pathologically good response in disease.133
terms of regressive histologic changes.130 The converse is
not always true.
TREATMENT OF RELAPSES
Important data on histology of postchemotherapy
specimens has been collected by SIOP investigators. Children with relapsed Wilms’ tumor have a variable
Although this information has only been reported for prognosis, depending on the initial stage, site of relapse,
children with unilateral tumors, the information can be time from initial diagnosis to relapse and prior therapy.
extrapolated to those with BWT. The distribution of his- Adverse prognostic factors include relapse <12 months
tologic subtypes is different in children treated with pre- after diagnosis, prior treatment with DOX, and intra-
operative chemotherapy compared to those undergoing abdominal relapse in patients who had previously
primary surgery.83,125,130 Differentiation of the tumor received abdominal irradiation.134 The risk of tumor
after chemotherapy has been noted by several investiga- relapse in NWTS-3 at 3 years was 9.6%, 11.8%, 22%,
tors. Stromal predominant tumors were noted in 14% and 22%, respectively, for stages I to IV. Relapses
Chapter 47 Wilms’ Tumor 767

occurred in 36% of stages I to III and 45% of stage IV hyperfiltration injury.144,145 There is both clinical and
patients with unfavorable histology.48 experimental evidences of hyperfiltration damage of
remnant nephrons after a loss of renal mass.146 Most
LATE EFFECTS OF CANCER TREATMENT experimental studies involve a loss of >3/4 of the total
renal mass, but there is only limited data assessing renal
One consequence of the tremendous improvement in function long term in children following unilateral
survival of children with malignancies is that there are nephrectomy.147,148 While several studies have found no
many long-term survivors who have been exposed to significant alterations in renal function following unilat-
both radiation and chemotherapeutic agents. These eral nephrectomy for Wilms’ tumor,149–151 other reports
children require long-term surveillance, as the sequelae of have noted an increased incidence of proteinuria and
this therapy may not be evident for many years. One decreased glomerular filtration rate (GFR).147,148,152,153
of the more serious concerns is the increased risk for However, clinically important hypertension or renal
second malignant neoplasms (SMN). Investigators from insufficiency is rare.
the NWTSG have noted a 1.6% cumulative incidence of One of the more important risk factors for a decrease in
SMNs at 15 years posttreatment.135 Prior treatment GFR is the amount of radiation to the remaining kidney.
for relapse, the amount of abdominal irradiation, and use of The correlation of functional impairment with the renal
DOX were all associated with an increased incidence radiation dose was reported by Mitus et al.154 a review of
of SMNs. 100 children treated for Wilms’ tumor. The incidence of
Another risk associated with DOX is cardiac toxicity. impaired creatinine clearance was significantly greater for
Congestive heart failure is a well-known acute complica- children who received >1200 cGy to the remaining kidney,
tion of treatment with anthracycline and the incidence is and all cases of overt renal failure occurred in patients who
dose related.136 For NWTS-1, NWTS-2, NWTS-3, and had received more than 2300 cGy.
NWTS-4 patients, the cumulative frequency of conges- The concern of renal dysfunction has led some centers
tive heart failure was 4.4% at 20 years after diagnosis to consider parenchyma-sparing procedures for unilateral
among patients treated initially with doxorubicin.137 The tumors.155,156 Because the majority of tumors are too
relative risk of congestive heart failure was increased in large for partial nephrectomy at presentation, the patient
females by cumulative doxorubicin dose, lung irradiation, must be given preoperative chemotherapy, after that less
and left abdominal irradiation. than 10% of patients may then be amenable to partial
Numerous other organ systems are subject to the late nephrectomy. However, in order to scientifically justify
sequelae of anticancer therapy. An early report of this approach, many children would have to be treated
NWTSG patients found that musculoskeletal problems, accordingly to demonstrate its benefits since the inci-
such as scoliosis, were 7 times more common in children dence of renal failure is quite low for unilateral Wilms’
those were treated with radiation.138 A more recent tumor. A review of the NWTSG database found that
review of 2778 NWTSG patients found that reduction in <0.2% of unilateral Wilms’ tumor patients developed
stature following RT to the pediatric spine is dose renal failure. Moreover, the majority of these patients suf-
dependent and age dependent. However, average height fered from the Denys-Drash syndrome, a disorder
deficits observed at maturity for children receiving doses whereby all patients invariably develop end-stage renal
currently recommended by the NWTSG are clinically disease irrespective of the cancer therapy.129 A recent
nonsignificant.139 report from the NWTSG did find an increased risk of
Damage to reproductive systems can lead to problems renal failure in children with Wilms’ tumor who also had
with hormonal dysfunction and/or infertility. Female aniridia or genitourinary abnormalities.157 The cumula-
Wilms’ tumor patients who received abdominal radiation tive risk of renal failure at 20 years was 38% for the
have a 12% incidence of ovarian failure.140 In addition, patients with aniridia.
women with prior abdominal radiation have the potential
for adverse pregnancy outcomes.141,142 Perinatal mortal-
ity rates are higher, and infants are more likely to have REFERENCES
low birth weights.143 Gonadal radiation in males can
result in temporary azoospermia and hypogonadism.144 1. Zantinga AR, Coppes MJ: Max Wilms (1867–1918): the
man behind the eponym. Med Pediatr Oncol 1992;
The severity of damage to the testis is dependent on the
20:515–518.
dose or radiation. 2. Zantinga AR, Coppes, MJ: Historical aspects of the
There has been concern about the late occurrence of identification of the entity Wilms’ tumor, and of its
renal dysfunction in children who have undergone management. Hematol Oncol Clin North Am 1995;
nephrectomy. Patients have been found to develop pro- 9:1145–1155.
teinuria, hypertension, and renal insufficiency secondary 3. Priestly JT, Schulte TL: The treatment of Wilms’ tumor.
to focal glomerulosclerosis, which has been attributed to Urology 1942; 47:7–10.
768 Part VIII Pediatric Malignancies

4. Farber S: Chemotherapy in the treatment of leukemia and Wiedemann syndrome and Wilms’ tumor: a report from
Wilms’ tumor. JAMA 1966; 198:826–836. the National Wilms’ Tumor Study Group. J Clin Oncol
5. Sutow WW: Chemotherapy in childhood cancer (except 2000; 18:2026–2031.
leukemia). An appraisal. Cancer 1965; 18:1585. 22. Green DM, Breslow NE, Beckwith JB, Norkool P:
6. Birch JM, Breslow N: Epidemiologic features of Wilms’ Screening of children with hemihypertrophy, aniridia, and
tumor. Hematol Oncol Clin North Am 1995; 9:1157–1178. Beckwith-Wiedemann syndrome in patients with Wilms’
7. Breslow N, Olshan A, Beckwith JB, et al: Ethnic variation tumor: a report from the National Wilms’ Tumor Study.
in the incidence, diagnosis, prognosis and follow-up of Med Pediatr Oncol 1993; 21:188–192.
children with Wilms’ tumor. J Natl Cancer Inst 1994; 23. Choyke PL, Siegel MJ, Craft AW, et al: Screening for
86:49–51. Wilms’ tumor in children with Beckwith-Wiedemann
8. Olshan AF, Breslow NE, Daling JR, et al: Wilms’ tumor syndrome or idiopathic hemihypertrophy. Med Pediatr
and paternal occupation. Cancer Res 1990; 50:3212–3217. Oncol 1999; 32:196–200.
9. Miller RW, Fraumeni JF, Manning MD: Association of 24. Choyke PL, Siegel MJ, Oz O, et al: Nonmalignant renal
Wilms’ tumor with aniridia, hemihypertrophy and other disease in pediatric patients with Beckwith-Wiedemann
congenital malformations. N Engl J Med 1964; syndrome. Am J Roentgenol 1998; 171:733–737.
270:922–927. 25. Coppes MJ, Haber DA, Grundy P: Genetic events in the
10. Clericuzio CL: Clinical phenotypes and Wilms’ tumor. development of Wilms’ tumor. N Engl J Med 1994;
Med Pediatr Oncol 1993; 21:182–187. 331:586–590.
11. Breslow NE, Beckwith JB: Epidemiological features of 26. Knudson AG, Strong LC: Mutation and cancer: a model
Wilms’ tumor: results of the National Wilms’ Tumor for Wilms’ tumor of the kidney. J Natl Cancer Inst 1972;
Study. J Natl Cancer Inst 1982; 68:429–436. 48:313–324.
12. Denys P, Malvaux P, Van Den Berghe H, Tanghe 27. Koufos A, Hansen MF, Lampkin BC, et al: Loss of alleles
W, Proesmans W: Association d’un syndrome at loci on human chromosome 11 during genesis of
anatomo-pathologique de pseudohermaphrodisitism Wilms’ tumor. Nature 1984; 309:170–172.
masculin, d’une tumeur de Wilms, d’une nephropathie 28. Huff V: Inheritance and functionality of Wilms’ tumor
parenchymateuse dt d’un mosaicism XX/XY. Arch Fr genes. Cancer Bull 1994; 46:255–259.
Pediatr 1967; 24:729–39. 29. Bonetta L, Kuehn SE, Huang A, et al: Wilms’ tumor
13. Drash A, Sherman F, Hartmann WH, Blizzard RM: locus on 11p13 defined by multiple CpG island-associated
A syndrome of pseudohermaphroditism, Wilms’ tumor, transcripts. Science 1990; 250:994–997.
hypertension and degenerative renal disease. J Pediatr 30. Call KM, Glaser T, Ito CY, et al: Isolation and
1970; 76:585–593. characterization of a zinc finger polypeptide gene at the
14. Jadresic L, Leake J, Gordon I, et al: Clinicopathologic human chromosome 11 Wilms’ tumor locus. Cell 1990;
review of twelve children with nephropathy, Wilms’ 60:509–520.
tumor, and genital abnormalities (Drash syndrome). 31. Gessler M, Poustka A, Cavenee W, et al: Homozygous
J Pediatr 1990; 117:717–725. deletion in Wilms’ tumour of a zinc-finger gene
15. Pelletier J, Bruening W, Kashtan CE, et al: Germline identified by chromosome jumping. Nature 1990;
mutations in the Wilms’ tumor suppressor gene are 343:774–778.
associated with abnormal urogenital development in 32. Coppes MJ, Campbell CE, Williams BRG: The role of
Denys-Drash syndrome. Cell 1991; 67:437–447. WT1 in Wilms’ tumorigenesis. FASEB J 1993; 7:886–895.
16. Coppes MJ, Huff V, Pelletier J: Denys-Drash syndrome: 33. Kreidberg JA, Sariola H, Loring JM, et al: WT-1 is
relating a clinical disorder to genetic alterations in the required for early kidney development. Cell 1993; 74:679.
tumor suppressor gene WT1. J Pediatr 1993; 34. Reeve AE, Sih SA, Raizis AM, Feinberg AP: Loss of
123:673–678. allelic heterozygosity at a second locus on chromosome 11
17. Riccardi VM, Sujansky E, Smith AC, Francke U: in sporadic Wilms’ tumor cells. Mol Cell Biol 1989;
Chromosomal imbalance in the aniridia-Wilms’ tumor 44:711–719.
association: 11p interstitial deletion. Pediatrics 1978; 35. Koufos A, Grundy P, Morgan K, et al: Familial
61:604–610. Wiedemann-Beckwith syndrome and a second Wilms’
18. Sotelo-Avila C, Gonzalez-Crussi F, Fowler JW: Complete tumor locus both map to 11p15.5. Am J Human Genet
and incomplete forms of Beckwith-Wiedemann 1989; 44:711–719.
syndrome: Their oncogenic potential. J Pediatr 1980; 36. Coppes MJ, Bonetta L, Huang A, et al: Loss of
96:47–50. heterozygosity mapping in Wilms’ tumor indicates the
19. Beckwith JB: Macroglossia, omphalocele, adrenal involvement of three distinct regions and a limited role
cytomegaly, gigantism and hyperplastic visceromegaly. for non-disjunction or mitotic recombination. Genes,
Birth Defects: Original Article Series 1969; 5:188–196. Chromosomes & Cancer 1992; 5:326–334.
20. Debaun MR, Siegel MJ, Choyke PL: Nephromegaly in 37. Grundy P, Telzerow P, Haber D, et al: Chromosome 11
infancy and early childhood: a risk factor for Wilms’ uniparental isodisomy predisposing to embryonal
tumor in Beckwith-Wiedemann syndrome. J Pediatr 1998; neoplasms. Lancet 1991; 338:1079.
132:401–404. 38. Maw MA, Grundy PE, Millow LJ, et al: A third Wilms’
21. Porteus MH, Norkool P, Neuberg D, et al: tumor locus on chromosome 16q. Cancer Res 1992;
Characteristics and outcome of children with Beckwith- 52:3094–3098.
Chapter 47 Wilms’ Tumor 769

39. Grundy PE, Telzerow PE, Breslow N, et al: Loss of 57. Wooton SL, Rumack CM, Albano EA, et al: Pulmonary
heterozygosity for chromosomes 16q and 1p in Wilms’ metastases in Wilms’ tumor. Pediatr Radiol 1994; 24:463.
tumor predicts an adverse outcome. Cancer Res 1994; 58. Cohen MD: Current controversey: Is CT scan of the
54:2331–2333. chest needed in patients with Wilms’ tumor? J Pediatr
40. Coppes MJ: Serum biological markers and paraneoplastic Hematol Oncol 1994; 16:191.
syndromes in Wilms’ tumor. Med Pediatr Oncol 1993, 59. Green DM, Fernbach DJ, Norkool P, Kollia G, D’Angio
21:213–221. GJ: The treatment of Wilms’ tumor patients with
41. Ritchey ML, Kelalis PP, Breslow N, et al: Intracaval and pulmonary metastases detected only with computed
atrial involvement with nephroblastoma: Review of tomography: a report from the National Wilms’ Tumor
National Wilms’ Tumor Study-3. J Urol 1988; Study. J Clin Oncol 1991; 9:1776–1781.
140:1113–1118. 60. Meisel JA, Guthrie KA, Breslow NE, et al: Significance
42. Shamberger RC, Ritchey ML, Haase GM, et al: and management of computed tomography detected
Intravascular extension of Wilms’ tumor: Report of the pulmonary nodules: a report from the National Wilms’
National Wilms’ Tumor Study Group. Ann Surg 2001; Tumor Study Group. Int J Radiat Oncol Biol Phys 1999;
234:116–121. 44:579–585.
43. Cohen MD: Staging of Wilms’ tumor. Clin Radiol 1993; 61. Owens CM, Veys PA, Pritchard J, et al: Role of chest
47:77–81. computed tomography at diagnosis in the management
44. D’Angio GJ, Rosenberg H, Sharples K, et al: Position of Wilms’ tumor: a study by the United Kingdom
paper: imaging methods for primary renal tumors of Children’s Cancer Study Group. J Clin Oncol 2002;
childhood: Cost versus benefits. Med Pediatr Oncol 1993; 20:2768–2773.
21:205–212. 62. Argani P, Perlman EJ, Breslow NE, et al: Clear cell
45. Ritchey ML, Kelalis PP, Breslow N, et al: Surgical sarcoma of the kidney: a review of 351 cases from the
complications following nephrectomy for Wilms’ tumor: a National Wilms’ Tumor Study Pathology Center.
report of National Wilms’ Tumor Study-3. Surg Gynecol Am J Surg Pathol 2000; 24:4–18.
Obstet 1992; 175:507–514. 63. Amar AM, Tomlinson G, Green DM, et al: Clinical
46. Weese DL, Applebaum H, Taber P: Mapping presentation of rhabdoid tumors of the kidney. J Pediatr
intravascular extension of Wilms’ tumor with magnetic Hematol Oncol 2001; 23:105–108.
resonance imaging. J Pediatr Surg 1991; 26:64–67. 64. Glass RBJ, Davidson AJ, Fernbach SK: Clear cell sarcoma
47. Laissy JP, Menegazzo D, Debray MP, et al: Renal of the kidney: CT, sonographic and pathologic
carcinoma: diagnosis of venous invasion with correlation. Radiology 1991; 180:715–717.
Gd-enhanced MR venography. Eur Radiol 2000; 65. White KS, Grossman H: Wilms’ and associated renal
10:1138–1143. tumors of childhood. Pediatr Radiol 1991; 21:81–88.
48. D’Angio GJ, Breslow N, Beckwith JB, et al: Treatment of 66. Broecker B: Renal cell carcinoma in children. Urology
Wilms’ tumor: results of the Third National Wilms’ 1991; 38:54–56.
Tumor Study. Cancer 1989; 64:349–360. 67. Raney RB Jr, Palmer N, Sutow WW, et al: Renal cell
49. Othersen HB Jr, DeLorimer A, Hrabovsky E, et al: carcinoma in children. Med Pediatr Oncol 1983;
Surgical evaluation of lymph node metastases in Wilms’ 11:91–98.
tumor. J Pediatr Surg 1990; 25:3:1–2. 68. Blute ML, Malek RS, Segura JW: Angiomyolipoma: a
50. Blute ML, Kelalis PP, Offord KP, et al: Bilateral Wilms’ clinical metamorphosis and concepts for management.
Tumor. J Urol 1987; 138:968–973. J Urol 1988; 139:20–24.
51. Ritchey ML, Kelalis PP, Haase GM, et al: Preoperative 69. Ewalt DH, Sheffield E, Sparagana SP, et al: Renal lesion
therapy for intracaval and atrial extension of Wilms’ growth in children with tuberous sclerosis complex.
tumor. Cancer 1993; 71:4104–4110. J Urol 1998; 160:141–145.
52. Montgomery BT, Kelalis PP, Blute ML, et al: Extended 70. Howell CJ, Othersen HB, Kiviat NE, et al: Therapy and
follow-up of bilateral Wilms’ tumor: results of the outcome in 51 children with mesoblastic nephroma: a
National Wilms’ Tumor Study. J Urol 1991; 146:514–518. report of the National Wilms’ Tumor Study. J Pediatr
53. Ritchey ML, Green DM, Breslow NE, Norkool P: Surg 1982; 17:826–830.
Accuracy of current imaging modalities in the diagnosis 71. Ritchey ML, Azizkhan RG, Beckwith JB: Neonatal
of synchronous bilateral Wilms’ tumor. Cancer 1995; Wilms’ tumors. J Pediatr Surg 1995; 30:856–859.
75:600–604. 72. Gormley TS, Skoog SJ, Jones RV, Maybee D: Cellular
54. Ng YY, Hall-Craggs MA, Dicks-Mireaux C, Pritchard congenital mesoblastic nephroma: What are the options?
J: Wilms’ tumour: Pre- and post-chemotherapy CT J Urol 1989; 142:479–483.
appearances. Clin Radiol 1991; 43:255–259. 73. Joshi VV, Kasznica J, Walters TR: Atypical mesoblastic
55. Thomas PRM, Shochat SJ, Norkool P, et al: Prognostic nephroma. Pathologic characterization of a potentially
implications of hepatic adhesion, invasion, and metastases aggressive variant of conventional congenital
at diagnosis of Wilms’ tumor. Cancer 1991; 68:2486–2488. mesoblastic nephroma. Arch Pathol Lab Med 1986;
56. Wilimas J, Douglass EC, Magill HL, Fitch S, Hutsu HO: 110:100–106.
Significance of pulmonary computed tomography at 74. Heidelberger KP, Ritchey ML, Dauser RC, McKeever
diagnosis in Wilms’ tumor. J Clin Oncol 1988; PE, Beckwith JB: Congenital mesoblastic nephroma
6:1144–1146. metastatic to the brain. Cancer 1993; 72:2499–2502.
770 Part VIII Pediatric Malignancies

75. Beckwith JB: Congenital mesoblastic nephroma. When 92. Rowe DH, Huang J, Kayton ML, et al: Anti-VEGF
should we worry? Arch Pathol Lab Med 1986; antibody suppresses primary tumor growth and
110:98–99. metastasis in an experimental model of Wilms’ tumor. J
76. Schmidt D, Beckwith JB: Histopathology of childhood Pediatr Surg 2000; 35:30–33.
renal tumors. Hematol Oncol Clin North Am 1995; 93. Huang J, Moore J, Soffer S, et al: Highly specific
9:1179–1200. antiangiogenic therapy is effective in suppressing growth
77. Beckwith JB, Zuppan CE, Browning NG, et al: of experimental Wilms’ tumors. J Pediatr Surg 2001;
Histological analysis of aggressiveness and responsiveness 36:357–361.
in Wilms’ tumor. Med Pediatr Oncol 1996; 27:422–428. 94. Green DM, Breslow N, Beckwith JB, et al:
78. Beckwith JB, Palmer NF: Histopathology and prognosis Comparison between single-dose and divided-dose
of Wilms’ tumor. Results from the National Wilms’ administration of dactinomycin and doxorubicin for
Tumor Study. Cancer 1978; 41:1937–1948. patients with Wilms’ tumor: a report from the
79. Breslow NB, Churchill G, Beckwith JB, et al: Prognosis National Wilms’ Tumor Study Group. J Clin Oncol
for Wilms’ tumor patients with nonmetastatic disease at 1998; 16: 237–245.
diagnosis – results of the Second National Wilms’ Tumor 95. Weeks DA, Beckwith JB, Luckey DW: Relapse-
Study. J Clin Oncol 1985; 3:521–531. associated variables in stage I, favorable histology Wilms’
80. Weirich A, Schmidt D, Harms D, et al: Distribution of tumor. Cancer 1987; 60:1204–1212.
subtypes in standard Wilms’ tumor after pre-operative 96. Shamberger RC, Guthrie KA, Ritchey ML, et al:
chemotherapy and its possible influence of the patients Surgery-related factors and local recurrence of Wilms’
cure rate. Med Pediatr Oncol 1994; 23:217. tumor in National Wilms’ Tumor Study 4. Ann Surg
81. Bonadio JF, Storer B, Norkool P, et al: Anaplastic Wilms’ 1999; 229:292–297.
tumor: clinical and pathological studies. J Clin Oncol 97. Breslow N, Churchill G, Nesmith B, et al:
1985; 3:513–520. Clinicopathologic features and prognosis for Wilms’
82. Faria P, Beckwith JB, Mishra K, et al: Focal versus diffuse tumor patients with metastases at diagnosis. Cancer
anaplasia in Wilms’ tumor—new definitions with 1986; 58:2501–2511.
prognostic significance: a report from the National Wilms’ 98. Ritchey ML, Lally KP, Haase GM, Shochat SJ, Kelalis
Tumor Study Group. Am J Surg Pathol 1996; 20: 909–920. PP: Superior mesenteric artery injury during
83. Zuppan CW, Beckwith JB, Weeks DA, Luckey DW, nephrectomy for Wilms’ Tumor. J Pedatr Surg 1992;
Pringle KC: The effect of preoperative therapy on the 27:612–615.
histologic features of Wilms’ tumor. An analysis of cases 99. Ritchey ML, Othersen HB Jr, deLorimier AA, et al:
from the Third National Wilms’ Tumor Study. Cancer Renal vein involvement with nephroblastoma: a report of
1991; 68:385–394. National Wilms’ Tumor Study 3. Eur Urol 1990;
84. Beckwith JB, Kiviat NB, Bonadio JF: Nephrogenic rests, 17:139–144.
nephroblastomatosis, and the pathogenesis of Wilms’ 100. Nakayama DK, deLorimier AA, O’Neill JA Jr, et al:
tumor. Pediatr Pathol 1990, 10:1–36. Intracardiac extension of Wilms’ tumor: a report of the
85. Beckwith JB: Nephrogenic rests and the pathogenesis of National Wilms’ Tumor Study. Ann Surg 1986;
Wilms’ tumor: developmental and clinical considerations. 204:693–697.
Am J Med Genet 1998; 79:268–273. 101. Shurin SB, Gauderer MWL, Dahms BB, Conrad WG:
86. Coppes MJ, Arnold M, Beckwith JB, et al: Factors Fatal intraoperative pulmonary embolization of Wilms’
affecting the risk of contralateral Wilms’ tumor tumor. J Pediatr 1982; 101:559–562.
development. Cancer 1999; 85:1616–1625. 102. Dykes EH, Marwaha RK, Dicks-Mireaux C, et al: Risks
87. Dome JS, Chung S, Bergemann T, et al: High telomerase and benefits of percutaneous biopsy and primary
reverse transcriptase (hTERT) messenger RNA level chemotherapy in advanced Wilms’ tumour. J Pediatr
correlates with tumor recurrence in patients with Surg 1991;26:610–612.
favorable histology Wilms’ tumor. Cancer Res 1999; 103. Borden TA: Wilms’ tumor and pulmonary embolism.
59:4301–4307. Soc Pediatr Urol Newsletter 1992:27–29.
88. Douglass EC, Look AT, Webber B, et al: Hyperdiploidy 104. Bracken RB, Sutow WW, Jaffe N, Ayala A, Guarda L:
and chromosomal rearrangements define the anaplastic Preoperative chemotherapy for Wilms’ tumor. Urology
variant of Wilms’ tumor. J Clin Oncol 1986; 4:975. 1982; 19:55–60.
89. Rainwater LM, Hosaka Y, Farrow GM, et al: Wilms’ 105. Ritchey ML, Pringle K, Breslow N, et al: Management
tumors: relationship of nuclear deoxyribonucleic acid and outcome of inoperable Wilms’ tumor. a report of
ploidy to patient survival. J Urol 1987; 138:974–977. National Wilms’ Tumor Study. Ann Surg 1994;
90. Layfield LJ, Ritchie AWS, Ehrlich R: The relationship 220:683–690.
of deoxyribonucleic acid content to conventional 106. Ritchey ML, Shamberger RC, Haase G, et al: Surgical
prognostic factors in Wilms’ tumor. J Urol 1989; complications after nephrectomy for Wilms’ tumor:
142:1040–1043. report from the National Wilms’ Tumor Study Group.
91. Kayton ML, Rowe DH, O’Toole KM, et al: Metastasis Pediatrics 1999; 104:816–817.
correlates with production of vascular endothelial growth 107. Ritchey ML, Kelalis P, Breslow N, Etzioni R, Haase G:
factor in a murine model of human Wilms’ tumor. Small bowel obstruction following nephrectomy for
J Pediatr Surg 1999; 34:743–748. Wilms’ tumor. Ann Surg 1993; 218:654–659.
Chapter 47 Wilms’ Tumor 771

108. Godzinski J, Tournade MF, deKraker J, et al: Rarity of Wilms’ tumor trial and study: a risk-adapted therapeutic
surgical complications after postchemotherapy approach in Wilms’ tumor. J Clin Oncol 1993;
nephrectomy for nephroblastoma. Experience of the 11:1014–1023.
International Society of Paediatric Oncology—Trial and 122. deKraker J, Weitzman S, Voûte PA: Preoperative
study “SIOP-9”. International Society of Paediatric strategies in the management of Wilms’ tumor. Hematol
Oncology Nephroblastoma Trial and Study committee. Oncol Clin North Am 1995; 9:1275–1286.
Eur J Pedatr Surg 1998; 8:83–86. 123. Tournade MF, Com-Nogoue, deKraker J, et al: Optimal
109. D’Angio GJ, Evans AE, Breslow N, et al: The treatment duration of preoperative chemotherapy in unilateral and
of Wilms’ tumor: results of the National Wilms’ Tumor nonmetastatic Wilms’ tumor in children over 6 months
Study. Cancer 1976; 38:633–646. of age: Result of the 9th SIOP Wilms’ tumor trial and
110. D’Angio GJ, Evans A, Breslow N, et al: The treatment study. J Clin Oncol 2001; 19:488–500.
of Wilms’ tumor: results of the Second National Wilms’ 124. Graf N, Tournade MF, deKraker J: The role of
Tumor Study, Cancer 1981; 47:2302–2311. preoperative chemotherapy in the management of
111. Farewell VT, D’Angio GJ, Breslow N, Norkool P: Wilms’ tumor. Urol Clin North Am 2000; 27:443–454.
Retrospective validation of a new staging system for 125. Boccon-Gibod L, Rey A, Sandstedt B, et al: Complete
Wilms’ tumor. Cancer Clin Trials 1981; 4:167–171. necrosis induced by preoperative chemotherapy in
112. Green DM, Breslow N, Evans I, et al: The treatment of Wilms’ tumor as an indicator of low risk: Report of the
children with stage IV Wilms’ tumor. A report from the International Society of Pediatric Oncology (SIOP)
National Wilms’ Tumor Study Group. Med Pediatr Nephroblastoma Trial and Study 9. Med Pediatr Oncol
Oncol 1996; 26:147–152. 2000; 34:183–190.
113. Green DM, Breslow NE, Evan I, et al: Effect of dose 126. Vujanic GM, Sandstedt B, Harms D, et al: Revised
intensity of chemotherapy on the hematological toxicity International Society of Pediatric Oncology (SIOP)
of the treatment of Wilms’ tumor. A report from the working classification of renal tumors of childhood. Med
National Wilms’ Tumor Study. Am J Pediatr Hematol Pediatr Oncol 2002; 38:79–82.
Oncol 1994; 16:207–212. 127. Coppes MJ, deKraker J, Van Dijken PJ, et al: Bilateral
114. Green DM, Breslow NE, Beckwith JBB, et al: Effect of Wilms’ tumor: Long-term survival and some
duration of treatment on treatment outcomes and cost of epidemiological features. J Clin Oncol 1989;
treatment for Wilms’ tumor: a report from the National 7:310–315.
Wilms’ Tumor Study Group. J Clin Oncol 1998; 128. Kumar R, Fitzgerald R, Breatnach F: Conservative
16:3744–3751. surgical management of bilateral Wilms’ tumor: results
115. Green DM, Beckwith JB, Breslow NB, et al: The of the United Kingdom Children’s Cancer Study Group.
treatment of children with Stage II-IV anaplastic Wilms’ J Urol 1998; 160:1450–1453.
tumor. A report from the National Wilms’ Tumor Study. 129. Ritchey ML, Green DM, Thomas P, et al: Renal failure
J Clin Oncol 1994; 12:2126–2131. in Wilms’ tumor patients: a report of the NWTSG. Med
116. Green DM, Breslow NE, Beckwith JB, Ritchey ML, Pediatr Oncol 1996; 26:75–80.
et al: Treatment with nephrectomy only for small, stage 130. Weirich A, Leuschner I, Harms D, et al: Clinical impact
I/favorable histology Wilms’ tumor. A Report From of histologic subtypes in localized non-anaplastic
The National Wilms’ Tumor Study Group. J Clin Oncol nephroblastoma treated according to the trial and study
2001; 19:3719–3724. SIOP-9/GPOH. Ann Oncol 2001; 12:311–319.
117. Larsen E, Perez-Atayde A, Green DM, et al: Surgery 131. Maes P, Delemarrre J, deKraker J, Ninane J: Fetal
only for the treatment of patients with stage I (Cassady) rhabdomyomatous nephroblastoma: a tumour of good
Wilms’ tumor. Cancer 1990; 66:264–266. prognosis but resistant to chemotherapy. Eur J Cancer
118. Green DM, Beckwith JB, Weeks DA, et al: The 1999; 35:1356–1360.
relationship between microsubstaging variables, tumor 132. Anderson J, Slater O, McHugh K, et al: Response
weight and age at diagnosis of children with stage without shrinkage in bilateral tumor: significance of
I/favorable histology Wilms’ tumor. A report from the rhabdomyomatous histology. J Pediatr Hematol Oncol
National Wilms’ Tumor Study. Cancer 1994; 2002; 24:31–34.
74:1817–1820. 133. Penn I: Renal transplantation for Wilms’ tumor: report
119. Lemerle J, Voûte PA, Tournade MF, et al: Preoperative of 20 cases. J Urol 1979; 122:793–794.
versus postoperative radiotherapy, single versus multiple 134. Grundy P, Breslow N, Green DM, et al: Prognostic
courses of actinomycin D, in the treatment of Wilms’ factors for children with recurrent Wilms’ tumor: results
tumor: preliminary results of a controlled clinical trial from the second and third National Wilms’ Tumor
conducted by the International Society of Pediatric Study. J Clin Oncol 1989; 7:638–647.
Oncology (SIOP). Cancer 1976; 38:647–654. 135. Breslow NE, Takashima JR, Whitton JA, et al: Second
120. Lemerle J, Voûte PA, Tournade MF, et al: Effectiveness malignant neoplasms following treatment for Wilms’
of preoperative chemotherapy in Wilms’ tumor: results tumor: a report from the National Wilms’ Tumor Study
of an International Society of Pediatric Oncology (SIOP) Group. J Clin Oncol 1995; 13:1851–1859.
clinical trial. J Clin Oncol 1983; 1:604–609. 136. Gilladoga AC, Manuel C, Tan CT, et al: The
121. Tournade MF, Com-Nougue C, Voûte PA, et al: Results cardiotoxicity of adriamycin and daunomycin in children.
of the Sixth International Society of Pediatric Oncology Cancer 1976; 37:1070–1078.
772 Part VIII Pediatric Malignancies

137. Green DM, Grigoriev YA, Nan B, et al: Congestive 147. Argueso LR, Ritchey ML, Boyle ET Jr, et al: Prognosis
failure after treatment for Wilms’ tumor. A report from of the solitary kidney after unilateral nephrectomy in
the National Wilms’ Tumor Study Group. J Clin Oncol childhood. J Urol 1992; 148:747–751.
2001; 19:1926–1934. 148. Robitaille P, Mongeau JG, Lortie L, Sinnassamy P:
138. Evans AE, Norkool P, Evans I, Breslow N, D’Angio Long-term follow-up of patients who underwent
GJ: Late effects of treatment for Wilms’ tumor: a report nephrectomy in childhood. Lancet 1985; 1:1297–1299.
from the National Wilms’ Tumor Study Group. Cancer 149. Barrera M, Roy LP, Stevens M: Long-term follow-up
1991; 67:331–336. after unilateral nephrectomy and radiotherapy for
139. Hogeboom CJ, Grosser SC, Guthrie KA, Thomas PR, Wilms’ tumor. Pediatr Nephrol 1989; 3:430–432.
D’Angio GJ, Breslow NE: Stature loss following 150. Bhisitkul DM, Morgan ER, Vozar MA, Langman CB:
treatment for Wilms’ tumor. Med Pediatr Oncol 2001; Renal functional reserve in long-term survivors of
36:295–304. unilateral Wilms’ tumor. J Pediatr 1990; 118:698–702.
140. Stillman RJ, Schinfeld JS, Schiff I, et al: Ovarian failure 151. Bailey S, Roberts A, Brock C, et al: Nephrotoxicity in
in long term survivors of childhood malignancy. survivors of Wilms’ tumour in the North of England.
Am J Obstet Gynecol 1987; 139:62–66. Br J Cancer 2002; 87:1092–1098.
141. Li FP, Gimbrere K, Gelber RD, et al: Outcome of 152. Levitt GA, Yeomans E, Dicks Mireaux C, et al: Renal
pregnancy in survivors of Wilms’ tumor. JAMA 1987; size and function after cure of Wilms’ tumor. Br J
257:216–219. Cancer 1992; 66:877–882.
142. Green DM, Peabody EM, Nan B, et al: Pregnancy 153. Donckerwolcke RM, Coppes MJ: Adaptation of renal
outcome after treatment for Wilms’ tumor: a report function after unilateral nephrectomy in children with
from the National Wilms’ Tumor Study Group. J Clin renal tumors. Pediatr Nephrol 2001; 16:568–574.
Oncol 2002; 20:2506–2513. 154. Mitus A, Tefft M, Feller FX: Long-term follow-up
143. Kinsella TJ, Trivette G, Rowland J, et al: Long-term of renal function of 108 children who underwent
follow-up of testicular function following radiation for nephrectomy for malignant disease. Pediatrics 1969;
early-stage Hodgkin’s disease. J Clin Oncol 1989; 44:912–921.
7:718–724. 155. McLorie GA, McKenna PH, Greenberg M, et al:
144. Scully RE, Mark J, McNeely BU: Case records of the Reduction in tumor burden allowing partial
Massachusetts General Hospital. NEJM 1985; nephrectomy following preoperative chemotherapy in
312:1111–119. biopsy proved Wilms’ tumor. J Urol 1991; 146:509–513.
145. Welch TR, McAdams AJ: Focal glomerulosclerosis 156. Moorman-Voestermans C, Aronson D, Staalman CR,
as a late sequela of Wilms’ tumor. J Pediatr 1986; et al: Is partial nephrectomy appropriate treatment for
108:105–109. unilateral Wilms’ tumor? J Pediatr Surg 1998;
146. Anderson S, Meyer TW, Brenner BM: The role of 33:165–170.
hemodynamic factors in the initiation and progression 157. Breslow NE, Takashima JR, Ritchey ML, et al: Renal
of renal disease. J Urol 1985; 363–368. failure in the Denys-Drash and Wilms’ tumor-aniridia
syndromes. Cancer Res 2000; 60:4030–4032.
C H A P T E R

48 Rhabdomyosarcoma of the Pelvis


and Paratesticular Structures
Hsi-Yang Wu, MD, and Howard M. Snyder III, MD

One of the goals in pediatric oncology is to individualize phamide (VAC). Failure-free survival at 3 years is now
therapy for the low-risk patient versus the high-risk 77%.1 During the same time, the surgical approach
patient. Therefore, the least amount of chemotherapy has changed from exenterative surgery to organ-
and radiation for the low-risk patient, which will achieve preserving surgery following chemotherapy. The func-
a cure, is desirable, since it decreases long-term morbid- tional bladder salvage rate has risen from 25% to 60%
ity. For the high-risk patient, molecular identification of with this change in management. IRS-V was designed to
poor prognostic factors can suggest that intensified evaluate new agents, such as topotecan for advanced dis-
treatment is necessary. The management of rhab- ease, while attempting to decrease cyclophosphamide
domyosarcoma (RMS) remains controversial, because and XRT dosing for low-risk patients. Unlike neurob-
while a combination of surgery, chemotherapy, and radi- lastoma, there are no plans for high-dose chemotherapy
ation therapy is more successful than any single modal- followed by bone marrow transplantation in advanced
ity approach, the optimal timing and dose of each disease since there are no data showing that this has been
remains unclear. This is evident when reviewing the dif- helpful.2,3
ferent approaches to the disease taken by North
American and European groups. To maintain an overall
PATHOLOGY AND MOLECULAR BIOLOGY
perspective in reviewing the literature on RMS, it is use-
ful to remember three key points: RMS consists of small, blue, round cells (similar to neu-
roblastoma, Ewing’s sarcoma, non-Hodgkin’s lymphoma,
1. Chemotherapy cures microscopic disease. and leukemia) with a microscopic appearance of spindle
2. Residual mass does not equal disease. cells resembling fetal skeletal muscle. The cells derive
3. Radiation therapy renders pathology very difficult to from undifferentiated mesoderm and, therefore, can be
read. present in tissues, which do not normally express skeletal
muscle. One surgical problem is that although there may
Twenty percent of RMS involves the bladder, prostate, appear to be a pseudocapsule, the tumor will often infil-
vagina, or paratesticular area. The incidence peaks trate through the tissue. Multiple biopsies are required to
between ages 2 to 4 and 15 to 19. The tumor is nonen- ensure that the excision is complete. Embryonal pathol-
capsulated, grows rapidly, and spreads to regional lymph ogy, which is a favorable factor, accounts for 90% of gen-
nodes, as well as hematogenously. In the United States, itourinary RMS. Sarcoma botryoides (bunch of grapes) is
patients are managed by Intergroup Rhabdomyosarcoma a polypoid form of embryonal pathology, which occurs in
Study Group (IRSG) protocols. (The IRSG is now the hollow organs like the bladder. Alveolar pathology, which
Children’s Oncology Group Soft Tissue Sarcoma tends to occur in extremities, is less favorable. Anaplastic
Committee.) These studies have been ongoing since and undifferentiated pathologies rarely occur in geni-
1972 to achieve better survival with less morbidity. tourinary tumors (Figure 48-1). Desmin and actin stains
Three-year patient survival, which was only 40% to are sometimes useful in determining the diagnosis.
73% prior to chemotherapy, has improved to 86% in Current lab efforts have found genetic translocations
IRS-IV with vincristine, dactinomycin, and cyclophos- between chromosomes 1;13 (favorable) and 2;13 (very

773
774 Part VIII Pediatric Malignancies

Figure 48-1 Histologic subtypes of RMS: A, embryonal; B, spindle cell; C, botryoid; D,


alveolar. (H/E 33X.) (Courtesy Dr. Philip Faught.)

high risk) to be prognostic markers for survival in predominance. Sometimes, a child can void a piece of the
patients with alveolar tumors with metastatic disease.4 tumor per urethra. In females, the sarcoma botryoides
Unfortunately, there are no good molecular markers for type of tumor can prolapse through the urethra.
embryonal tumor behavior that can be followed at the Occasionally, the tumor can project posteriorly and cause
present time.3 constipation. Determining which organ the tumor arose
from can be difficult, but initial treatment does not
depend on this distinction. Computed tomography (CT)
PRESENTATION AND EVALUATION
or magnetic resonance imaging (MRI) (T2 weighted) of
Bladder and prostate primaries tend to present with uri- the pelvis is the initial study. MRI may give better reso-
nary retention, a palpable lower abdominal mass, gross lution of tumor invasion.
hematuria, and tend to be located at the trigone and blad- Vaginal primaries present with vaginal bleeding or an
der neck (Figure 48-2). The tumors have a 2.5/1 male introital mass and tend to occur on the anterior vaginal

Figure 48-2 Sarcoma botryoides of bladder base showing characteristic polypoid


appearance on cystogram.
Chapter 48 Rhabdomyosarcoma of the Pelvis and Paratesticular Structures 775

wall. CT or MRI (T2 weighted) of the pelvis is the initial Table 48-1 Preoperative Staging
study. Uterine primaries were formerly considered to be a
T1 Confined to organ of origin, a: ≤5 cm in size,
disease of older females, but more recent studies suggest b: >5 cm in size
that they present at the same age as vaginal primaries and
can be successfully treated with limited excision followed T2 Extension or fixed to surrounding tissue, a:
by chemotherapy. Hysterectomy is rarely carried out.5 ≤5 cm in size, b: >5 cm in size
Vulvar and cervical lesions tend to occur in teenagers.6
N0 Regional nodes clinically negative
Staging for bladder, prostate, and vaginal primaries is
completed by endoscopic examination and biopsy of the N1 Regional nodes clinically positive
lesion. Cold-cup biopsy will give better results than using
a resectoscope loop, since cautery artifact can make inter- Nx Unknown
pretation difficult. If the lesion does not appear to involve
M0 No distant metastasis
the mucosa of the bladder, prostate, or vagina, then a
needle biopsy may be necessary via a percutaneous M1 Metastasis present
(transperineal), transrectal, or laparoscopic approach.
Paratesticular RMS has two peak incidences, in the Stage I Vaginal and paratesticular RMS, any
3- to 4-month-old boy and in the teenager. The bio- T, any N, M0
logic activity of the tumor is different between the
Stage II Bladder/prostate RMS, T1a or T2a, N0
neonate and the teenager (90% versus 63% failure-free or Nx, M0
survival at 3 years).1 The primary tumor presents as a
painless scrotal mass. Scrotal ultrasound, serum β- Stage III Bladder/prostate RMS (T1a or T2a) and N1,
HCG, and AFP should be obtained to confirm that the M0, OR (T1b or T2b), any N, M0
mass is not a testicular primary. Imaging of the
retroperitoneum by CT or MRI is usually carried out Stage IV Any tumor with M1
before surgery. Thirty percent to forty percent will have
microscopic metastases to the retroperitoneum. The
tumor is resected, along with the testis in a radical
Table 48-2 Postoperative Grouping
inguinal orchiectomy. Pathologic staging of retroperi-
toneal lymph nodes is currently recommended for Group 1: Localized disease, completely excised, no microscopic
residual
patients 10 years of age or older, regardless of radiologic
findings, and in patients <10 years of age with retroperi- A Confined to site of origin, completely resected
toneal disease visible on CT or MRI.3
Metastatic workup for all lesions is completed with a B Infiltrating beyond site of origin, completely
chest x-ray (and possibly chest CT), liver function tests, resected
bone scan and bone marrow biopsy.
Group 2: Total gross resection

STAGING A Gross resection with microscopic local residual

The IRS study includes both preoperative staging and B Regional disease with involved lymph nodes,
postoperative grouping (Tables 48-1 and 48-2). The IRS- completely resected with no microscopic residual
I to IRS-III studies grouped patients based on complete-
C Microscopic local and/or nodal residual
ness of resection, introducing biases (shifting patients
from group 1 to group 3), which are not seen with the use Group 3: Incomplete resection or biopsy with gross residual
of the TNM system in IRS-IV and IRS-V. Eighty per-
cent of bladder and prostate primaries are group 3 (gross Group 4: Distant metastases
residual), whereas 50% of nonbladder/prostate primaries
are group 1 (complete resection), with about 20% in
group 2 (complete gross resection, with microscopic or Since cyclophosphamide is cheaper and has less toxicity,
nodal tumor) and group 3, respectively.7 VAC is the current standard chemotherapy. The addition
of doxorubicin or melphalan to VAC has not been bene-
ficial. In IRS-V, low-risk patients are randomized to
GENERAL APPROACH
receive either VA or VAC, plus or minus XRT.
Chemotherapy/Radiation
Intermediate-risk patients receive chemotherapy and
In IRS-IV, VAC was shown to be as effective as two other XRT and are randomized to VAC or VAC alternating
three drug regimens (VIE/VAE: ifosfamide, etoposide).1 with vincristine, topotecan, and cyclophosphamide.
776 Part VIII Pediatric Malignancies

TREATMENT AND OUTCOME


High-risk patients receive CPT-11 (irinotecan), VAC,
Bladder and Prostate
and XRT.3
Chemotherapy and Radiation
In IRS-IV, hyperfractionated radiation therapy did not
achieve better results than conventional radiation ther- IRS-III included intensified chemotherapy (dactino-
apy.8 For IRS-V, patients with group 1 embryonal pathol- mycin, VP-16) and 6 weeks of XRT, increasing the func-
ogy will not receive XRT. For other patients, the dose of tional bladder salvage rate from 25% to 60%. For
radiation to the residual primary will be higher than to persistent disease, group 2 was treated with 41 Gy and
microscopic residual disease, and metastases will be irra- group 3 was treated with 50 Gy. At 24 weeks, a third
diated as well.3 Surgery as treatment of gross residual dis- operative evaluation was performed with consideration
ease after chemotherapy is still an option. At the for exenteration.14
Children’s Hospital of Philadelphia, we have favored this The difficulty with radiation therapy is that since the
approach, mainly because of the difficulty of interpreting tumors tend to be located at the bladder neck, even the
the pathology after radiation. lowest dose (41 Gy) that the radiation oncologists are
willing to deliver may significantly risk urinary conti-
nence. Current attempts at limiting radiation toxicity to
Surgery
adjacent organs involve both conformal radiation therapy
The goal of the initial procedure is to obtain adequate and brachytherapy. The long-term risk of radiation vas-
tissue for a definitive diagnosis. If possible, one should culitis, which is inevitably progressive, as well as possible
remove the tumor without removing the affected bony pelvis deformity in these children, is another issue
organ, except in the case of paratesticular tumors. If to consider. Therefore, one must sometimes weigh
excision is not possible, chemotherapy will be given whether preserving a bladder without an outlet is better
with a plan for a second look operation. In follow-up than removing the bladder entirely.15,16
staging, a biopsy is needed during the second look to
confirm whether a residual mass is tumor. The tumor
Surgery for Bladder Primaries
can involute more rapidly than the supporting stroma,
so gross residual masses may not contain any viable While a full retroperitoneal lymphadenectomy is not
tumor. necessary, any suspicious lymph nodes along the great
The goal of the definitive operation is a good, radi- vessels between the obturator fossa and the renal veins
cal, but pelvic organ-sparing operation. If microscopic should be removed. Multiple frozen-section biopsies of
residual disease is found, brachytherapy or external the bladder are obtained around the tumor. If these are
beam radiation therapy can be used. Exenteration is negative, and the tumor is amenable to partial cystec-
reserved for patients who have failed previous thera- tomy with a 2- to 3-cm margin, then the bladder does not
pies. In Europe, an alternate approach is to give pri- need to be entirely removed. If the tumor extends down
mary chemotherapy without any attempt at initial local the urethra, then the symphysis should be split to gain
control (radiation therapy or surgery), and to offer better access. After completing distal dissection of the
local therapy based on the initial chemotherapy urethra, the symphysis is closed with absorbable suture.
response.9,10 With this improved exposure, it is also possible to per-
Review of the pathology may reveal persistent rhab- form a nerve-sparing dissection. The placement of
domyoblasts in a patient who has received as much brachytherapy catheters for afterloading (to treat micro-
chemotherapy as can safely be given. Currently, there scopic positive margins if needed) should be considered.
is debate concerning the malignant potential of these For patients who require total cystectomy, we have
cells, which represent matured rhabdomyoblasts.11,12 often placed Dexon mesh across the abdomen to hold the
Normally, resection of the involved organ is carried out. intestines out of the pelvis at the level of the sacral
However, if this would require destruction of a functional promontory. This is done by attaching it to the sacral
bladder, observation with frequent radiologic follow-up promontory and wrapping it around the sigmoid. This
may be an option to consider. limits the exposure of the bowel to the radiation field if
postoperative radiation is required. The final surgical
decision is whether to proceed with continent urinary
Relapse
reconstruction at the same time. We believe that it is not
Five-year survival after relapse is 64% for botryoid wise to perform the reconstruction at the same time,
embryonal, 26% for other embryonal, and 5% for alveo- unless the patient is both motivated and able to perform
lar or undifferentiated pathology. Survival after relapse clean intermittent catheterization to drain a urinary
was also improved in lower-stage embryonal lesions at reservoir. For the younger patients who are not ready to
the time of diagnosis: 52% stage I, 20% stages II and III, manage a urinary reservoir, we have either brought up
and 12% stage IV.13 the remaining bladder plate with ureterovesical junctions
Chapter 48 Rhabdomyosarcoma of the Pelvis and Paratesticular Structures 777

intact as a vesicostomy, or performed low end-cutaneous patients were treated with surgery and chemotherapy, 19%
ureterostomies, with the ureters placed side-by-side as a required additional radiation therapy, 21% were treated
single stoma on the abdominal wall. with biopsy and chemotherapy alone, and 12% were man-
aged with biopsy, chemotherapy, and radiation therapy.
(There were insufficient data regarding local therapy for
Surgery for Prostate Primaries
the remaining patients.) Five-year patient survival and fail-
The technique is similar to that for localized prostatic ure-free survival were 82% and 69%.6 In IRS-IV, only 19%
adenocarcinoma. The use of nerve-sparing procedures of patients required wide excision of vaginal tumors.18 Age
may preserve erectile potency in these patients. Splitting between 1 and 9 was a favorable factor for survival (94%
the symphysis is useful as it is essential to remove the ure- versus 76%).6 The experience of the International Society
thra to the mid-bulbar level. The placement of of Pediatric Oncology (SIOP) in Europe has been similar,
brachytherapy catheters for afterloading (to treat micro- although they prefer intracavitary brachytherapy for local
scopic positive margins if needed) should be considered. control rather than hysterectomy.19

Outcome Paratestis
Chemotherapy and Radiation
In IRS-I and IRS-II, half of the patients survived with
intact bladders, whereas the other half had either total or In IRS-I to IRS-III, all patients underwent retroperi-
partial cystectomy. In those patients with intact bladders, toneal lymph node dissection (RPLND).1 Based on the
function (as assessed by questionnaire) was normal in European experience that 17 of 19 patients with clinical
73%, whereas 8% had incontinence and 9% had fre- stage I disease were cured with adjuvant chemotherapy
quency and nocturia. Overall renal function as assessed alone20; RPLND was not recommended in IRS-IV for
by serum BUN and creatinine was normal in 95%, stage I disease with a normal CT of the abdomen and
although 29% of the patients had abnormal renal scans, pelvis. This led to a significant understaging of disease
which was often hydronephrosis in the setting of a uri- based on radiologic evaluation alone and a decrease in
nary diversion.15 In IRS-I to IRS-III, over 25% of failure-free survival because patients with negative CT
patients treated with partial cystectomy had complica- scan did not receive radiation. Thirty percent of those
tions of gross hematuria, bladder contracture, or inconti- patients who were clinically stage I and over 10 years of
nence. The risk of complications increased as the XRT age required retreatment. Since the outcome for patients
dose raised over 40 Gy.16 <10 years old with stage I disease was good in IRS-IV,
More recent outcomes show that 50% of patients were those patients who are <10 years old have stage I disease,
managed with biopsy, 37% had partial cystectomy, and and negative abdominal/pelvic CT are treated with VA
13% had prostatectomy. Patients with embryonal pathol- (vincristine, dactinomycin) only in IRS-V. All patients
ogy had an 83% 3-year failure-free survival, compared to with stage I disease who are 10 years or older undergo
40% in those with alveolar pathology. However, all RPLND regardless of CT findings. Group 2 tumors
patients with group 4 (metastatic) prostate disease died.14 (positive lymph nodes on pathology) are treated with
radiation therapy and VAC.21
Vagina/Uterus
Chemotherapy and Radiation Surgery
Primary treatment with VAC chemotherapy is usually During inguinal orchiectomy, frozen section of the prox-
successful, and a biopsy 8 to 12 weeks after chemother- imal cord should reveal no tumor. If one makes a scrotal
apy is recommended. Pelvic lymph node dissection is not incision for what turns out to be a solid paratesticular
necessary. Vaginectomy or hysterectomy should only be mass, chemotherapy will often cure residual disease.
considered for persistent disease after a complete course However, some cases have required hemiscrotectomy
of chemotherapy.17 Rhabdomyoblasts on biopsy are evi- due to tumor infiltration.
dence of chemotherapy response, and therefore further The technique for RPLND is identical to that used
chemotherapy, instead of resection, is the proper treat- for retroperitoneal involvement by testicular tumors.
ment. Radiation therapy should only be used for persist- Ipsilateral template and nerve-sparing techniques can be
ent disease or relapse.17,18 used to maintain ejaculation.22

Outcome Outcome
Surgery and chemotherapy cure most cases of vaginal Three-year patient survival and failure-free survival are
RMS. In the overall IRS-I to IRS-IV experience, 42% of 92% and 81%, respectively, for group 1 tumors overall.
778 Part VIII Pediatric Malignancies

The failure-free survival was worse than the 95% rationale for Intergroup Rhabdomyosarcoma Study
achieved in IRS-III, although the patient survival rate is V. J Ped Hem/Oncol 2001; 23:215–220.
the essentially the same as the 96% achieved in IRS-III. 4. Sorenson PH, Lynch JC, Qualman SJ, et al: PAX3-FKHR
Again, this was felt to be due to understaging when the and PAX7-FKHR gene fusions are prognostic indicators
in alveolar rhabdomyosarcoma: a report from the
retroperitoneal lymph nodes were not pathologically
Children’s Oncology Group. J Clin Oncol 2002;
evaluated. Those patients more than 10 years old only
20:2672–2679.
had a 63% survival, leading to the recommendation that 5. Corpron C, Andrassy RJ, Hays CM, et al: Conservative
all patients of that age undergo RPPLND.1 management of uterine rhabdomyosarcoma: a report from
the Intergroup Rhabdomyosarcoma Studies III and IV
Pilot. J Pediatr Surg 1995; 30:942–944.
COMPLICATIONS
6. Arndt CAS, Donaldson SS, Anderson JR, et al: What
The majority of patients have acute toxicity from the constitutes optimal therapy for patients with
chemotherapy; 90% developed myelosuppression, 55% rhabdomyosarcoma of the female genital tract? Cancer
developed significant infections, and renal toxicity was 2001; 91:2454–2468.
seen in 2%.1 Most relapses occur within 3 years of initial 7. Barksdale EM, Weiner ES: Rhabdomyosarcoma.
In Pediatric Surgery. Philadelphia, WB Saunders,
diagnosis.13 Late recurrences can occur in patients who
2000.
are treated with chemotherapy alone. Out of 883
8. Donaldson SS, Neza J, Breneman JC, et al: Results from
patients, 10 developed a secondary cancer. Patients with the IRS-IV randomized trial of hyperfractionated
preexisting renal abnormalities were at a higher risk of radiotherapy in children with rhabdomyosarcoma – a
death (5% versus 1%).1 The experience from IRS-I and report from the IRSG. Int J Radiat Oncol Biol Phys 2001;
IRS-II suggests a 27% long-term urinary complica- 51:718–728.
tion rate, with incontinence being the major issue. 9. Flamant F, Rodary C, Rey A, et al: Treatment of non-
Twenty-nine percent required sex hormone replacement metastatic rhabdomyosarcoma in childhood and
and 11% were shorter than expected.15 If radiation ther- adolescence. Results of the second study of the
apy has been used, there is an increased risk of a second- International Society of Pediatric Oncology: MMT84.
ary neoplasm, often another sarcoma, in the radiation Eur J Cancer 1998; 34:1050–1062.
10. Koscielniak E, Harms D, Henze G, et al: Results of
field. Complications specific to surgical treatment
treatment for soft tissue sarcoma in childhood and
include intestinal obstruction, anejaculation, and lower
adolescence: A final report of the German cooperative soft
extremity edema after RPLND. However, this series tissue sarcoma study CWS-86. J Clin Oncol 1999;
evaluated therapy from 1972 to 1984, and current tech- 17:3706–3719.
niques may result in better outcomes.23 11. Ortega JA, Rowland J, Monforte H, et al: Presence of
well-differentiated rhabdomyoblasts at the end of therapy
for pelvic rhabdomyosarcoma: implications for the
SUMMARY
outcome. J Pediatr Hem/Oncol 2000; 22:106–111.
RMS is treated with a combination of surgery, chemother- 12. Leuschner I, Harms D, Mattke A, et al:
apy, and radiation. Bladder, prostate, and vaginal primar- Rhabdomyosarcoma of the urinary bladder and vagina.
ies are treated with biopsy followed by chemotherapy, Am J Surg Pathol 2001; 25:856–864.
13. Pappo AS, Anderson JR, Crist WM, et al: Survival after
reresection, and radiation with periodic reevaluations.
relapse in children and adolescents with
Paratesticular primaries receive chemotherapy after
rhabdomyosarcoma: a report from the Intergroup
orchiectomy and possibly require a RPLND if spread to Rhabdomyosarcoma Study Group. J Clin Oncol 1999;
the retroperitoneum is seen or if the boy is >10 years of 17:3487–3493.
age. 14. Paidas CN: Results of rhabdomyosarcoma of the bladder
and prostate: Is bladder preservation successful? In
American Academy of Pediatrics Meeting, Chicago, IL,
REFERENCES Section on Surgery, 2000.
15. Raney B, Heyn R, Hays DM, et al: Sequelae of treatment
1. Crist WM, Anderson JR, Meza JL, et al: Intergroup in 109 patients followed 5 to 15 years after diagnosis of
Rhabdomyosarcoma Study-IV: Results for patients with sarcoma of the bladder and prostate. Cancer 1993;
nonmetastatic disease. J Clin Oncol 2001; 19:3091–3102. 71:2387–2394.
2. Carli M, Colombatti R, Oberlin O, et al: High-dose 16. Hays DM, Raney RB, Wharam MD, et al: Children with
melphalan with autologous stem cell rescue in metastatic vesical rhabdomyosarcoma (RMS) treated by partial
rhabdomyosarcoma. J Clin Oncol 1999; 17:2796–2803. cystectomy with neoadjuvant or adjuvant chemotherapy,
3. Raney RB, Anderson JR, Barr FG, et al: with or without radiotherapy. J Pediatr Hem/Oncol 1995;
Rhabdomyosarcoma and undifferentiated sarcoma in the 17:46–52.
first two decades of life: a selective review of Intergroup 17. Andrassy RJ, Weiner ES, Raney RB, et al: Progress in the
Rhabdomyosarcoma Study Group experience and surgical management of vaginal rhabdomyosarcoma: a
Chapter 48 Rhabdomyosarcoma of the Pelvis and Paratesticular Structures 779

25-year review from the Intergroup Rhabdomyosarcoma 21. Weiner ES, Anderson JR, Ojimba JI, et al: Controversies
Study Group. J Pediatr Surg 1999; 34:731–735. in the management of paratesticular rhabdomyosarcoma:
18. Andrassy RJ: Modern approach to rhabdomyosarcoma of Is staging retroperitoneal lymph node dissection necessary
the vagina and uterus. In American Academy of Pediatrics for adolescents with resected paratesticular
Meeting, Chicago, IL, Section on Surgery, 2000. rhabdomyosarcoma? Semin Pediatr Surg 2001;
19. Martelli H, Oberlin O, Rey A, et al: Conservative 10:146–152.
treatment for girls with nonmetastatic rhabdomyosarcoma 22. Donohue JP, Foster RS, Rowland RG, et al: Nerve-
of the genital tract: a report from the study committee of sparing retroperitoneal lymphadenectomy with
the International Society of Pediatric Oncology. J Clin preservation of ejaculation. J Urol 1990; 144:287–291.
Oncol 1999; 17:2117–2122. 23. Heyn R, Raney B, Hays D, et al: Late effects of therapy in
20. Olive D, Flamant F, Zucker JM, et al: Paraaortic patients with paratesticular rhabdomyosarcoma.
lymphadenectomy is not necessary in the treatment of Intergroup Rhabdomyosarcoma Study Committee. J Clin
localized paratesticular rhabdomyosarcoma. Cancer 1984; Oncol 1992; 10:614–623.
54:1283–1287.
C H A P T E R

49 Prepubertal Testicular Tumors


Peter D. Metcalfe, MD,
and Darius J. Bägli, MD, CM, FRCSC, FAAP

Pediatric testicular tumors differ significantly from their between benign from malignant masses is not always
adult counterparts in several fundamental aspects. They demonstrable sonographically.11–14 Color doppler can
are less common, have different histologic distribution, demonstrate increased vascularity, increasing the speci-
and they are more likely to be benign. There is also a ficity of a malignant process.12 Teratoma may be sus-
much higher incidence of metastases to the pediatric tes- pected by a characteristic solid and cystic appearance,15,16
ticle, primarily of lymphopoietic origin. and epidermoid cysts may have an onionskin or target
The reported incidence of pediatric testicular tumors appearance17 on ultrasound.
is between 0.5 and 2 per 100,000 children, and they
account for 1% of solid tumors in this age group.1–3
HISTOLOGIC TUMOR TYPES
Their rarity has prompted collaborative efforts at data
Yolk Sac Tumors
collection, and the development of the Prepubertal
Testicular Tumor Registry by the Section of Urology of Previous historic data suggested that yolk sac tumors were
the American Association of Pediatrics (AAP) in 1980. the most commonly diagnosed testis tumor in the pedi-
atric population, and this has been confirmed by AAP reg-
istry data. It comprised 62% to 63% of their tumors.3,18
PRESENTATION
However, other retrospective, single institution reviews
Eighty percent to ninety percent of patients present with have incidences of 66%,1 19%,5 and 8%.19 Most diag-
a painless mass.3–5 Trauma and swelling (5%) and hydro- noses occur in boys <2 years of age, with the median age
cele (2%) are the next most common modes of presenta- 16 months in data from both the AAP18 and the Hospital
tion.3 Leydig cell or granulosa tumors may present with for sick children (HSC) in Toronto, Canada.19
manifestations of testosterone production.6 In managing pediatric yolk sac tumors, it is important
Serology is a fundamental part of the initial evalua- to remember the physiologic elevations of AFP in the
tion. Given the preponderance of yolk sac tumors, alpha neonatal age group. Production from the gastrointestinal
fetoprotein (AFP) is very useful in the evaluation of the tract or the liver can result in levels as high as 50,000
prepubertal testicular mass. However, because it is pro- ng/ml in the neonate.20 Physiologic elevations diminish
duced during normal fetal development, AFP may be ele- quickly after birth but most remain above adult levels for
vated beyond adult norms in neonates.7 Because most up to 8 months.7 The prolonged half-life of 33 days dur-
yolk sac tumors in this age group are not associated with ing months 2 to 4 of life implies an element of neonatal
mixed germ cell tumors, human chorionic gonadotropin production.7 AFP binding affinity to concanavalin A may
(HCG) is not elevated. be useful in the assessing an elevation from a benign ver-
Scrotal ultrasound is an invaluable adjunct to treat- sus a malignant source.21
ment, as the age of the child or the presence of a large, Staging is according to the Children’s Cancer
tense hydrocele precludes a good physical examination.8 Group/Pediatric Oncology Group system. Stage I is
Sensitivity for detecting testicular neoplasia approaches defined as tumor confined to testis; stage II as micro-
100%9 and is accurate in distinguishing intratesticular scopic disease within the scrotum or spermatic cord and
from extratesticular lesions in up to 90% of cases.10 retroperitoneal adenopathy is < 2 cm; stage III has
However, the specificity of the histologic diagnosis retroperitoneal nodes >2 cm, but without visceral
afforded by ultrasound is much lower, and differentiation metastases; and stage IV is defined by distant metastases.

780
Chapter 49 Prepubertal Testicular Tumors 781

Eighty-five percent of patients present with stage I dis- Rhabdomyosarcoma Study (IRS) have been credited with
ease.8 the dramatic improvement in survival from 10% in the
Unlike adults, in this age group, retroperitoneal 1960s to over 70%.35 Paratesticular rhabdomyosarcomas
lymph node dissection (RPLND) has been associated are generally considered separate from other genitouri-
with a high rate of complications, including chylous nary rhabdomyosarcomas because of the difference in
ascites, bowel obstruction, gastrointestinal bleeding, lig- treatment and prognosis.34
ation of the renal artery, and a 10% to 40% incidence of The progenitor cell of the rhabdomyoblast is believed
ejaculatory dysfunction.22 Improved clinical staging with to be premature striated muscle derived from embryonic
computed tomography (CT) and AFP combined with mesenchyme. The most common subtype is embryonal
the success of cisplatin, etoposide, and bleomycin rhabdomyosarcoma, which also accounts for 97% of
chemotherapy has reduced the role of RPLND for yolk paratesticular cases.36 Microscopically, the cells are pri-
sac tumors. Contemporary indications are limited to a marily spindle shaped with abundant cytoplasm resem-
persistent retroperitoneal mass, an elevated AFP post- bling those of a 7- to 10-week fetus.37
orchiectomy with no radiographic evidence of metastatic Most paratesticular rhabdomyosarcoma patients
disease, or an unknown AFP preoperatively.8 (81%) present with a scrotal mass.36 Serum markers are
Disease-free survival at 5 years is comparable to that used to rule out primary testicular processes. Ultrasound
in the adult population, with the results of stages II to IV is very sensitive in differentiating testicular masses from
reported at >90%.23,24 Liu et al.25 demonstrated a 10% paratesticular masses,5,11,16 and thereby increases the
5-year survival advantage (91% versus 100%) using 2 doses probability that the scrotal lesion is a rhabdomyosar-
of cisplatin, vincristine, and bleomycin in stage I disease. coma. If discrete scrotal lymphatic drainage is violated
during transscrotal surgery for rhabdomyosarcoma in
particular, a hemiscrotectomy is recommended.38
Teratoma
The success of chemotherapy developed during the
The recently published 2002 AAP registry data listed ter- IRS studies I to III has resulted in RPLND no longer
atoma as accounting for 23% of testis tumors, a marked being required in all cases.39 This most recent protocol
increase from the 14% published in 1993.3,18 Single insti- mandates that patients with localized disease receive vin-
tution studies have shown an incidence of up to cristine and actinomycin for 1 year, with more advanced
60%,1,19,26 where it exceeds the incidence of yolk sac stages requiring actinomycin and cyclophosphamide in
tumors.20,26 This discrepancy may be accounted for by an longer, more intense regimes, combined with radiation
underreporting of benign tumors in the registry data.19,26 therapy. IRS-IV allowed for CT surveillance of the
Median age at presentation is 13 to 16 months, with a retroperitoneum, and this demonstrated that children
range of 0 to 146 months.18,19 There are at least two under the age of 10 years do not require an RPLND, as
reports of an antenatal diagnosis of teratomas.19,27 their incidence of metastatic nodal disease was signifi-
Preoperative findings consistent with a teratoma include cantly less. Moreover, successful treatment with
normal serology, as these tumors neither secrete nor stain chemotherapy was above 95%.40 However, in those boys
for AFP.20,28–30 Scrotal ultrasonography may show charac- over the age of 10 years, there is a 50% incidence of
teristic cystic areas with septa and solid stromal ele- retroperitoneal disease, and an ipsilateral RPLND is rec-
ments,10,31 but the specificity is only 50% to 59%.14,16,31 ommended.41,42 Positive lymph node metastasis man-
Mature teratomas in the prepubescent male contrast dates radiation therapy, as this is associated with a
sharply with their adult counterparts, as metastasis from significantly worse prognosis.42
teratoma have never been reported in children.3,18,26,32 The Children younger than 10 years of age have been
benign properties of this tumor are likely responsible for reported to have up to a 97% 5-year survival, while those
the increasing the popularity of treatment with partial older than 10 years had an 84% 5-year survival.43 Overall
orchiectomy. The finding of immature elements does not survival using IRS-II and IRS-I is 89% at 3 years, and 93%
deter from its benign natural history, provided all other for localized disease.44,45 Survivals estimated from IRS-III
investigations are in keeping with localized disease.33 and IRS-IV are 96% and 92%, respectively, the difference
not achieving statistical significance (p = 0.30).42 This suc-
cess has resulted in a secondary goal of reducing the mor-
Rhabdomyosarcoma
bidity of treatment and identifying biologic characteristics
Although a paratesticular tumor, it is included in this dis- to allow a more selective treatment regimens.46
cussion because it is an important consideration in the
differential diagnosis of a pediatric scrotal mass.4 It is the
Epidermoid Cysts
most frequent tumor of paratesticular origin and, despite
its aggressive behavior, has recently been associated with This universally benign entity accounts for 2% to 10% of
a good prognosis.34 The efforts of the Intergroup childhood testicular tumors3,18,19,26,27 and are less common
782 Part VIII Pediatric Malignancies

than in the adult population. Histologically, they consist of broma, hamartoma, leiomyoma, and neuroectodermal
a cyst wall filled with keratinizing squamous epithelium,47 tumor. Malignant tumors include leiomyosarcoma,
and their origin is thought to represent the monodermal fibrosarcoma, and reticular cell sarcoma.
development of a teratoma.48,49 Epidermoid cysts are hor-
monally inactive and usually present as a smooth, firm
Metastases to the Testis
intratesticular mass. They have a characteristic ultrasono-
graphic appearance that is consistent with the histology: a Infiltration of the testis with acute lymphoblastic
central hypoechoic mass with through transmission, a sur- leukemia (ALL) is a common cause of a prepubertal tes-
rounding echogenic rim, or mixed internal echogenic- ticular mass and may be bilateral.4,26 Hematologic malig-
ity.50–52 However, while ultrasound can raise the index of nancies accounted for half of testicular biopsy results in a
suspicion it cannot rule out a malignant process. single institution review.19 Children with bulky ALL at
Testis conserving surgery is accepted by many urolo- diagnosis have up to a 20% incidence of experiencing tes-
gists as the standard of care for this benign tumor.* There ticular metastases.58 Synchronous testicular metastases at
have never been any reports of metastases nor associa- the time of initial ALL diagnosis are associated with a
tions with intratubular germ cell neoplasia. The benign poor prognosis.59 The testicle is the second most com-
nature suggested by physical examination, serology, and mon site of a relapse of an extra-medullary ALL.
ultrasound, however, requires confirmation by frozen Moreover, Askin et al.58 found occult testicular leukemia
section before organ sparing can be performed safely. in 8.5% of biopsies in patients in clinical remission.
Follicular, Hodgkin’s, and Burkitt’s lymphomas can
also present in the testicle and have a favorable prognosis
Gonadal Stromal Tumors
if the tumor is completely intraparenchymal.60
This subgroup of tumors comprises Leydig cell, Sertoli
cell, and juvenile granulosa tumors, and accounts for 8%
TESTIS PRESERVING SURGERY
to 11% of pediatric testicular tumors.3,18 Although the
majority follows a benign course, reports of metastases The high incidence of benign tumors and the ability to
do exist. These tumors are unique as they are often hor- exclude malignancy histologically account for the
monally active. Leydig cell tumors account for approxi- increasing use of testis-sparing surgery in the prepuber-
mately 10% of cases of male precocious puberty.6 tal population. First advocated by Weissbach in 1984 for
Both adult Leydig cell and Sertoli cell tumors have a a solitary testicle,61,62 it has been advocated as the surgi-
10% malignancy rate in the adult population. This cal procedure of choice for all potentially benign
appears to be much lower in the pediatric population, but tumors.18
at least 5 cases of metastatic disease have been The combination of negative serology, an ultrasound
reported.54–56 No reliable histologic characteristics have that excludes a paratesticular mass, and the presence of
been identified, but invasion of the spermatic cord may salvageable testicular tissue in a prepubertal patient
be an important prognostic finding.6 Metastatic Sertoli should invite consideration of a testicular preserving
cell tumor is reported to be responsive to combination approach. After early control of the spermatic cord, a
chemotherapy.56 frozen section is obtained as a final determinant. Frozen
Juvenile granulosa tumors typically present in the section is very accurate in differentiating benign from
neonatal period. The median age at presentation in the malignant tumor, and is an essential confirmatory
latest AAP registry was 0.1 months, with a range of 0 to step.62,63 If the presumptive diagnosis is a teratoma, suf-
6 months. The clinical behavior has been universally ficient normal tissue should be analyzed to rule out
benign.18 pubertal changes, as the latter would necessitate radical
orchiectomy. A treatment algorithm incorporating the
principal benign pathologies and position for nonradical
Other Pediatric Testicular Tumors
surgery is depicted in Figure 49-1.
Gonadoblastoma may develop from a dysgenetic testis.
This tumor most commonly presents with virilization of a
SUMMARY
phenotypic female, often harboring some degree of an XY
karyotype.57 The tumor usually arises from an intraab- Testicular tumors in the prepubertal population are a dis-
dominal testis and histologically resembles a seminoma. tinct entity compared to their adult counterparts. There
Spread beyond the testicle has not been reported.1,4,57 is a predisposition towards benign tumors, while mixed
Connective tissue tumors arising from the testis that germ cell tumors are very rare. Serum HCG is not help-
have been reported include neuroblastoma, hemangiofi- ful, while an elevated AFP must be interpreted with cau-
tion in the neonate. Ultrasound is an excellent and
*References 13, 33, 48, 49, 53. invaluable tool, essential for the determination of a
Chapter 49 Prepubertal Testicular Tumors 783

History and physical consistent with painless


scrotal mass in prepubertal patient

Testicular
ultrasound

Suggests mature
Confirms or suggests teratoma, epidermoid
malignancy cyst; salvageable
parenchyma

Plan parenchymal
Paratesticular Baseline AFP sparing surgery; control
rhabdomyosarcoma serology cord, intraoperative
frozen section

Benign histology confirmed


on frozen section?
Technically salvageable
parenchyma?

NO TO YES TO
EITHER BOTH

Proceed with testis


preserving procedure:
Radical orchiectomy enucleate tumor, sample
parencymal bed as
indicated

Figure 49-1 Treatment algorithm for the prepubertal patient with a painless scrortal mass.

testicular from a paratesticular tumor. Although not REFERENCES


accurate enough to be definitive, ultrasound may help to
predict benign histology. However, frozen histopathol- 1. Brosman SA: Testicular tumors in prepubertal children.
ogy is the final arbiter of any testis-sparing procedure. Urology 1979; 13(6):581–588.
Characteristics of both the pediatric patients the 2. Kaplan GW, et al: Prepubertal yolk sac testicular
tumors themselves make testicular preserving surgery a tumors—report of the testicular tumor registry. J Urol
viable option. 1988; 140(5 Pt 2): 1109–1112.
784 Part VIII Pediatric Malignancies

3. Kay R: Prepubertal testicular tumor registry. J Urol 1993; 23. Hawkins EP, et al: Nongerminomatous malignant germ
150(2 Pt 2): 671–674. cell tumors in children. A review of 89 cases from the
4. Skoog SJ: Benign and malignant pediatric scrotal masses. Pediatric Oncology Group, 1971–1984. Cancer 1986;
Pediatr Clin North Am 1997; 44(5):1229–1250. 58(12): 2579–2584.
5. Aragona F, et al: Painless scrotal masses in the pediatric 24. Huddart SN, et al: The UK Children’s Cancer Study
population: prevalence and age distribution of different Group: testicular malignant germ cell tumours
pathological conditions—a 10 year retrospective 1979–1988. J Pediatr Surg 1990; 25(4): 406–410.
multicenter study. J Urol 1996; 155(4): 1424–1426. 25. Liu P, et al: Sonographic findings of testicular teratoma
6. Thomas JC, Ross JH, Kay R: Stromal testis tumors in with pathologic correlation. Pediatr Radiol 1992; 22(2):
children: a report from the prepubertal testis tumor 99–101.
registry. J Urol 2001; 166(6): 2338–2340. 26. Khoury AE, Bagli DJ, et al: Prepubertal testicular and
7. Wu JT, Book L, Sudar K: Serum alpha fetoprotein (AFP) paratesticular tumors. . J Urol 1996; 155: 392 (abstract).
levels in normal infants. Pediatr Res 1981; 15(1): 50–52. 27. Siu SS, et al: Prenatal diagnosis of intra-abdominal mature
8. Grady RW: Current management of prepubertal yolk sac testicular teratoma. J Ultrasound Med 2001; 20(11):
tumors of the testis. Urol Clin North Am 2000; 27(3): 1257–1260.
503–508 (ix). 28. Rushton HG, et al: Testicular sparing surgery for
9. Benson CB, Doubilet PM, Richie JP: Sonography of the prepubertal teratoma of the testis: a clinical and
male genital tract. Am J Roentgenol 1989; 153(4): 705–713. pathological study. J Urol 1990; 144(3): 726–730.
10. Krone KD, Carroll BA: Scrotal ultrasound. Radiol Clin 29. Gupta DK, Kataria R, Sharma MC: Prepubertal testicular
North Am 1985; 23(1): 121–139. teratomas. Eur J Pediatr Surg 1999; 9(3): 173–176.
11. Rifkin MD, et al: Diagnostic capabilities of 30. Klein EA: Tumor markers in testis cancer. Urol Clin
high-resolution scrotal ultrasonography: prospective North Am 1993; 20(1): 67–73.
evaluation. J Ultrasound Med 1985; 4(1): 13–19. 31. Kennedy PT, et al: Ultrasonography of intratesticular
12. Luker GD, Siegel MJ: Pediatric testicular tumors: lesions: its role in clinical management. Ulster Med
evaluation with gray-scale and color Doppler US. J 1999; 68(2): 54–58.
Radiology 1994; 191(2): 561–564. 32. Brown NJ: Teratomas and yolk-sac tumours. J Clin Pathol
13. Altadonna V, et al: Simple cysts of the testis in children: 1976; 29(11): 1021–1025.
preoperative diagnosis by ultrasound and excision with 33. Walsh C, Rushton HG: Diagnosis and management of
testicular preservation. J Urol 1988; 140(6): 1505–1507. teratomas and epidermoid cysts. Urol Clin North Am
14. Tackett RE, et al: High resolution sonography in 2000; 27(3): 509–518.
diagnosing testicular neoplasms: clinical significance of 34. Blyth B, et al: Paratesticular rhabdomyosarcoma: results
false positive scans. J Urol 1986; 135(3): 494–496. of therapy in 18 cases. J Urol 1990; 144(6): 1450–1453.
15. Bauer S, Hendren H: Infantile cystic teratoma of the 35. Crist WM, et al: Prognosis in children with
testis: Children’s Hospital experience and review of the rhabdomyosarcoma: a report of the intergroup
literature. J Urol 1998; 159(163) (abstract No. 624). rhabdomyosarcoma studies I and II. Intergroup
16. Polak V, Hornak M: The value of scrotal ultrasound in Rhabdomyosarcoma Committee. J Clin Oncol 1990; 8(3):
patients with suspected testicular tumour. Int Urol 443–452.
Nephrol 1990; 22(5): 467–473. 36. Raney B Jr, et al: Primary site as a prognostic variable for
17. Maxwell AJ, Mamtora H: Sonographic appearance of children with pelvic soft tissue sarcomas. J Urol 1986;
epidermoid cyst of the testis. J Clin Ultrasound 1990; 136(4): 874–878.
18(3): 188–190. 37. Patton RB, Horn RC Jr.: Rhabdomyosarcoma: clinical and
18. Ross JH, Rybicki L, Kay R: Clinical behavior and a pathological features and comparison with human fetal
contemporary management algorithm for prepubertal and embryonal skeletal muscle. Surgery 1962; 52:572.
testis tumors: a summary of the Prepubertal Testis Tumor 38. Rogers DA, et al: Indications for hemiscrotectomy in the
Registry. J Urol 2002; 168(4 Pt 2): 1675–1678 (Discussion management of genitourinary tumors in children.
1678-9). J Pediatr Surg 1995; 30(10): 1437–1439.
19. Metcalfe PD, Farivar-Mohseni H, Farhat W, et al: 39. Crist WM, et al: Intergroup rhabdomyosarcoma study-IV:
Pediatric testicular tumors: contemporary incidence and results for patients with nonmetastatic disease. J Clin
efficacy of testicular preserving surgery. J Urol 2003; Oncol 2001; 19(12): 3091–3102.
170(6pt1):2412–2415 (Discussion 2415–2416). 40. Rodney C, Flanant F, Maurer H, et al: Initial
20. Grady RW, Ross JH, Kay R: Epidemiological features of lymphadenectomy is not necessary in localized and
testicular teratoma in a prepubertal population. J Urol completely resected paratesticular rhabdomyosarcoma.
1997; 158(3 Pt 2): 1191–1192. Med Pediatr Oncol 1992; 20: 430 (abstract).
21. Lahdenne P, et al: Biphasic reduction and concanavalin 41. Kattan J, et al: Paratesticular rhabdomyosarcoma in adult
A binding properties of serum alpha-fetoprotein in patients: 16-year experience at Institut Gustave-Roussy.
preterm and term infants. J Pediatr 1991; 118(2): Ann Oncol 1993; 4(10): 871–875.
272–276. 42. Wiener ES, et al: Controversies in the management of
22. Green DM: The diagnosis and treatment of yolk sac paratesticular rhabdomyosarcoma: Is staging
tumors in infants and children. Cancer Treat Rev 1983; retroperitoneal lymph node dissection necessary for
10(4): 265–288. adolescents with resected paratesticular
Chapter 49 Prepubertal Testicular Tumors 785

rhabdomyosarcoma? Semin Pediatr Surg 2001; 10(3): 53. Eisenmenger M, et al: Epidermoid cysts of the testis:
146–152. organ-preserving surgery following diagnosis by
43. Wiener ES, et al: Retroperitoneal node biopsy in ultrasonography. Br J Urol 1993; 72(6):955–957.
paratesticular rhabdomyosarcoma. J Pediatr Surg 1994; 54. Kolon TF, Hochman HI: Malignant Sertoli cell tumor in
29(2): 171–177 (Discussion 178). a prepubescent boy. J Urol 1997; 158(2): 608–609.
44. Maurer HM, et al: The Intergroup Rhabdomyosarcoma 55. Kapoor HL, et al: Malignant interstitial cell tumour of
Study-I. A final report. Cancer 1988; 61(2): 209–220. testis in a child. Indian J Cancer 1988; 25(4): 241–245.
45. Maurer HM, et al: The Intergroup Rhabdomyosarcoma 56. Sharma S, Seam RK, Kapoor HL: Malignant Sertoli cell
Study-II. Cancer 1993; 71(5): 1904–1922. tumour of the testis in a child. J Surg Oncol 1990; 44(2):
46. Shapiro DN, et al: Relationship of tumor-cell ploidy to 129–131.
histologic subtype and treatment outcome in children and 57. Scully RE: Gonadoblastoma. A review of 74 cases. Cancer
adolescents with unresectable rhabdomyosarcoma. J Clin 1970; 25(6): 1340–1356.
Oncol 1991; 9(1): 159–166. 58. Askin FB, et al: Occult testicular leukemia: testicular
47. Shah KH, Maxted WC, Chun B: Epidermoid cysts of the biopsy at three years continuous complete remission of
testis: a report of three cases and an analysis of 141 cases childhood leukemia: a Southwest Oncology Group Study.
from the world literature. Cancer 1981; 47(3): 577–582. Cancer 1981; 47(3): 470–475.
48. Ross JH, Kay R, Elder J: Testis sparing surgery for 59. Godinho C, et al: Testicular relapse in acute
pediatric epidermoid cysts of the testis. J Urol 1993; lymphoblastic leukemia. The experience of a pediatrics
149(2): 353–356. service. Acta Med Port 1995; 8(11): 613–618.
49. Heidenreich A, et al: Organ preserving surgery in 60. Lamm DL, Kaplan GW: Urological manifestations of
testicular epidermoid cysts. J Urol 1995; 153(4): Burkitt’s lymphoma. J Urol 1974; 112(3): 402–405.
1147–1150. 61. Weissbach L, Altwein JE, Stiens R: Germinal testicular
50. Cohen EL, et al: Epidermoid cyst of testicle. tumors in childhood. Report of observations and literature
Ultrasonographic characteristics. Urology 1984; 24(1): review. Eur Urol 1984; 10(2): 73–85.
79–81. 62. Elert A, et al: Accuracy of frozen section examination of
51. Nichols J, et al: Epidermoid cyst of testis: a report of 3 testicular tumors of uncertain origin. Eur Urol 2002;
cases. J Urol 1985; 133(2): 286–287. 41(3): 290–293.
52. Shapeero LG, Vordermark JS: Epidermoid cysts of testes 63. Tokuc R, et al: Accuracy of frozen section examination of
and role of sonography. Urology 1993; 41(1): 75–79. testicular tumors. Urology 1992; 40(6): 512–516.
INDEX
Ablation Adenocarcinoma of prostate (Continued) Adenocarcinoma of prostate (Continued)
adrenal tumors with, 149 NHT with radiation therapy in, 495–496 biochemical relapse-free survival with,
renal tumor with, 207–210, 208t nilutamide in, 495 477, 478, 479–481, 480t, 481t, 482,
cryoablation in, 207–208, 208t orchiectomy as, 494–495 482f, 485–489, 485t, 487f, 489f
CT with, 208, 209 PEP with, 506 CT scan with, 478, 483, 484, 484f
HIFU, 209–210 peripheral androgen blockade and, 508 CTV with, 478–479, 479f
MRI with, 208, 209 progesterone supplementation as, 495 3DCRT with, 478–479, 478f, 482, 489
radiofrequency, 208–209, 208t prostatectomy with, 498–499 dose escalation with, 481–482, 481t
ACD. See ASTRO Consensus Definition radiation therapy with, 495–498, 497f EBRT historic perspective, 478
ACTH. See Adrenocorticotrophic hormone rationale in, 494 EBRT technique for, 478–479, 479f
Acute leukemia (AL) RTOG and, 495–498, 499, 500, 501t, GTV with, 478–479, 479f
nongerm cell tumors of, 636 502 HIFU with, 477
Acute lymphocytic leukemia (ALL) secondary hormonal therapy and, 508 IMRT with, 478, 482
nongerm cell tumors of, 636, 636f standards of care in, 505–506 MR with, 478, 483
Acute myelogenous leukemia (AML) timing of, 498, 507 NHT with, 495–496
diagnosis of, 179, 180, 180f toxicity with, 499 PSA levels with, 477–478, 479, 480,
Acute urinary retention (AUR) future directions for, 500–502 481t, 482f, 485, 485f, 486–488, 487t
brachytherapy with, 119 interstitial brachytherapy for, 483–489, PTV with, 478–479, 479f
Adaptive cellular therapy 484f, 484t, 485t, 487t, 489t risk stratification with, 478
immunotherapy with, 93, 94f BRFS following, 485–489, 485t, 487f, RTC with, 477
LAK, 93, 94f 489f RTOG with, 477, 478, 482, 483, 485
TIL, 93, 94f HDR, 488–489, 489t T1-2 PC in, 477, 482–483
ADCC. See Antibody-dependent cell- historic perspective on, 483 timing/duration of hormonal therapy
mediated cytotoxicity PPB EBRT combined in, 486–488, with, 498
Adenocarcinoma of prostate 487t radical prostatectomy for, 465–474, 466t,
androgen deprivation therapy for, PPB technique in, 483–484, 484f 468t, 469t, 471t, 472t, 473t
494–502, 497f, 500t, 501t T1-2 cancer with, 488 biopsy Gleason sum with, 471, 472f
adjuvant hormonal with radiation TPB technique in, 484–485 blood loss during, 469, 469f
therapy in, 496–498, 497f management of, 465–474, 466t, 468t, cancer control with, 469f, 470–473,
alternative approaches with, 507–508 469t, 471t, 472t, 473t, 494–502, 471f, 472f
aminoglutethimide in, 495 497f, 500t, 501t CT scan assessment with, 466
antiandrogens as, 495 general, 494 early complications with, 468–470,
bicalutamide in, 495 metastasis of, 505–511, 509t, 510t 468f, 469f
CAB with, 505, 506, 508 bone-directed therapies for, 511 erectile function after, 474
cyproterone acetate in, 495 chemotherapy for, 509–511, 510t hospitalization days with, 469f
EBRT with, 494, 496–498, 499, 500, hormone therapy for, 505–509, 509t intraoperative complications with,
501, 502 summary of, 511 468–470, 468f, 469f
EORTC and, 496–497, 497f, 501, 501t, outcomes for, 500 late complications with, 470
506, 507 particle beam therapy for, 489 lymph node metastases with, 472t
estrogen supplementation as, 495 prognosis for, 465–474, 466t, 468t, 469t, mortality with, 468f
flutamide in, 495 471t, 472t, 473t, 494–502, 497f, MRI assessment with, 466
FSH in, 495, 500t 500t, 501t obturator nerve injury from, 470
goserelin in, 495 radiation therapy for, 477–489, 478f, outcomes after, 469f, 470–474, 471f,
intermittent adenocarcinoma and, 479t, 480t, 481t, 482f, 484f, 484t, 472f, 473f
507–508 485t, 487t, 489t patient selection for, 466
leuprolide in, 495 ACD with, 479, 483, 485, 487–488 PC natural history and, 465–466, 466t
LH in, 495, 500t adjuvant hormonal with, 496–498, 497f pelvic lymph node dissection with,
LHRH in, 495, 496, 498, 500t, ADT and, 479, 480, 482–483, 486, 487, 467–468
505–507, 508, 511 488 pretreatment risk stratification with,
medroxyprogesterone in, 495 androgen deprivation therapy with, 466–467
megestrol in, 495 495–498, 497f PSA risk assessment with, 466–467,
methods of, 494–495 ASTRO with, 479, 481 470, 471f

Page numbers followed by f indicate figures; page numbers followed by t indicate tables.

787
788 Index

Adenocarcinoma of prostate (Continued) Adrenal tumors (Continued) Adrenal tumors (Continued)


PSA survival with, 469f hyperaldosteronism in, 137–138, urologic oncology and, 131–151, 132f,
rectal injury from, 469 137f, 138f 133t, 134f, 135f, 136t, 137f, 138f,
results of, 468 aldosterone secretion with, 138, 138f 140t, 141f, 142t, 143f, 144f, 145f,
risk factors with, 472f, 473f CT scan for, 137f, 138 146f, 147f, 148f, 150f, 151f,
seminal vesicle involvement with, 472f diagnosis of, 154t, 155 153–167
treatment rationale, 465 hypernatremia with, 137 Adrenalectomy
urinary function with, 473, 473f hypertension with, 137, 137f, 154–155 adrenal tumors treated with, 139–149, 142t,
urinary incontinence after, 473, 473f hypokalemia with, 137–138, 155 143f, 144f, 145f, 146f, 147f, 148f
regionally advanced, 494–502, 497f, 500t, PRA with, 137, 137f approaches to, 141, 142t
501t RASS with, 137 Cushing’s syndrome in, 141, 142t
androgen deprivation and secondary, 138 flank approach to, 145–147, 145f, 146f,
prostatectomy for, 498–499 incidentally discovered, 133–136, 135f 156–158, 157f
androgen deprivation and radiation CT scan in, 133–136, 135f hyperaldosteronism in, 142t
therapy for, 494–498, 497f MRI in, 134, 135f lateral flank approach in, 156–158, 157f
androgen deprivation therapy for, ultrasound in, 133 left, 163–165, 163f
494–502, 497f, 500t, 501t laparoscopic surgery for, 149, 150–151f, modified posterior approach to, 143–145,
future directions for, 500–502 160–167, 161f, 161t, 162f, 163f, 144f, 159, 160f
outcomes for, 500 164f, 166t nephrectomy with, 146, 146f
prognosis for, 500 anterior transperitoneal approach in, partial, 148–149, 148f, 167
toxicity with, 499–500 161–164, 162f, 163f pheochromocytoma in, 142t
urethral cancer pathology with, 674 approaches to, 161 posterior approach to, 142–143, 143f,
Adenomatoid tumor of epididymis complications of, 165 159, 159f
nongerm cell tumors of, 618t, 633, 633f, indications for, 160–161, 161t preoperative management with, 141, 142t
634f left adrenalectomy in, 163–165, 163f RCC with, 204–205
Adenovirus vectors malignant tumors in, 167 right, 161–163, 162f, 165
applications of, 21–22, 21t posterior retroperitoneal approach thoracoabdominal approach to, 158, 158f
attributes in, 21–22, 21t in, 165 transabdominal approach to, 147–148, 147f
gene delivery in, 21 postoperative care with, 165 Adrenocortical carcinoma
gene expression in, 21 preoperative preparation in, 161, Cushing’s syndrome with, 155
gene therapy with, 21–22, 21t, 22f, 26t 161f description of, 155–156
vector production in, 21 references to, 166t Adrenocorticotrophic hormone (ACTH)
ADEPT. See Antibody-dependent enzyme- results of, 165 Cushing’s syndrome with, 132–133
mediated cytotoxicity retroperitoneal technique in, 164, 164f ADT. See Androgen deprivation therapy
Adrenal carcinoma right adrenalectomy in, 161–163, EBRT with, 482–483
adrenal tumors with, 136–137, 136t 162f, 165 PPB with, 487
adrenocortical, 155–156 transthoracic technique in, 165 radiation therapy for adenocarcinoma
classification of, 136t pheochromocytoma in, 138–139, 138f, with, 479, 480, 482–483, 486, 487,
Cushing’s syndrome with, 136 140t, 141f, 142t, 143f, 148f 488
DHEA with, 136 APUD with, 139 Adult granulosa cell tumors (AGCT)
gynecomastia with, 136 cardiomyopathy with, 139 nongerm cell tumors as, 624–625, 626
Mitotane with, 136 CT scan for, 139, 155 AFP. See α-fetoprotein
oligospermia with, 136 diagnosis of, 155 AGCT. See Adult granulosa cell tumors
Adrenal tumors glomus jugulare tumors with, 139, 148f Agent Orange
ablation for, 149 identifying, 138, 138f, 154t retroperitoneal tumors from, 652
carcinoma, 136–137, 136t MEA syndromes with, 139 AgNOR. See Silver staining nucleolar
classification of, 136t MIBG scan for, 139, 155 organizer regions
Cushing’s syndrome with, 136 MRI scan for, 139, 141f, 143f, 148f, AGR syndrome. See Aniridia, genitourinary
DHEA with, 136 155 malformations and mental
gynecomastia with, 136 neurofibromatosis with, 139, 140t retardation syndrome
Mitotane with, 136 Sturge-Weber syndrome with, 139 AIS. See Androgen insensitivity syndrome
oligospermia with, 136 surgical approach to, 155 AJCC. See American Joint Committee of
cryosurgery for, 149 symptoms of, 140t, 155 Cancer
Cushing’s syndrome and, 131, 132–133, von Hippel-Landau with, 139, 140t AL. See Acute leukemia
133t, 134f physiological understanding for, Aldosterone secretion
ACTH with, 132–133 131, 132f hyperaldosteronism with, 138, 138f
CRH with, 132–133 arterial supply in, 132f Alkylating agents
description of, 132, 155 CT in, 131 genitourinary cancer treatment with,
diagnosis of, 132, 134f, 154t, 155 IVC in, 131, 132f 67–68
manifestations of, 133t MRI in, 131 ALL. See Acute lymphocytic leukemia
diagnosis of, 153–154, 154f, 154t venous drainage in, 132f American Joint Committee of Cancer
CT scan in, 153, 154f summary of, 149 (AJCC), 196, 196t, 259
Cushing’s syndrome in, 154t surgery for, 139–149, 142t, 143f, 144f, bladder cancer staging with, 308, 319
hyperaldosteronism in, 154t 145f, 146f, 147f, 148f, 153–167, PC staging with, 462, 549–461
MRI in, 153, 154f 154f, 154t, 157f, 158f, 159f, 160f, TCC staging of, 274t
pheochromocytoma in, 154t 161f, 161t, 162f, 163f, 164f, 166t testis tumors staging with, 569, 571, 572t
Index 789

American Society of Therapeutic Radiology Aniridia, genitourinary malformations and Bicalutamide


and Oncology (ASTRO) mental retardation syndrome androgen deprivation therapy
radiation therapy for adenocarcinoma (AGR syndrome) with, 495
with, 479, 481 Wilms’ tumor with, 754, 754t, 759t Bilateral pelvic lymph node dissection
Amine precursor uptake and Anorexia (BPLND)
decarboxylation (APUD) orthotopic bladder substitution with, 453t RPP patient selection and, 528
pheochromocytoma with, 139 Anthracycline antibiotics Biochemical relapse-free survival
Aminoglutethimide genitourinary cancer treatment with, 62f, (BRFS)
androgen deprivation therapy with, 495 63–64 adenocarcinoma of prostate with, 477,
AML. See Acute myelogenous leukemia Antiandrogens 478, 479–481, 480t, 481t, 482,
Amyloidosis androgen deprivation therapy with, 495 482f, 485–489, 485t, 487f, 489f
RCC associated with, 174t Antibody-dependent cell-mediated brachytherapy with, 485, 485t
Anastomotic margin biopsy cytotoxicity (ADCC) dose escalation and, 481t, 482, 482f
orthotopic bladder substitution with, 444 immunotherapy with, 98t EBRT with, 479–481, 480t
Androgen deprivation therapy (ADT) Antibody-dependent enzyme-mediated HDR brachytherapy with, 488–489, 489t
prostate adenocarcinoma treated with, cytotoxicity (ADEPT) PPB alone with, 485–486, 485t
494–502, 497f, 500t, 501t immunotherapy with, 98t PPB EBRT combined with, 486–488,
adjuvant hormonal with radiation Anticancer antibiotics 487t, 489t
therapy in, 496–498, 497f anthracycline antibiotics, 62f, 63–64 Birt-Hogg-Dubé (BHD)
alternative approaches with, 507–508 bleomycin, 61–63, 62f RCC with, 176, 197t
aminoglutethimide in, 495 doxorubicin, 62f, 63–64 Bladder cancer. See also Bladder preservation;
antiandrogens as, 495 mitomycin, 62f, 63 Transitional cell carcinoma
bicalutamide in, 495 mitoxantrone, 62f, 64 cystectomy for, 328–329, 329t
CAB in, 505, 506, 508 Antifolates indications for, 328, 329t
cyproterone acetate in, 495 genitourinary cancer treatment with, 70 partial, 329
EBRT with, 494, 496–498, 499, 500, Antimetabolites radical, 328–329
501, 502 genitourinary cancer treatment with, 70 development pathways of, 303f
EORTC and, 496–497, 497f, 501, 501t Antioncogene therapy dysplasia in, 303f
estrogen supplementation as, 495 gene therapy with, 30–31 hyperplasia in, 303f
flutamide in, 495 APUD. See Amine precursor uptake and lymphatic permeation in, 303f
FSH in, 495, 500t decarboxylation vascular permeation in, 303f
goserelin in, 495 ASTRO. See American Society of diagnosis of, 301–313, 302f, 303f, 304f
intermittent ADT in, 507–508 Therapeutic Radiology and follow-up for, 329–330, 329t
leuprolide in, 495 Oncology genetic changes with, 304f
LH in, 495, 500t ASTRO Consensus Definition (ACD) invasive carcinoma in, 304f
LHRH in, 495, 496, 498, 500t, radiation therapy for adenocarcinoma lymphatic permeation in, 304f
505–507, 508, 511 with, 479, 483, 485, 487–488 vascular permeation in, 304f
medroxyprogesterone in, 495 Atraumatic urethra dissection HRQOL with, 108–109
megestrol in, 495 orthotopic bladder substitution with, 446 mucosa distant from tumor in, 320
methods of, 494–495 staging of, 301, 302f, 303f, 308–313
NHT with radiation therapy in, Bacille Calmette-Guerin (BCG) AJCC in, 308
495–496 bladder cancer treated with, 92–93 carcinoma in situ in, 310
nilutamide in, 495 IFN and, 92 dysplasia in, 312
orchiectomy as, 494–495 immunotherapy with, 92–93, 94f lymph node involvement in, 311
PEP in, 506 TCC prognostic markers with, 321 muscle-invasive cancer in, 310–311
peripheral androgen blockade and, 508 TCC with, 276 TNM classification in, 308, 309
progesterone supplementation as, 495 Balanitis xerotica obliterans (BXO) statistics on, 338
prostatectomy with, 498–499 penis carcinoma with, 711, 712 TUR for
radiation therapy with, 495–498, 497f Ball electrode anesthesia for, 359
rationale in, 494 TUR instrumentation with, 359 bimanual examination for, 359, 359f
RTOG and, 495–498, 499, 500, 501t, BCG. See Bacille Calmette-Guerin bladder cancer diagnosis with, 306, 338
502 Beckwith-Wiedemann syndrome (BWS Bladder-sparing therapy with, 347–348
secondary hormonal therapy and, 508 syndrome) bladder tumors with, 358–365, 359f,
standards of care in, 505–506 Wilms’ tumor with, 754, 754t, 759t 360f, 364t, 365f
timing/duration of, 498 Benchekroun nipple complications with, 362–364
timing in, 507 urinary diversion approach of, 409 equipment for, 359–360
toxicity with, 499 Benign lesions fluoroscopic cystoscopy for, 365
seminal vesicles treatment options with, ganglioneuroma as, 652t instrumentation for, 359–360
562 hemangiopericytoma as, 652t intraoperative problem management in,
Androgen insensitivity syndrome (AIS) leiomyoma as, 652t, 653 361–362
SCA with, 622, 624 lipomas as, 652–653, 652t irrigation fluid in, 361
Angiogenic factors myelolipoma as, 652t, 653 laser surgery for, 364–365, 364t, 365f
MVD with, 341–342 retroperitoneal tumors with, 652–653, 652t postoperative management in, 362
TCC with, 341–342 Schwannoma as, 652t preparation for, 358–359
TSP-1 with, 341 BEP. See Bleomycin, etoposide and cisplatin primary muscle invasive tumor
VEGF with, 341 BHD. See Birt-Hogg-Dubé treatment with, 362
790 Index

Bladder cancer (Continued) Brachytherapy Carcinoma in situ (CIS). See also Testicular
second look resection in, 362 CT-guided, 119 Intraepithelial neoplasia
TCC bladder with, 317–318 outcome of, 119 bladder cancer staging with, 310.339
TCC diagnosis with, 338 procedure for, 119 laparoscopic partial nephrectomy for, 231
technique for, 360–361 HDR, 488–489, 489t penis carcinoma with, 712–713
types of, 338 HRQOL with, 107 seminoma with, 579
Bladder explosion IGT with, 118–122, 120f, 121f Transurethral resection (TUR), 359
TUR complications with, 364 MR-guided, 114f, 115f, 119–122, 120f, Cardiomyopathy
Bladder perforation 121f pheochromocytoma with, 139
TUR complications with, 363 outcome of, 120–122 Carney’s syndrome
Bladder preservation patient selection for, 119 LCCSCT with, 623–624
5-FU, 391t procedure for, 114f, 115f, 119–120, Case histories
5-FU for, 390, 390t 120f, 121f TCC bladder, 330–331
chemotherapy for, 389, 390t TURP and, 119 CDC. See Complement-dependent
combined modality treatment for, penis carcinoma with, 705–706 cytotoxicity
389–397, 390t, 391t, 393f, 394f, prostate adenocarcinoma treated with, CDKI. See Cycle kinase inhibitor
395f, 396t, 397t 483–489, 484f, 484t, 485t, 487t, 489t Cell adhesion molecules (CAMS), 13, 13f
MCV for, 390t BRFS following, 485–489, 485t, 487f, TCC with, 341
monotherapy’s influence on, 389, 390t 489f Cell-cycle regulators
multimodal treatment schema for, 394f historic perspective on, 483 TCC prognosis with, 342–343
multimodality treatment results for, 390, PPB EBRT combined in, 486–488, 487t erb-B-2 in, 342–343
390t PPB technique in, 483–484, 484f P53 in, 342
outcome of, 395f, 396t T1-2 cancer with, 488 retinoblastoma gene in, 342
radiation therapy for, 389, 390t TPB technique in, 484–485 TGF in, 343
RTOG and, 391 radiation oncology with, 40–41, 42f Cellular biology
single agent activity in, 391, 391t ultrasound guided, 118–119 aging/telomerase in, 16
5-FL, 391t BRFS. See Biochemical relapse-free survival aptamers in, 16–17
carboplatin, 391t Bropirimine cancer genes in, 8–10
cisplatin, 391, 391t bladder TCC with, 327 DNA methylation, 9
docetaxel, 391t Bulbourethral DNA repair, 8
doxorubicin, 391t cancer in, 3, 4f oncogenes, 8
gemcitabine, 391t Buschke-Löwenstein tumor suppressor, 8
ifosfamide, 391t penis carcinoma with, 712, 723 viral, 8–9
methotrexate, 391, 391t BWS syndrome. See Beckwith-Wiedemann carcinogens in, 4–5
mitomycin C, 391t syndrome cell cycle in, 11–12, 12f
paclitaxel, 391, 391t BXO. See Balanitis xerotica obliterans cell signaling in, 12f, 14–15, 14f
vinblastine, 391t CGH in, 8
TUR for, 389, 390t, 394f CAB. See Combination androgen blockade epigenetic effects in, 3–4, 4f
TURBIT for, 390t, 392 CAH. See Congenital adrenal hyperplasia familial cancer in, 7–8
Bladder-sparing therapy CAIX. See Carbonic anhydrase IX inflammation in, 5–7, 6t
complications of, 349 Camptothecins microarrays in, 10–11
multimodality approach to, 348 genitourinary cancer treatment with, proteomics in, 10–11
outcomes of, 348–349 72–74, 73f SKY in, 8
quality of life concerns with, 349 irinotecan as, 73–74, 73f stromal epithelial interactions in, 15–16,
rationale for, 347 topotecan as, 73f, 74 15f
single modality approaches to, CAMS. See Cell adhesion molecules tumor cell heterogeneity in, 16–17, 17f
347–348 Cancer genes urologic cancers in, 3–17, 4f, 6t, 10f, 12f,
TCC with, 347–349 DNA methylation, 9 13f, 14f, 15f, 17f
TUR in, 347–348 DNA repair, 8 CGH. See Comparative genome
Bleomycin molecular biology of, 8–10 hybridization
genitourinary cancer treatment with, oncogenes, 8 Chemotherapy
61–63, 62f suppressor, 8 agents used in, 59–74, 60f, 62f, 66f, 69f,
Bleomycin, etoposide and cisplatin viral, 8–9 72f, 73f
(BEP) CAPD. See Continuous ambulatory alkylating agents as, 67–68
NSGCT treatment with, 606 peritoneal dislysis anthracycline antibiotics as, 62f, 63–64
Bone marrow transplantation Carbonic anhydrase IX (CAIX) anticancer antibiotics as, 61–64, 62f
RCC with, 265 RCC with, 176 antifolates as, 70
Bosniak classification, 182, 182t Carboplatin antimetabolites as, 70
Bosniak lesions bladder preservation with, 391t bleomycin as, 61–63, 62f
RCC diagnosis with, 181–182 genitourinary cancer treatment with, camptothecins as, 72–74, 73f
Bowenoid papulosis 60–61, 60f capecitabine as, 71
penis carcinoma with, 712 Carcinogens carboplatin as, 60–61, 60f
Bowen’s disease molecular biology of, 4–5 cisplatin as, 59–60, 60f
penis carcinoma with, 712–713 Carcinoid tumors (CT) cyclophosphamide as, 68
BPLND. See Bilateral pelvic lymph node nongerm cell tumors as, 618t, 632–633, docetaxel as, 66f, 67
dissection 633f doxorubicin as, 62f, 63–64
Index 791

Chemotherapy (Continued) Chronic leukemia (CL) Computed tomography (CT) (Continued)


epipodophyllotoxin as, 69–70, 69f nongerm cell tumors of, 636 PC staging with, 461
estramustine as, 67 CIS. See Carcinoma in situ penis carcinoma with, 714
etoposide as, 69, 69f CISCA. See Cisplatin, cyclophosphamide, pheochromocytoma with, 139
fluoropyrimidines as, 70–71 adriamycin prostatectomy assessed with, 466
gemcitabine as, 71–72, 72f Cisplatin radiation therapy for adenocarcinoma
ifosfamide as, 68–69 bladder preservation with, 391, 391t with, 478, 483, 484, 484f
irinotecan as, 73–74, 73f genitourinary cancer treatment with, radical nephrectomy with, 218
methotrexate as, 70 59–60, 60f RCC staging with, 186–188, 186f
mitomycin as, 62f, 63 Cisplatin, cyclophosphamide, adriamycin renal diagnosing with, 179–182, 180f,
mitoxantrone as, 62f, 64 (CISCA) 181f, 182f, 197
oxaliplatin as, 60f, 61 TCC chemotherapy treatment with, 281 renal tumor ablation with, 208, 209
paclitaxel as, 65–67, 67f Cisplatin, methotrexate, vinblastine (CMV) retroperitoneal tumors diagnosis of,
platinum complexes as, 59–61, 60f TCC chemotherapy treatment with, 281 657–658, 657f
taxanes as, 65–67, 67f Cisplatin, Velban and bleomycin (PVB) RMS evaluation with, 767–768
thiotepa as, 69 NSGCT treatment with, 605 RPLND with, 642, 647, 647f
topotecan as, 73f, 74 CL. See Chronic leukemia seminal vesicles treatment options with,
tubulin modulation drugs as, 64–65 Classic Sertoli cell tumors (CSCT) 558, 563f
vinca alkaloids as, 64–65 nongerm cell tumors as, 622–623, 622f, squamous cell carcinoma of penis with,
anticancer agent pharmacology of, 54–56 623f 696
absorption in, 54 pathologic characteristic of, 622–623, TCC diagnosis with, 271–272
distribution in, 54–55 622f, 623f testis tumors staging with, 569–571
excretion in, 55–56 Clinical target volume (CTV) Wilms’ tumor evaluation with, 756,
factors modifying, 56 brachytherapy and, 119–120 756f, 758f
metabolism in, 55 radiation oncology with, 41 Conditionally replication-competent
transport in, 54–55 radiation therapy for adenocarcinoma oncolytic adenovirus (CRAd), 30
bladder TCC with, 323–324 with, 478–479, 479f Condyloma acuminatum
doxorubicin in, 324 CMV. See Cisplatin, methotrexate, penis carcinoma with, 712
MMC in, 323–324, 326t vinblastine Congenital adrenal hyperplasia (CAH)
thiotepa in, 323 Cold-cup biopsy Leydig cell tumors v., 621, 621f
valrubicin in, 324 TUR equipment including, 360 Continent mechanism
drug resistance mechanisms in, 56–58, 57t Colonic conduits appendiceal approach to, 409
models for overcoming, 56–58, 57t noncontinent cutaneous urinary diversion Benchekroun nipple approach to, 409
genitourinary cancer treatment with, with, 404–406, 405f cutaneous urinary diversion with,
51–74, 57t, 59t, 60f, 62f, 66f, 69f, ileocecal conduit for, 405 409–411, 410f
72f, 73f sigmoid colon conduit for, 406 flap valve approach to, 409
macro-pharmacokinetic mechanisms with, transverse colon conduit for, 404–406, four surgical techniques for, 409
57t 405f hydraulic valve approach to, 409
micro-pharmacokinetic mechanisms with, Colony-stimulating factor (CSF) intussuscepted nipple valve approach to,
57t cytokine therapy with, 92, 93t 409
penis carcinoma with, 718 HCT with, 93 pseudoappendiceal approach to, 409
pharmacodynamic intracellular immunotherapy with, 92, 93, 93t Continent reservoirs
mechanisms with, 57t Combination androgen blockade (CAB) cutaneous urinary diversion with,
prostate adenocarcinoma treated with, androgen deprivation therapy with, 505, 411–427, 412f, 413f, 414f, 415f,
509–511, 510t 506, 508 416f, 417f, 418–420f, 421f, 422f,
RMS treated with, 773–774 peripheral androgen blockade and, 508 422t, 423–424t, 425f, 426f, 427f,
stage I seminoma with, 586 prostate adenocarcinoma treated with, 428f, 436–437
summary of, 74 505, 506, 508 Duke pouch, 422t, 424t
TCC radical cystectomy with, 346–347 Comparative genome hybridization Florida pouch, 422t, 424t
treatment efficacy assessment for, 58–59, (CGH) gastric pouch, 424t, 426–427, 427f, 428f,
59t cancer with, 8 436–437
classification by phase in, 59 Complement-dependent cytotoxicity Indiana pouch, 416–421, 422t, 423–424t,
explanatory trial in, 58 (CDC) 425f, 436
pragmatic trial in, 58–59 immunotherapy with, 98t Kock pouch, 411–413, 412f, 413f, 423t,
tumor cell biology related to, 51–54 Computed tomography (CT). See also 436
cell cycle control in, 52–53 Multidetector spiral computed LeBag, 422t
cell proliferation/apoptosis balance in, tomography Mainz pouch, 414–416, 418–420f, 423t,
53–54 bladder cancer diagnosis with, 302, 304, 436
cellular kinetics in, 52–53 306, 339 Penn pouch, 424t, 426, 426f
clonality in, 54 hyperaldosteronism with, 137f, 138 T pouch, 413–414, 414f, 415f, 416f,
EGF in, 51 IGT with, 113–114, 117f, 119 417f, 423t
EGFR in, 52 incidentaloma with, 133–136, 135f U. Miami pouch, 424t
PC, 53 MRI v., 182, 187 UCLA pouch, 422t, 423t
Chimeric antibodies, 97, 97f NSGCT staging with, 601, 603f, 604f, Continuous ambulatory peritoneal dislysis
Chromosome 605f, 611t, 614t (CAPD)
TCC with abnormalities in, 340 partial nephrectomy for, 230–231 RCC with, 174
792 Index

Core Quality of Life Questionnaire Cutaneous urinary diversion (Continued) Cystectomy (Continued)
(QLQ-C30) Duke pouch, 422t, 424t partial, 329
cutaneous urinary diversion influence in, Florida pouch, 422t, 424t radical, 328–329
437–438, 438t gastric pouch, 424t, 426–427, 427f, urethrectomy with, 680
HRQOL measured with, 105 428f, 436–437 Cystogram
Corticotropin-releasing hormone (CRH) ileocecal, 411–416, 412f, 413f, 414f, orthotopic bladder substitution
Cushing’s syndrome with, 132–133 415f, 416f, 417f, 418–420f, 421f, with, 449
CRAd. See Conditionally replication- 422f, 422t, 423–424t Cystoprostatectomy
competent oncolytic adenovirus Indiana pouch, 416–421, 422t, urethrectomy with, 686–688, 687f, 688f,
CRH. See Corticotropin-releasing hormone 423–424t, 425f, 436 689f, 690f, 691f, 692f, 693f
Cryoablation Kock pouch, 411–413, 412f, 413f, 423t, pelvic dissection in, 686, 687, 687f,
renal tumor ablation with, 207–208, 208t 436 688f
Cryosurgery LeBag, 422t perineal dissection in, 687–688, 689f,
adrenal tumors with, 149 Mainz pouch, 414–416, 418–420f, 423t, 690f, 691f, 692f, 693f
Cryotherapy 436 Cytokine therapy
ED with, 118 Penn pouch, 424t, 426, 426f CSF with, 92, 93, 93t
IGT with, 117–118, 118f right colon, 416–427, 422t, 423–424t, IFN with, 88–89, 89t, 90t, 91–92, 92f
MR image of, 118f 425f, 426f, 427f, 428f, 436–437 IL2 with, 88–89, 89t, 90t, 91, 91f, 92, 92f
outcomes of, 118 T pouch, 413–414, 414f, 415f, 416f, immunotherapy with, 88–92, 89t, 90t,
procedure of, 118 417f, 423t 91f, 92f
Cryptorchid testes U. Miami pouch, 424t intravesical interferon therapy with, 92
seminoma with, 591 UCLA pouch, 422t, 423t MHC with, 88, 89t
CSCT. See Classic Sertoli cell tumors continent v. noncontinent, 399 RCC with, 88, 89–92, 90t
CSF. See Colony-stimulating factor intraoperative pouch testing for, 411 superficial bladder cancer treated with, 92
CT. See Carcinoid tumors; Computed laparoscopic ileal conduit for, 438, 439t Cytology
tomography nasogastric tube for, 400 TCC prognostic factors with, 320–321
CT-guided brachytherapy (CTBT) noncontinent, 400–406, 401f, 402f, 403f, DNA ploidy in, 321
IGT with, 117 404f, 405f urine in, 320–321
CTBT. See CT-guided brachytherapy colocolostomy in, 405f Cytoreductive nephrectomy, 247–248
CTV. See Clinical target volume colonic conduits for, 404–406, 405f
Cushing’s syndrome cutaneous pyelostomy for, 400, 401f Denonvilliers’ fascia
ACTH with, 132–133 cutaneous ureterostomy for, 400–401, Radical cystectomy with, 279f, 377, 378f,
adrenal carcinoma with, 136 402f 380, 380f
adrenal tumors and, 131, 132–133, 133t, ileal conduit for, 401–403, 403f, 404f radical prostatectomy incision of, 521f
134f ileocecal conduit for, 405 RRP incision in, 529
adrenalectomy for, 141, 142t jejunal conduit for, 404 wide-field dissection RRP with, 530
CRH with, 132–133 rectus fascia incision in, 404f Denys-Drash syndrome
description of, 132, 155 renal pelvis incision in, 401f Wilms’ tumor with, 753, 758, 759t
diagnosis of, 132, 134f, 154t, 155 sigmoid colon conduit for, 406 DHEA
LCCSCT with, 623 technique, 400, 401, 401f, 402f, 403, adrenal carcinoma with, 136
manifestations of, 133t 403f, 404f, 405, 405f, 406 Digital rectal exam (DRE)
Cutaneous horn transverse colon conduit for, 404–406, PC staging with, 549
penis carcinoma with, 712 405f RPP patient selection with, 529
Cutaneous pyelostomy novel techniques for, 438–439, 439t US-guided prostate biopsy with, 122
noncontinent cutaneous urinary diversion postoperative care for, 400 Dioxin
with, 400, 401f preoperative preparation for, 399–400 retroperitoneal tumors from, 652
technique for, 400, 401f quality of life with, 437–438, 438t Direct ureterointestinal anstomoses,
Cutaneous ureterostomy rectal bladder urinary diversion for, 427–430, 429f
noncontinent cutaneous urinary diversion 406–407, 407f, 408f DLI. See Donor T-cells
with, 400–401, 402f ureterointestinal anastomoses with, Docetaxel
technique for, 401, 402f 427–430, 429f, 430f, 431f bladder preservation with, 391t
Cutaneous urinary diversion Cycle kinase inhibitor (CDKI), 9, 11 genitourinary cancer treatment with, 66f,
complications with, 430–437, 433t, 435t Cyclophosphamide 67
continent, 406–427, 407f, 408f, 409f, genitourinary cancer treatment with, Donor T-cells (DLI)
410f, 412f, 413f, 414f, 415f, 416f, 68 HCT with, 96f
417f, 418f, 419f, 420f, 421f, 422f, Cyclosporine (CSA) Dorsal venous complex
422t, 423–424t, 425f, 426f, 427f HCT with, 93 radical prostatectomy with, 516–519,
surgical technique for, 406 Cyproterone acetate 518f, 519f, 520f, 521f
continent catheterizable urinary androgen deprivation therapy with, Dose volume histograms (DVH)
diversions for, 407–409 495 brachytherapy and, 120
continent mechanism in, 409–411, 410f Cystectomy. See also Partial cystectomy; radiation oncology with, 42, 42f
continent reservoirs for, 411–427, 412f, Radical cystectomy Doxorubicin
413f, 414f, 415f, 416f, 417f, orthotopic bladder substitution with, bladder preservation with, 391t
418–420f, 421f, 422f, 422t, 445–446, 445f, 446f, 453f bladder TCC with, 324
423–424t, 425f, 426f, 427f, 428f, TCC bladder with, 328–329, 329t genitourinary cancer treatment with, 62f,
436–437 indications for, 328, 329t 63–64
Index 793

DRE. See Digital rectal exam Etoposide Fibrin gradation products (FDP)
Duke pouch genitourinary cancer treatment with, 69, 69f bladder cancer diagnosis with, 305
complications with, 435t European Organization for Research and Fibrosarcoma
continent colon pouches compared to, 422t Treatment of Cancer (EORTC) malignant lesions of, 653t, 656
continent reservoirs compared to, 424t androgen deprivation therapy and, Fine needle aspiration (FNA)
reoperation rate with, 435t 496–497, 497f, 501, 501t, 506, 507 RCC with, 197–198
urinary diversion with, 422t, 424t, 435t HRQOL measured with, 105 FISH. See Fluorescent in situ
DVH. See Dose volume histograms Expanded Prostate Cancer hybridization
Dyspepsia Index-Composite (EPIC) 5-fluorouracil (5-FU)
orthotopic bladder substitution with, 453t HRQOL measured with, 105 Bladder preservation with, 391t
Dysplasia External beam radiation therapy (EBRT) 5-FU. See 5-fluorouracil
urethral cancer pathology with, 673–674 ADT and, 479, 480, 482–483, 486, 487, Flap valve
488 urinary diversion approach of, 409
Eastern Cooperative Oncology Group patient androgen deprivation therapy for, 494, Flexible cystoscope
performance status (ECOG PS) 496–498, 499, 500, 501, 502 TUR equipment including, 360
RCC staging with, 186–187, 186t, BRFS through, 479–482, 480t, 481t Florida pouch
260–261 CT scan with, 478, 483, 484, 484f complications with, 435t
EBRT. See External beam radiation therapy 3DCRT with, 478–479, 478f, 482, 489 continent colon pouches compared to,
ECE. See Extracapsular extension dose escalation with, 481–482, 481t 422t
ECM. See Extracellular matrix HCT with, 95t continent reservoirs compared to, 424t
ECOG PS. See Eastern Cooperative historic perspective of, 478, 478f reoperation rate with, 435t
Oncology Group patient HRQOL with, 107, 108, 108t urinary diversion with, 422t, 424t, 435t
performance status IMRT with, 478 Fluorescent in situ hybridization (FISH)
ED. See Erectile dysfunction interstitial brachytherapy with, 483–489, bladder cancer diagnosis with, 305
EGCT. See Extragonadal germ cell tumors 484f, 484t, 485t, 487t, 489t TCC prognostic markers with, 321
EGF. See Epidermal growth factor BRFS following, 485 Fluoropyrimidines
EGFR. See Epidermal growth factor receptor historic perspective on, 483 genitourinary cancer treatment with,
18-fluoro-2-d3oxyglucose (FDG) PPB technique in, 483–484, 484f, 70–71
bladder cancer diagnosis with, 306, 339 486–488, 487t Fluoroscopic cystoscopy
RCC diagnosis with, 183, 188–189 TPB technique in, 484–485 TUR with, 365
Endopelvic fascia MR with, 478, 483 Flutamide
radical prostatectomy incision of, penis carcinoma with, 704, 705t androgen deprivation therapy with, 495
515–516, 517f, 518f prostate adenocarcinoma treated with, Focused ultrasound surgery (FUS)
EORTC. See European Organization for 477–489, 478f, 479t, 480t, 481t, IGT with, 114, 116, 122
Research and Treatment of Cancer 482f, 484f, 484t, 485t, 487t, 489t Follicle stimulating hormone (FSH)
EPIC. See Expanded Prostate Cancer Index- PSA levels with, 486–488, 487t androgen deprivation therapy for, 495,
Composite radiation oncology with, 39–40, 40f 500t
Epidermal growth factor (EGF) RMS treated with, 773–774 Food and Drug Administration (FDA)
tumor cell biology and, 51 seminal vesicles treatment options with, IL-2 approval by, 88
Epidermal growth factor receptor (EGFR) 558, 560f valrubicin approval by, 324
tumor cell biology and, 52 T1-2 PC in, 477, 482–483 Fossa navicularis cancer, 675, 677t
Epidermoid cysts technique of, 478–479, 479t FSH. See Follicle stimulating hormone
nongerm cell tumors of, 618t, 627–628, CTV with, 478–479, 479t Functional Assessment of Cancer Therapy
627f, 628f GTV with, 478–479, 479t (FACT)
testicular tumors in pediatrics, 781–782 PTV with, 478–479, 479t cutaneous urinary diversion influence in,
Epigenetic effects Extracapsular extension (ECE) 438t
HAT in, 3–4 IGT influence on, 117 HRQOL measured with, 105
HDAC in, 4 PC staging with, 461
methylation in, 4 Extracellular matrix (ECM), 11, 12f, 13f G-CSF. See Granulocyte colony-stimulating
RNAi in, 4 Extragonadal germ cell tumors (EGCT) factor
urologic cancers with, 3–4, 4f seminoma with, 591 Ganglioneuroma
Epipodophyllotoxin Extramustine benign lesions of, 652t
genitourinary cancer treatment with, genitourinary cancer treatment with, 67 Gastric pouch
69–70, 69f complications with, 436–437
Erb-B-2 FACT. See Functional Assessment of Cancer continent reservoirs compared to, 424t
TCC prognosis with, 342–343 Therapy urinary diversion with, 424t, 426–427,
Erectile dysfunction (ED) Familial renal oncocytoma 427f, 428f, 436–437
cryotherapy with, 118 RCC with, 176–177 Gastrointestinal anastomosis (GIA)
Erythrocytosis Fatigue LRN with, 247, 249f
RCC associated with, 174t orthotopic bladder substitution with, 453t GCT. See Testis tumors
Erythroplasia of Queyrat FDA. See Food and Drug Administration Gemcitabine
penis carcinoma with, 710–711 FDG. See 18-fluoro-2-d3oxyglucose bladder preservation with, 391t
Estramustine FDP. See Fibrin gradation products Gene therapy
chemotherapy with, 67 α-fetoprotein (AFP) adenovirus vectors for, 21–22, 21t, 22f, 26t
Estrogen supplementation nongerm cell tumors with, 617, 626 antioncogene therapy for, 30–31, 31f
androgen deprivation therapy with, 495 serum tumor markers of, 568, 596, 598 antisense construct approach to, 30–31
794 Index

Gene therapy (Continued) Genitourinary cancer (Continued) Granulocyte colony-stimulating factor


emerging vectors for, 25 anticancer agent pharmacology for, 54–56 (Continued)
future directions in, 33–34, 33f absorption in, 54 HCT with, 93
gene transfer vectors for, 18–19 distribution in, 54–55 immunotherapy with, 92, 93
liposomal gene transfer for, 21t, 24–25, excretion in, 55–56 Granulocyte-macrophage colony-
24f, 25f, 26t factors modifying, 56 stimulating factor (GM-CSF)
modified vector tropism in, 33–34 metabolism in, 55 cytokine therapy with, 92, 93t
molecular targets of, 25–33, 26t, 27f, transport in, 54–55 HCT with, 93
31f, 32f chemotherapy principles for, 51–74, 57t, immunotherapy with, 92, 93, 93t, 264
direct induction of, 26t, 29–30 59t, 60f, 62f, 66f, 69f, 72f, 73f Granulosa cell tumors
immunogene therapy (ex vivo) in, drug resistance mechanisms with, 56–58, 57t clinical presentation of, 624–625
25–28, 26t, 27f models for overcoming, 56–58, 57t evaluation of, 626
immunogene therapy (in vivo) in, 23f, macro-pharmacokinetic mechanisms management of, 626
26t, 28–29 with, 57t nongerm cell tumors as, 618t, 624–626,
nonviral vectors for, 21t, 24–25, 24f, micro-pharmacokinetic mechanisms 625f, 626f
25f, 26t with, 57t pathologic characteristic of, 625–626,
oncolytic virotherapy for, 30 pharmacodynamic intracellular 625f, 626f
other delivery systems for, 25 mechanisms with, 57t Gross tumor volume (GTV)
poxvirus vectors for, 21t, 22–24, 23f, 26t summary of, 74 radiation oncology with, 41
restricted transgene expression in, 34 treatment efficacy assessment for, 58–59, radiation therapy for adenocarcinoma
retrovirus vectors for, 19–21, 19f, 20f, 59t with, 478–479, 479f
21t, 26t classification by phase in, 59 Growth factor (GF), 12–14, 12f, 13f
ribozyme construct approach to, 30–31, 31f explanatory trial in, 58 GSH. See Glutathione
summary of, 34 pragmatic trial in, 58–59 GSTPi. See Glutathione-S-transferase pi
target cell death with, 26t, 29–30 tumor cell biology and, 51–54 GTV. See Gross tumor volume
targeting vector specificity in, 33–34, 33f cell cycle control in, 52–53 GVHD. See Graft-versus-host disease
transcriptional targeting in, 34 cell proliferation/apoptosis balance in, GVL. See Graft-versus-leukemia
tumor suppressor gene restoration in, 53–54 GVT. See Graft-versus-tumor
31–33, 32f cellular kinetics in, 52–53 Gynecomastia
urologic cancers treated with, 18–34, 19f, clonality in, 54 adrenal carcinoma with, 136
20f, 21t, 22f, 23f, 24f, 25f, 26t, 27f, EGF in, 51
31f, 32f, 33f EGFR in, 52 HA. See Hyaluronic acid
Genitourinary cancer PC, 53 HA stimulating activity (HASA)
agents used against, 59–74, 60f, 62f, 66f, Gerota’s fascia Wilms’ tumor pathology with, 761
69f, 72f, 73f radical nephrectomy with, 224f, 226, 239 HAL. See Hand-assisted laparoscopy
alkylating agents as, 67–68 GF. See Growth factor HAMA. See Human antimouse antibody
anthracycline antibiotics as, 62f, 63–64 GIA. See Gastrointestinal anastomosis Hand-assisted laparoscopy (HAL)
anticancer antibiotics as, 61–64, 62f Giant condyloma acuminatum LRN with, 199
antifolates as, 70 penis carcinoma with, 712 HASA. See HA stimulating activity
antimetabolites as, 70 Gleason score HAT. See Histone acetyl transferases
bleomycin as, 61–63, 62f radical prostatectomy with, 471, 472f HCG. See Human chorionic
camptothecins as, 72–74, 73f seminal vesicles diagnosis of, 556, 556t, 564f gonadotropin
capecitabine as, 71 Glomus jugulare tumors HCT. See Hematopoietic cell
carboplatin as, 60–61, 60f pheochromocytoma with, 139, 148f transplantation
cisplatin as, 59–60, 60f Glucose transporter (GLUT-1) HDAC. See Histone deacetylation
cyclophosphamide as, 68 RCC with, 176 HDR techniques. See High-dose rate
docetaxel as, 66f, 67 GLUT-1. See Glucose transporter techniques
doxorubicin as, 62f, 63–64 Glutathione (GSH), 57 Health-related quality of life (HRQOL)
epipodophyllotoxin as, 69–70, 69f TCC prognosis with, 343 brachytherapy influencing, 107
estramustine as, 67 Glutathione-S-transferase pi (GSTPi) cutaneous urinary diversion with,
etoposide as, 69, 69f inflammation with, 5–6, 6t 437–438, 438t
fluoropyrimidines as, 70–71 GM-CSF. See Granulocyte-macrophage EBRT influencing, 107, 108, 108t
gemcitabine as, 71–72, 72f colony-stimulating factor established instruments for, 104–105
ifosfamide as, 68–69 Gonadal stromal tumors, 782 cancer-specific, 105
irinotecan as, 73–74, 73f Gonadoblastoma, 782 EORTC as, 105
methotrexate as, 70 Goserelin EPIC as, 105
mitomycin as, 62f, 63 androgen deprivation therapy with, 495 FACT as, 105
mitoxantrone as, 62f, 64 Graft-versus-host disease (GVHD) general, 104–105
oxaliplatin as, 60f, 61 HCT with, 93, 95–96, 97 NHP as, 104
paclitaxel as, 65–67, 67f Graft-versus-leukemia (GVL) QLQ-C30 as, 105
platinum complexes as, 59–61, 60f HCT with, 93 QWB as, 104
taxanes as, 65–67, 67f Graft-versus-tumor (GVT) SF-36 as, 104
thiotepa as, 69 HCT with, 93, 96f, 97 SIP as, 104
topotecan as, 73f, 74 Granulocyte colony-stimulating factor UCLA PCI as, 105
tubulin modulation drugs as, 64–65 (G-CSF) urologic malignancy specific, 105
vinca alkaloids as, 64–65 cytokine therapy with, 92 IB influencing, 107–108, 108t
Index 795

Health-related quality of life (Continued) Horseshoe kidney IGCCCG. See International Germ Cell
instrument methodology for, 102–104, seminoma with, 592 Cancer Collaborative
103t, 104t HPC. See Hereditary prostate cancer IGT. See Image-guided therapy
IPSS measuring, 107 HPLC. See High performance liquid IL-2. See Interleukin-2
RP v. WW in, 107 chromatography Ileal conduit
specific urologic malignancies with, HPRC. See Hereditary papillary renal noncontinent cutaneous urinary diversion
105–109, 108t carcinoma with, 401–403, 403f, 404f
bladder cancer in, 108–109 HPV. See Human papillomavirus technique for, 403, 403f, 404f
kidney cancer in, 109 HRPC. See Hormone-refractory PC Ileal segment resection
prostate cancer in, 105–108, 108t hTERT. See High telomerase reverse orthotopic bladder substitution with, 446,
testicular cancer in, 109 transcriptase 447f, 448f
summary of, 109 Human antimouse antibody (HAMA) Ileocecal conduit
urologic oncology issues with, 102–109, immunotherapy with, 97–98, 98t noncontinent cutaneous urinary diversion
103t, 104t, 108t Human chorionic gonadotropin (HCG) with, 405
Hemangiopericytoma serum tumor markers of, 568, 597, 599 ILNR. See Intralobar nephrogenic rest
benign lesions of, 652t Human immunodeficiency virus (HIV) Image-guided therapy (IGT)
malignant lesions of, 653t, 657 seminoma patients with, 592 brachytherapy with, 118–122, 120f,
Hematologic tumors testis tumors and, 637 121f
nongerm cell tumors of, 618t, 633–636, 635f Human papillomavirus (HPV) CT-guided, 119
Hematopoietic cell transplantation (HCT) penis carcinoma with, 711 MR-guided, 114f, 115f, 119–122, 120f,
CSA and, 93 squamous cell carcinoma of penis with, 121f
CSF and, 93 695–696, 710 ultrasound guided, 118–119
DLI with, 96f Humanized antibodies, 97 cryotherapy for, 117–118, 118f
GVHD with, 93, 95–96, 97 Hyaluronic acid (HA) ED with, 118
GVL with, 93 Wilms’ tumor pathology with, 761 MR image of, 118f
GVT with, 93, 96f, 97 Hydraulic valve outcomes of, 118
immunotherapy with, 93–97, 95t, 96f urinary diversion approach of, 409 procedure of, 118
myeloablative condition in, 95, 95t, 96f Hyperaldosteronism CT in, 113–114, 117f, 119
RCC treated with, 95–97, 96f adrenal tumors with, 137–138, 137f, CTBT with, 117
T-cells with, 93, 96, 96f 138f current applications overview of,
XRT conditioning for, 95t adrenalectomy for, 142t 115–116, 115f
Hemorrhage aldosterone secretion with, 138, 138f diagnosis in, 122–123
TUR complications with, 362–363 CT scan for, 137f, 138 ECE minimized with, 117
Hereditary leiomyomatosis renal carcinoma diagnosis of, 154t FUS in, 114, 116, 122
(HLRC) hypernatremia with, 137 history of, 113–114, 114f
RCC with, 177 hypertension with, 137, 137f image processing role of, 116, 117f
Hereditary papillary renal carcinoma (HPRC) hypokalemia with, 137–138 minimal invasiveness of, 113–125, 114f,
RCC with, 176, 197t PRA with, 137, 137f 115f, 117f, 118f, 120f, 121f, 123f,
Hereditary prostate cancer (HPC), 7 RASS with, 137 124f
HIF. See Hypoxia-inducible factor secondary, 138 MR-guided prostate biopsy in, 123–124,
HIFU. See High-intensity focused ultrasound Hypercalcemia 123f, 124f
High-dose rate (HDR) techniques RCC associated with, 174t procedure for, 123–124
prostate adenocarcinoma treated with, Hyperchloremic metabolic acidosis MRBT with, 117
488–489, 489t cutaneous urinary diversion with, 433t MRI in, 113–125, 114f, 115f, 117f, 118f,
radiation oncology with, 41 Hyperkalemia 120f, 121f, 123f, 124f
High-intensity focused ultrasound (HIFU) cutaneous urinary diversion with, 433t MRT in, 114, 115
radiation therapy for adenocarcinoma Hypernatremia, 137 rapid growth in, 113
with, 477 Hypertension summary of, 124–125
renal tumor ablation with, 209–210 hyperaldosteronism with, 137, 137f surgery focused ultrasound in, 122
High performance liquid chromatography RCC associated with, 174t outcome of, 122
(HPLC), 66 Hypochloremic metabolic acidosis procedure for, 122
High telomerase reverse transcriptase cutaneous urinary diversion with, 433t prostate cancer with, 122
(hTERT) Hypokalemia therapy/image integration with, 116
Wilms’ tumor pathology with, 780 cutaneous urinary diversion with, 433t TRUS in, 114, 117, 118, 122
Histone acetyl transferases (HAT) hyperaldosteronism with, 137–138 US-guided prostate biopsy in,
epigenetic effects with, 3–4 Hypoxia-inducible factor (HIF) 122–123
Histone deacetylation (HDAC) RCC with, 176 DRE with, 122
epigenetic effects with, 4 procedure for, 122–123
HIV. See Human immunodeficiency virus IB. See Interstitial brachytherapy ImmunoCyt
HLRC. See Hereditary leiomyomatosis IBD. See Inflammatory bowel disease bladder cancer diagnosis with, 305
renal carcinoma ICU. See Intensive care unit Immunogene therapy
Hormone-refractory PC (HRPC), 57 IFN. See Interferon ex vivo, 25–28, 26t, 27f
Hormone therapy. See Androgen Ifosfamide gene therapy with, 23f, 25–29, 26t, 27f
deprivation therapy bladder preservation with, 391t in vivo, 23f, 26t, 28–29
Horner’s syndrome genitourinary cancer treatment with, Immunosuppression
neuroblastoma with, 742, 748 68–69 patients seminoma with, 590
796 Index

Immunotherapy Intensity modulated radiation therapy Ionizing radiation (IR)


adaptive cellular therapy as, 93, 94f (IMRT) apoptosis in, 45–46
LAK, 93, 94f radiation oncology with, 42–43, 42f, 43f cellular repair response in, 44
TIL, 93, 94f radiation therapy for adenocarcinoma cellular target interacts with, 43–44
basic guidelines for, 88–98, 89t, 90t, 91f, with, 478 cycle and, 45
92f, 93t, 94f, 95t, 96f, 97f, 98t retroperitoneal tumors with, 664 DNA damage accumulation with, 44, 45
BCG as, 92–93, 94f, 324–326, 326t Intensive care unit (ICU) DNA repair following, 45
bladder TCC with, 324–326, 326t radical cystectomy with, 384 induced cell death with, 45–46
CIS with, 325, 326t Interferon (IFN) necrosis in, 45–46
prophylaxis with, 325–326 background of, 88–89, 89t normal tissue protection in, 44–45
bladder TCC with, 324–327 bladder TCC treated with, 326 OER in, 43
bropirimine in, 327 immunotherapy with, 88–89, 89t, 90t, PLDR in, 44
cytokine therapy in, 88–92, 89t, 90t, 91f, 91–92, 92f radiation oncology with, 43–46
92f intravesical interferon therapy with, 92 radiation sensitizers for, 44
CSF with, 92, 93t RCC treatment with, 89, 90t, 91–92, radioprotector agents in, 44–45
IFN with, 88–89, 89t, 90t, 91–92, 92f 263 ROS with, 43–44
IL2 with, 88–89, 89t, 90t, 91, 91f, superficial bladder cancer treated with, 92 SLDR in, 44
92, 92f systemic therapy with, 263 therapeutic ratio in, 44, 45
intravesical interferon therapy Interleukin-2 (IL-2) tumor cell interactions with, 43
with, 92 background of, 88–89, 89t tumoricidal effect enhancement in, 44
MHC with, 88, 89t FDA approval of, 88, 263 IORT. See Intraoperative radiation
RCC with, 88, 89–92, 90t immunotherapy with, 88–89, 89t, 90t, 91, therapy
superficial bladder cancer treated 91f, 92, 92f IPSS. See International Prostate Symptom
with, 92 RCC treatment with, 88, 89, 90t, 91–92, Score
HAMA in, 97–98, 98t 263–264 IR. See Ionizing radiation
HCT as, 93–97, 95t, 96f systemic therapy with, 263–264 Irinotecan
CSA and, 93 International Germ Cell Cancer genitourinary cancer treatment with,
CSF and, 93 Collaborative (IGCCCG) 73–74, 73f
DLI with, 96f NSGCT treatment and, 606, 611t, 615 IVC. See Inferior vena cava
GVHD with, 93, 95–96, 97 testis tumors staging of, 571, 573t IVP. See Intravenous pyelogram
GVL with, 93 International Prostate Symptom Score
GVT with, 93, 96f, 97 (IPSS) Jackson staging system
myeloablative condition in, 95, 95t, 96f HRQOL with, 107 penis carcinoma with, 714, 715t
RCC treated with, 95–97, 96f Interstitial brachytherapy (IB) Penis SCC with, 701t, 702
T-cells with, 93, 96, 96f HRQOL with, 107–108, 108t Jejunal conduit
XRT conditioning for, 95t Intralobar nephrogenic rest (ILNR) noncontinent cutaneous urinary diversion
IFN in, 326 Wilms’ tumor pathology with, 758–759, with, 404
KLH in, 326–327 758f JGCT. See Juvenile granulosa cell tumors
monoclonal antibodies in, 97–98, Intraoperative radiation therapy (IORT) Juvenile granulosa cell tumors (JGCT)
97f, 98t retroperitoneal tumors with, 664 nongerm cell tumors as, 625, 625f, 626
IMRT. See Intensity modulated radiation Intraperitoneal rupture
therapy cutaneous urinary diversion with, 434 Keyhole-limpet hemocyanin (KLH)
Incidentaloma Intravenous pyelogram (IVP) bladder TCC with, 326–327
adrenal tumors as, 133–136, 135f bladder Cancer diagnosis with, 302 Kidney cancer
CT scan in, 133–136, 135f RCC diagnosing with, 178, 178f, 197 HRQOL with, 109
MRI in, 134, 135f upper urinary tract TCC with, 287 KLH. See Keyhole-limpet hemocyanin
ultrasound in, 133 Intravesical interferon therapy Klinefelter’s syndrome
Indiana pouch superficial bladder cancer treated with, 92 LCT with, 619
complications with, 435t, 436 Intravesical therapy Kock pouch
continent reservoirs compared to, 423t bladder TCC with, 322–327, 322t, complications with, 435t, 436
reoperation rate with, 435t 326t continent reservoirs compared to, 423t
technique for, 420–421, 422t, 425f chemotherapy for, 323–324 reoperation rate with, 435t
urinary diversion with, 416–421, 422t, doxorubicin in, 324 technique for, 411–413, 412f, 413f
423–424t, 425f, 435t, 436 MMC in, 323–324, 326t urinary diversion with, 411–413, 412f,
Inferior vena cava (IVC) thiotepa in, 323 413f, 423t, 435t, 436
adrenal anatomy with, 131, 132f valrubicin in, 324
RCC involvement with, 261 combination therapy, 327 L-(methyl)-11C-methionine (LCM)
tumor thrombus with, 203–204, 203t, immunotherapy for, 324–327 bladder cancer diagnosis with, 306
204f, 227f, 228f, 229, 229f BCG in, 324–326, 326t Lactate dehydrogenase (LDH)
Inflammation bropirimine in, 327 serum tumor markers of, 568, 597, 599
GSTPi in, 5–6, 6t IFN in, 326 LAK. See Lymphokine-activated killer cells
molecular biology of, 5–7, 6t KLH in, 326–327 Laparoscopic adrenalectomy
PIN in, 5 rationale for, 322–323, 322t adrenal tumors with, 149, 150–151f
ROS in, 5–6 recommendations in, 327 Laparoscopic ileal conduit
Inflammatory bowel disease (IBD) Intussuscepted nipple valve cutaneous urinary diversion with, 438,
seminoma with, 590 urinary diversion approach of, 409 439t
Index 797

Laparoscopic nephroureterectomy Large cell calcifying Sertoli cell tumors LRN. See Laparoscopic radical nephrectomy
TCC with, 278–279 (LCCSCT) (Continued) LSA. See Lichen sclerosis et atrophicus
Laparoscopic partial nephrectomy pathologic characteristic of, 623–624, Luteinizing hormone (LH)
complications of, 252–283 625f androgen deprivation therapy for, 495,
contraindications for, 249 Peutz-Jeghers syndrome with, 623 500t
financial analysis of, 283 Laser surgery Luteinizing hormone releasing hormone
indications for, 249 advantages/disadvantages with, 364–365 (LHRH)
renal hypothermia with bladder tumor recurrence rate with, 365 androgen deprivation therapy with, 495,
Endocatch II bag in, 250, 254f techniques of, 364 496, 498, 500t, 505–507, 508, 511
renal parenchymal repair in, 253f TUR with, 364–365, 364t, 365f seminal vesicles treatment options with,
results of, 250–252, 255t types of 560f
technique for, 249–250, 250t, 251f, 252f, CO2, 364t Lymph nodes
253f diode, 364t bladder cancer staging with, 311
bulldog clamp in, 251f Ho:Yag, 364, 364t radical prostatectomy with, 472f
methylene blue injection in, 252f KTP, 364, 364t RCC involvement with, 261
Satinsky clamp in, 251f Nd:Yag, 364, 364t, 365, 365f TCC staging with, 274t
Laparoscopic radical nephrectomy (LRN) Laser therapy testis tumors staging of, 571, 572t
approach selection for, 237–238 penis with, 704–705, 705t Lymphadenectomy
complications of, 241, 243t, 244t complications in, 705 avoiding complications with, 730–731
concomitant adrenalectomy with, effectiveness with, 705, 705t classic inguinal type, 732
245–246 surgical technique in, 704–705 considerations in, 730
contra-indications for, 237 LCCSCT. See Large cell calcifying Sertoli modified inguinal type, 729f, 731–732,
cytoreductive, 247–248 cell tumors 732f
financial implications of, 248 LCM. See L-(methyl)-11C-methionine pelvic
follow-up for, 246t LCT. See Leydig cell tumors radical cystectomy with, 373–374, 375f,
indications for, 237 LDH. See Lactate dehydrogenase 376f
larger renal tumors with, 245, 247t LeBag penectomy and, 728, 729f,
oncologic outcome of, 244–245 continent colon pouches compared to, 422t 730–732, 732f
ORN v., 201–203, 202t, 247t urinary diversion with, 422t radical nephrectomy with, 222
blood loss in, 202t Leiomyoma radical prostatectomy with, 515
convalescence time in, 202t, 247t benign lesions of, 652t, 653 RCC with, 205–206, 205f
mean tumor size in, 247t Leiomyosarcoma TCC radical cystectomy with, 345–346
morcellation in, 202–203 malignant lesions of, 653t, 654–655 technique of, 728f, 729f, 731
oncologic adequacy in, 201–202 Leukemia Lymphangiogram
operating room time in, 202t, 247t nongerm cell tumors of, 618t, 636, 636f testis tumors staging with, 570
postoperative complications in, 247t Leuprolide Lymphokine-activated killer cells (LAK)
RCC with, 199–203, 202t, 237–248, 238f, androgen deprivation therapy with, 495 immunotherapy with, 93, 94f
239f, 240f, 241f, 242t, 243t, 244t, Leydig cell tumors (LCT)
245f, 246t, 247t, 248t, 249f CAH v., 621, 621f Macrophage scavenger receptor (MSR-1)
renal vein involvement with, 246–247, clinical presentation of, 618–619 prostate cancer, 5, 6t
248t, 249f evaluation of, 621–622 Magnetic resonance (MR), 113
results of, 241, 242t Klinefelter’s syndrome with, 619 Mainz pouch
specimen extraction with, 241–244, 245f management of, 621–622 complications with, 435t, 436
female, 244, 245f nongerm cell tumors of, 618–622, 618f, continent reservoirs compared to, 423t
male, 244 618t, 619f, 620f, 621f reoperation rate with, 435t
technique for, 200–201, 238–240, 238f, pathologic characteristics of, 619–621, technique for, 414–416, 418–420f
239f, 240f, 241f 620f urinary diversion with, 414–416,
balloon dilation in, 240f Reinke’s crystals in, 619, 621 418–420f, 423t, 435t, 436
clip-applier in, 239f LH. See Luteinizing hormone Malignant fibrous histiocytoma (MFH)
GIA stapler in, 247, 249f LHRH. See Luteinizing hormone releasing malignant lesions of, 652, 653t, 655–656,
HAL, 201 hormone 656f
retroperitoneal, 200–201, 239–240, Lichen sclerosis et atrophicus (LSA) Malignant lesions
240f, 241f Penis carcinoma with, 710 fibrosarcoma as, 653t, 656
transperitoneal, 200, 238–239, 238f, Lipomas leiomyosarcoma as, 653t, 654–655
239f benign lesions of, 652–653, 652t liposarcoma as, 653–654, 653t, 654f, 655f
three approaches to, 199 Liposarcoma malignant fibrous histiocytoma as, 653t,
HAL, 199 malignant lesions of, 653–654, 653t, 654f, 655–656, 656f
retroperitoneal, 199 655f malignant hemangiopericytoma as,
transperitoneal, 199 Liposomal gene transfer 653t, 657
Laparoscopic renal hypothermia, 250, applications of, 21t, 25 retroperitoneal tumors with, 653–657,
254f attributes in, 21t, 25 653t, 654f, 655f, 656f
Large cell calcifying Sertoli cell tumors gene delivery in, 24–25, 24f, 25f rhabdomyosarcoma as, 653t, 656–657
(LCCSCT) gene expression in, 25 Malignant lymphoma (ML)
Carney’s syndrome with, 623–624 gene therapy with, 21t, 24–25, 24f, 25f, clinical presentation of, 633–634
Cushing’s syndrome with, 623 26t evaluation of, 634–636
nongerm cell tumors as, 622, 623–624 vector production in, 24 management of, 634–636
798 Index

Malignant lymphoma (ML) (Continued) Mohs’ micrographic surgery for, (Continued) MR imaging (MRI) (Continued)
nongerm cell tumors of, 618t, fresh tissue technique in, 704 renal tumor ablation with, 208, 209
633–636, 635f technique in, 704 retroperitoneal tumors diagnosis with,
pathologic characteristics of, 634, 635f Molecular biology 658
Malignant mesothelioma (MM) aging/telomerase in, 16 RMS evaluation with, 772–774
clinical presentation of, 629–630, 630f aptamers in, 16–17 Seminal vesicles diagnosis with, 557
evaluation of, 631 cancer genes in, 8–10 squamous cell carcinoma of penis, 696
management of, 631 DNA methylation, 9 TCC diagnosis with, 271–272
nongerm cell tumors of, 629–631, DNA repair, 8 temperature sensitivity of, 116
630f, 631f oncogenes, 8 Wilms’ tumor evaluation with, 756
pathologic characteristics of, suppressor, 8 MR therapy (MRT)
630–631, 631f viral, 8–9 IGT with, 114, 115
Massachusetts General Hospital (MGH), carcinogens in, 4–5 MRBT. See MR-guided brachytherapy
392, 393 cell cycle in, 11–12, 12f MRI. See MR imaging
MDCT. See Multidetector spiral computed cell signaling in, 12f, 14–15, 14f MRT. See MR therapy
tomography CGH in, 8 MSR-1. See Macrophage scavenger receptor
MDR. See Multidrug resistance epigenetic effects in, 3–4, 4f Multidetector spiral computed tomography
MEA syndrome. See Multiendocrine familial cancer in, 7–8 (MDCT)
adenopathy syndrome inflammation in, 5–7, 6t renal diagnosing with, 179
Medroxyprogesterone microarrays in, 10–11 Multidrug resistance (MDR)
androgen deprivation therapy with, 495 proteomics in, 10–11 TCC prognosis with, 343
Megestrol SKY in, 8 Multiendocrine adenopathy syndrome
androgen deprivation therapy stromal epithelial interactions in, (MEA syndrome)
with, 495 15–16, 15f pheochromocytoma with, 139
Metabolic acidosis tumor cell heterogeneity in, 16–17, 17f MVAC. See Methotrexate, vinblastine,
orthotopic bladder substitution with, 450, urologic cancers in, 3–17, 4f, 6t, 10f, 12f, adriamycin, cyclophosphamide
453t 13f, 14f, 15f, 17f MVD. See Microvessel density
Metaiodobenzylguanidine (MIBG) Monoclonal antibodies Myeloablative condition
pheochromocytoma scanned for, 139, 155 immunotherapy with, 97–98, 97f, 98t HCT with, 95, 95t, 96f
Metaplasia Montsouris technique Myelolipoma
urethral cancer pathology with, 673 laparoscopic radical prostatectomy with, benign lesions of, 652t, 653
Metastatic tumors 537
nongerm cell tumors as, 618t, 631–632, Morcellation NAD. See Nicotinamide-adenine
632f ORN v. LRN with, 202–203 dinucleotide
Methotrexate MR. See Magnetic resonance National Bladder Cancer Collaborative
bladder preservation with, 391, 391t MR-guided brachytherapy (MRBT) Group, 303
genitourinary cancer treatment with, 70 IGT with, 114f, 115f, 117, 119–122, 120f, National Wilms’ Tumor Study Group
Methotrexate, vinblastine, adriamycin, 121f (NWTSG), 753, 756, 758,
cyclophosphamide (MVAC) outcome of, 120–122 762–764
TCC chemotherapy treatment with, 281 patient selection for, 119 Nausea
TCC prognosis with, 343 procedure for, 114f, 115f, 119–120, 120f, orthotopic bladder substitution with, 453t
Methylation 121f Nephrectomy
epigenetic effects with, 4 TURP and, 119 adrenalectomy with, 146, 146f
MFH. See Malignant fibrous histiocytoma MR imaging (MRI) cytoreductive, 247–248
MGH. See Massachusetts General Hospital bladder cancer diagnosis with, 302, 304, laparoscopic partial
MIBG. See Metaiodobenzylguanidine 306–307, 339 complications of, 252–283
Microvessel density (MVD) CT v., 182, 187 contraindications for, 249
RCC with, 175 current applications overview of, financial analysis of, 283
TCC prognosis with, 341–342 115–116, 115f indications for, 249
Mitomycin C (MMC) FUS guided by, 116 renal hypothermia with, 250, 254f
bladder preservation with, 391t history of, 113–114, 114f renal parenchymal repair in, 253f
bladder TCC with, 323–324, 326t IGT with, 113–125, 114f, 115f, 117f, results of, 250–252, 255t
genitourinary cancer treatment with, 118f, 120f, 121f, 123f, 124f technique for, 249–250, 250t, 251f,
62f, 63 incidentaloma with, 134, 135f 252f, 253f
Mitotane laser therapy with, 116 laparoscopic radical
adrenal carcinoma with, 136 NSGCT staging with, 600 approach selection for, 237–238
Mitoxantrone PC staging with, 461 complications of, 241, 243t, 244t
genitourinary cancer treatment with, 62f, penis carcinoma with, 714–717 concomitant adrenalectomy with,
64 pheochromocytoma with, 139, 141f, 143f, 245–246
ML. See Malignant lymphoma 148f contra-indications for, 237
MM. See Malignant mesothelioma prostatectomy assessed with, 466 financial implications of, 248
MMC. See Mitomycin C radiation therapy for adenocarcinoma follow-up for, 246t
Mohs’ micrographic surgery for, 704, 723 with, 478, 483 indications for, 237
complications in, 704 radical nephrectomy with, 218 larger renal tumors with, 245, 247t
effectiveness of, 704 RCC diagnosing with, 182, 183f, 197 oncologic outcome of, 244–245
fixed tissue technique in, 704 RCC staging with, 186f, 187–188 ORN v., 201–203, 202t, 247t
Index 799

Nephrectomy (Continued) Nephrectomy (Continued) Nongerm cell tumors (Continued)


RCC with, 199–203, 202t, 237–248, partial version of, 206–207 leukemia as, 618t, 636, 636f
238f, 239f, 240f, 241f, 242t, 243t, surgical margin in, 207 Leydig cell tumors as, 618–622, 618f,
244t, 245f, 246t, 247t, 248t, 249f technique in, 207 618t, 619f, 620f, 621f
renal vein involvement with, 246–247, Nephroblastoma. See Wilms’ tumor CAH v., 621, 621f
248t, 249f Nephroureterectomy clinical presentation of, 618–619
results of, 241, 242t laparoscopic, 278–279, 280t evaluation of, 621–622
specimen extraction with, 241–244, laparoscopic v. open, 280t Klinefelter’s syndrome with, 619
245f open, 277–278, 280t management of, 621–622
technique for, 200–201, 238–240, 238f, TCC with, 277–279, 280t pathologic characteristics of, 619–621,
239f, 240f, 241f Nerve preservation 620f
three approaches to, 199 orthotopic bladder substitution with, Reinke’s crystals in, 619, 621
open radical, 198–199, 199f, 201–203, 202t 445–446, 445f, 446f malignant lymphoma as, 618t, 633–636,
hilar vessel division in, 199f Nesbit technique 635f
technique for, 198–199, 199f orthotopic bladder substitution with, 449f clinical presentation of, 633–634
ORN v. LRN, 201–203, 202t Neuroblastoma evaluation of, 634–636
blood loss in, 202t classification of, 741–742 management of, 634–636
convalescence time in, 202t diagnostic evaluation of, 743–744, 743t, pathologic characteristics of, 634, 635f
morcellation in, 202–203 744f, 745f malignant mesothelioma as, 629–631,
oncologic adequacy in, 201–202 etiology of, 739–740 630f, 631f
operating room time in, 202t future considerations with, 751–752 clinical presentation of, 629–630, 630f
partial, 230–235, 233f, 234f Horner’s syndrome with, 742, 748 evaluation of, 631
AML with, 235 incidence of, 740 management of, 631
CT for, 230–231 pathology of, 740–742, 740f, 741f, 742f pathologic characteristics of, 630–631,
segmental polar, 232, 233f presentation of, 742–743 631f
simple enucleation, 235 prognostic considerations with, 744, 747t metastatic tumors of, 618t, 631–632, 632f
in situ, 231 staging of, 744, 746t miscellaneous tumors of, 627–636, 627f,
transverse resection, 234–235, 234f treatment for, 744–750 628f, 629f, 630f, 631f, 632f, 633f,
wedge resection, 232–234, 234f Neurofibromatosis 634f, 635f, 636f
radical laparoscopic pheochromocytoma with, 139, 140t mixed sex cord/gonadal stromal tumors
HAL, 199 Neuromyopathy as, 626–637, 627f, 628f, 629f, 630f,
ORN v., 201–203, 202t RCC associated with, 174t 631f, 632f, 633f, 634f, 635f, 636f
RCC with, 199–203, 202t Neuron-specific enolase (NSE) ovarian surface epithelial type tumors
retroperitoneal, 199, 200–201 serum tumor markers of, 568 of, 637
technique for, 200–201 Wilms’ tumor pathology with, 761 plasmacytoma as, 618t, 636, 636f
three approaches to, 199 Neurovascular bundles Rete testis carcinoma as, 628–629, 629f
transperitoneal, 199, 200 laparoscopic radical prostatectomy clinical presentation of, 628
radical type, 218–230, 219f, 220f, 221f, dissection of, 538 evaluation of, 628–629
222f, 223f, 224f, 225f, 226f, 227f, radical prostatectomy with separation of, management of, 628–629
228f, 229f, 230t 519, 521f pathologic characteristics of, 628, 629f
anterior subcostal transperitoneal NHP. See Nottingham health profile Sertoli cell tumors as, 618t, 622–624,
incision in, 220, 220f, 221f NHT with radiation therapy in 622f, 623f, 624f, 625f
bilateral subcostal incision in, 220 androgen deprivation therapy for, 495–496 clinical presentation of, 622
CT scan for, 218 Nicotinamide-adenine dinucleotide evaluation of, 624
diaphragmatic incision in, 220 (NAD), 11 management of, 624
evaluation for, 218–219 Nilutamide pathologic characteristics of, 622–624,
Gerota’s fascia in, 224f, 226, 239 androgen deprivation therapy with, 495 622f, 623f, 624f, 625f
indications for, 218–219 NMP-22. See Nuclear matrix protein sex cord/stromal origin with, 618–626,
IVC with, 222–226, 227f, 228f Nongerm cell tumors 618f, 618t, 619f, 620f, 621f, 622f,
left side, 220–222, 224f, 225f acquired immunodeficiency syndrome 623f, 624f, 625f, 626f
lymphadenectomy with, 222 with, 637 testis tumors as, 617–637, 618f, 618t,
MRI for, 218 adenomatoid tumor of epididymis as, 619f, 620f, 621f, 622f, 623f, 624f,
renal vein in, 224f 618t, 633, 633f, 634f 625f, 626f, 627f, 628f, 629f, 630f,
right side, 222, 225f carcinoid tumors of, 618t, 632–633, 633f 631f, 632f, 633f, 634f, 635f, 636f
ring retractor with, 224f epidermoid cysts as, 618t, 627–628, 627f, Nonseminomatous germ cell tumors
suprahepatic vena cava with, 226–230, 628f (NSGCT)
228f, 229f generalized stroma tumors of, 636–637 clinical staging of, 598–600, 601f, 602f,
surgical anatomy with, 219, 219f granulosa cell tumors as, 618t, 624–626, 603f, 604f, 604t, 605t
surgical incisions in, 219–220, 220f, 625f, 626f CT scan for, 600, 602f, 603f, 604f,
221f, 222f clinical presentation of, 624–625 610t, 613t
TEE for, 218 evaluation of, 626 designations in, 600, 604t, 605t
thoracoabdominal approach in, 220, management of, 626 imaging studies for, 600, 601f, 602f,
222f, 223f pathologic characteristic of, 625–626, 603f, 604f
thrombectomy with, 227f 625f, 626f MRI for, 600
RCC with, 206–207 hematologic tumors of, 618t, 633–636, 635f PET scan for, 600
advanced disease with, 206 HIV and, 637 serum markers for, 598
800 Index

Nonseminomatous germ cell tumors Oncolytic virotherapy Orthotopic bladder substitution (Continued)
(NSGCT) (Continued) gene therapy with, 30 heartburn in, 453t
diagnosis of, 597, 598f, 599f, 600f Open nephroureterectomy Laplace’s law in, 450
ultrasound for, 597, 598f, 599f, 600f TCC with, 277–278 late period, 449–450
pathology of, 598–600, 600t Open radical nephrectomy (ORN) metabolic acidosis in, 450, 453t
AJCC classifications of, 599, 604 LRN v., 201–203, 202t metabolic acidosis symptoms in, 453t
radical v. partial orchiectomy for, 598 blood loss in, 202t metabolic management in, 450–452,
serum tumor markers of, 568 convalescence time in, 202t 453t
AFP, 568, 597, 599 mean tumor size in, 247t nausea in, 453t
HCG, 568, 597, 599 morcellation in, 202–203 sitting in, 449
LDH, 568, 597, 599 oncologic adequacy in, 201–202 ultrasound in, 449
NSE, 568 operating room time in, 202t vomiting in, 453t
NSGCT, 568 postoperative complications in, 247t weight loss in, 453t
PLAP, 568, 597 RCC with, 198–199, 199f, 201–203, 202t, preoperative patient selection for,
summary of, 615 247t 443–444
treatment by stage for, 607f, 608–615, Orchiectomy anastomotic margin biopsy in, 444
609f, 610f, 612f, 613f, 614t androgen deprivation therapy with, bowel function in, 444
stage I, 609–611, 612f, 613f 494–495 compliance in, 443
stage IIA/IIB, 611–615, 612f, ORN. See Open radical nephrectomy continence in, 444
614t Orthotopic bladder substitution general preparation for, 444
stage IIC/III, 607, 609f, 610f, 612f, long-term follow-up for, 452–454, 454t hepatic function in, 444
613f, 615 blood tests in, 454t mental capacity in, 443
treatment tools for, 601–608, 607f, 608f, body weight in, 454t renal function in, 443–444
609f, 610f, 611t bone scan in, 454t urethral recurrence in, 444
adjuvant chemotherapy as, 606 chest x-ray in, 454t Oxaliplatin
BEP as, 606 clinical examination in, 454t genitourinary cancer treatment with,
high-stage disease with chemotherapy CT scan in, 454t 60f, 61
as, 606–607 folic acid in, 454t Oxygen enhancement ratio (OER), 43
IGCCCG with, 606, 611t, 615 IVU in, 454t
observation as, 601, 611t postvoid residual in, 454t P53 gene
postchemotherapy RPLND as, renal ultrasound in, 454t TCC prognosis with, 342
604–606, 607f urethral wash cytology in, 454t Paclitaxel
primary RPLND as, 601–604, 607f, urine culture in, 454t bladder preservation with, 391, 391t
608f, 609f, 610f, 611t males/females with, 443–454, 445f, 446f, genitourinary cancer treatment with,
primary shot course chemotherapy as, 447f, 448f, 449f, 450f, 451f, 452f, 65–67, 67f
606 453f, 453t, 454t Papillary transitional cell carcinoma
PVB as, 606 operative technique for, 444–448, 445f, urethral cancer pathology with, 674
salvage chemotherapy as, 607–608 446f, 447f, 448f, 449f, 450f, 451f, Papillary urothelial neoplasms of low
second-line chemotherapy as, 607–608 452f, 453f malignant potential (PUNLMP)
Nonviral vectors atraumatic urethra dissection as, 446 TCC staging with, 274
applications of, 21t, 25 bipolar electrocautery in, 446 Parastomal hernias
attributes in, 21t, 25 bladder construction as, 447–448, 450f, cutaneous urinary diversion with, 431
gene delivery in, 24–25, 24f, 25f 451f, 453f Paratesticular tumors
gene expression in, 25 cystectomy as, 445–446, 445f, 446f, retroperitoneal tumors with, 667
gene therapy with, 21t, 24–25, 24f, 25f, 453f Partial cystectomy, 368–369
26t ileal segment resection as, 446, 447f, Particle beam therapy
vector production in, 24 448f prostate adenocarcinoma treated with,
Nottingham health profile (NHP) mesenteric window close in, 448f 489
HRQOL measured with, 104 nerve preservation in, 445–446, 445f, PC. See Prostate cancer
NSE. See Neuron-specific enolase 446f PCNA. See Proliferating cell nuclear
NSGCT. See Nonseminomatous germ cell Nesbit technique in, 449f antigen
tumors orthotopic ileal bladder substitute as, PCSM. See Prostate cancer-specific
Nuclear matrix protein (NMP-22) 446, 447f, 448f mortality
bladder cancer diagnosis with, 305 U-shaped distal ileum in, 450f PDGF. See Platelet-derived growth factor
ureteroileal anastomosis as, 446–447, Pediatric testicular tumors, 780–785
Obturator nerve reflex 449f, 450f Penectomy
TUR complications with, 363 ureters in, 446 partial penis, 703, 723–725, 724f
OER. See Oxygen enhancement ratio urethral anastomosis as, 447–448, 450f, complications in, 703
Oligospermia 451f, 453f impact of, 704
adrenal carcinoma with, 136 patient assessment for, 443–444 results in, 703
Oncocytoma postoperative management for, 448–452, surgical technique in, 703
familial renal, 176–177 453t total penis, 703–704, 725–726, 725f
RCC mimicked by, 181f anorexia in, 453t complications in, 704
RCC risk factor of, 196t cystogram for, 449 impact of, 704
Oncologic adaquacy dyspepsia in, 453t surgical technique in, 703
ORN v. LRN with, 201–202 fatigue in, 453t Penile urethra cancer, 675, 677t
Index 801

Penis Pheochromocytoma Prostate cancer (PC)


brachytherapy for, 705–706 cardiomyopathy with, 139 cell proliferation/apoptosis balance in, 53
external beam radiation for, 706, 707t CT scan for, 139 detection of, 460f, 461f, 462f, 463f,
inguinal node management with, glomus jugulare tumors with, 139, 148f 549–463
706–707, 707f identifying, 138, 138f, 154t GSTPi in, 5–6, 6t
invasive carcinoma of, 710–718, 715t, 716t MEA syndromes with, 139 HPC and, 7
Buschke-Löwenstein tumor in, 712, MIBG scan for, 139, 155 HRQOL with, 105–108, 108t
723 MRI scan for, 139, 141f, 143f, 148f brachytherapy influencing, 107
laser therapy for, 704–705, 705t neurofibromatosis with, 139, 140t EBRT influencing, 107, 108, 108t
complications in, 705 Sturge-Weber syndrome with, 139 IB influencing, 107–108, 108t
lymphadenectomy for, 728f, 729f, symptoms of, 140t IPSS measuring, 107
730–732, 732f von Hippel-Landau with, 139, 140t RP v. WW in, 107
Mohs’ micrographic surgery for, 704, 723 PIN. See Prostatic interepithelial neoplasia inflammation with, 5–7, 6t
partial penectomy for, 703 Placental alkaline phosphatase (PLAP) localized, 547–552
radiation therapy for, 705 serum tumor markers of, 568, 597 operative complication with, 548–449
SCC of, 695–697, 697t, 699–701, 701t, Planning target volume (PTV) patient selection with, 548
702, 702t radiation oncology with, 41, 42f postoperative complication with,
summary of carcinoma of, 707 radiation therapy for adenocarcinoma 449–552
superficial carcinoma of, 699–707, 700f, with, 478–479, 479f surgical approaches to, 547–548
701t, 702t, 705t, 706t, 707f, 707t PLAP. See Placental alkaline phosphatase MR-guided biopsy for, 123–124, 123f,
superficial SCC of, 702–703 Plasma renin activity (PRA) 124f
surgical procedures for, 723–732, 724f, hyperaldosteronism with, 137, 137f procedure in, 123–124
725f, 726f, 727f, 728f, 729f, 732f Plasmacytoma MRBT for, 120f
total penectomy for, 703–704 nongerm cell tumors of, 618t, 636, 636f MSR-1 in, 5, 6t
complications in, 704 Platelet-derived growth factor (PDGF), 9 natural history of, 465–466, 466t
impact of, 704 Platinum complexes PIN in, 5
surgical technique in, 703 carboplatin, 60–61, 60f RNASEL in, 5, 6t
Penn pouch cisplatin, 59–60, 60f ROS in, 5–6
complications with, 435t genitourinary cancer treatment with, staging of, 460f, 461f, 462f, 463f, 549–463
continent reservoirs compared to, 424t 59–61, 60f AJCC system for, 462, 549–461
reoperation rate with, 435t oxaliplatin, 60f, 61 combined modality, 460f, 461f,
urinary diversion with, 424t, 426, 426f, 435t PLDR. See Potentially lethal damage repair 549–461
PEP. See Polyestradiol phosphate PLNR. See Perilobar nephrogenic rest CT scan in, 461
Percutaneous needle aspiration cytology Polyestradiol phosphate (PEP) DRE with, 549
RCC diagnosis with, 189, 189t androgen deprivation therapy with, 506 ECE in, 461
Perilobar nephrogenic rest (PLNR) Positron emission tomography (PET) MRI in, 461
Wilms’ tumor pathology with, 758–759, bladder cancer diagnosis with, 305, 307, PCSM with, 460f, 461f, 462, 463, 549
758f 339 pretreatment nomograms in, 263f,
Permanent prostate brachytherapy (PPB) FDG with, 183, 188–189, 339 461–262
prostate adenocarcinoma treated with, NSGCT staging with, 600 prognostic factors in, 460f, 461f, 462f,
483–484, 484f, 486–488, 487t RCC diagnosing with, 178, 182–183, 463f, 549–463
PET. See Positron emission tomography 188–189, 197 PSA in, 460f, 461f, 462f, 463f,
Peutz-Jeghers syndrome testis tumors staging with, 570 549–463
LCCSCT with, 623 Potentially lethal damage repair (PLDR), 44 radiologic, 461, 462f
Pharmacology of anticancer agents Pouchitis RT with, 460f, 461f, 462f, 463, 463f,
absorption in, 54 cutaneous urinary diversion with, 434 549
distribution in, 54–55 Poxvirus vectors summary for, 262
drug resistance mechanisms with, 56–58, applications of, 21t, 23–24 SVI in, 461
57t attributes in, 21t, 23–24 TRUS in, 461
models for overcoming, 56–58, 57t gene delivery in, 23 TURP with, 549
excretion in, 55–56 gene expression in, 23 surgery focused ultrasound for, 122
factors modifying, 56 gene therapy with, 21t, 22–24, 23f, 26t outcome of, 122
genitourinary cancer and, 54–56 vector production in, 19f, 22–23 procedure for, 122
macro-pharmacokinetic mechanisms PPB. See Permanent prostate brachytherapy surgical treatment complications with,
with, 57t PRA. See Plasma renin activity 547–552
metabolism in, 55 Prepubertal testicular tumours, 780–785 bladder neck contracture, 549
micro-pharmacokinetic mechanisms Pretreatment nomograms early postoperative, 549
with, 57t PC staging with, 263f, 461–262 erectile dysfunction, 451–452
pharmacodynamic intracellular Progesterone supplementation late postoperative, 549–452
mechanisms with, 57t androgen deprivation therapy with, 495 operative, 547–549
transport in, 54–55 Proliferating cell nuclear antigen (PCNA) postoperative, 549–452
Pheochromocytoma RCC with, 175 quality of life as, 452
adrenal tumors with, 138–139, 138f, 140t, Prostate cancer (PC). See also urinary incontinence, 450–451
141f, 142t, 143f, 148f Adenocarcinoma of prostate US-guided biopsy for, 122–123
adrenalectomy for, 142t bulbourethral v., 3, 4f DRE with, 122
APUD with, 139 burnt meat and, 5 procedure for, 122–123
802 Index

Prostate cancer-specific mortality (PCSM) Radiation oncology Radiation therapy (RT) (Continued)
PC staging with, 460f, 461f, 462, 463, 549 CTV in, 41 GTV with, 478–479, 479f
Prostate-specific antigen (PSA) DVH in, 42, 42f HIFU with, 477
external beam radiation therapy with, EBRT in, 39–40, 40f IMRT with, 478, 482
460f, 461f GTV in, 41 MR with, 478, 483
PC staging with, 460f, 461f, 462f, 463f, HDR techniques for, 41 NHT with, 495–496
549–463 IMRT in, 42–43, 42f, 43f PSA levels in, 486–488, 487t
pretreatment nomogram for, 463f ionizing radiation in, 41–46 PSA with, 477–478, 479, 480, 481t,
radiation therapy for adenocarcinoma apoptosis in, 45–46 482f, 485, 485f, 486–488
with, 477–478, 479, 480, 481t, cellular repair response in, 44 PTV with, 478–479, 479f
482f, 485, 485f, 486–488 cellular target interacts with, 43–44 risk stratification with, 478
radical prostatectomy with, 462f cycle and, 45 RTC with, 477
RPP patient selection with, 528 DNA damage accumulation with, RTOG with, 477, 478, 482, 483, 485
seminal vesicles diagnosis of, 555–558, 44, 45 T1-2 PC in, 477, 482–483
556t, 557t, 561, 562, 564f DNA repair following, 45 timing/duration of hormonal therapy
Prostate-specific membrane antigen (PSMA) induced cell death with, 45–46 with, 498
immunotherapy with, 98 necrosis in, 45–46 retroperitoneal tumors with, 664
Prostatic interepithelial neoplasia (PIN) normal tissue protection in, 44–45 RMS treated with, 773–774
prostate cancer with, 5 OER in, 43 seminal vesicles treatment options with,
Prostatic pedicles PLDR in, 44 557–558, 557t
laparoscopic radical prostatectomy radiation oncology with, 43–46 stage I seminoma with, 587–588
dissection of, 538 radiation sensitizers for, 44 Radical cystectomy
radical prostatectomy with transection/ radioprotector agents in, 44–45 abdominal exploration in, 370
control of, 519–522, 522f ROS with, 43–44 adjuvant v. neoadjuvant, 346–347
Prostatic urethra SLDR in, 44 bladder treated with, 368, 369–387, 370f,
CIS only, 328f therapeutic ratio in, 44, 45 371f, 372f, 373f, 375f, 376f, 377f,
cystoprostatectomy for, 328f tumor cell interactions with, 43 378f, 379f, 380f, 381f, 382f, 383f,
ductal, 328f physics of, 39–41, 40f, 41f 384f, 385f, 386f
intravesical therapy for, 328f principles of, 39–48, 40f, 41f, 42f, 43f, 47t bowel mobilization in, 370–372, 372f,
stromal invasion, 328f PTV in, 41, 42f 373f
TCC in, 327–328, 328f summary of, 47–48 chemotherapy with, 346–347
Prostatic urethral cancer, 675–676, 677t treatment planning/delivery for, 41–43, complications of, 346
PSA. See Prostate-specific antigen 42f, 43f Denonvilliers’ fascia in, 279f, 377, 378f,
PSMA. See Prostate-specific membrane Radiation sensitizers, 44 380, 380f
antigen Radiation Therapy Oncology Group discussion of, 385–387
Psoralen and ultraviolet A (PUVA) (RTOG) dorsal venous complex in, 380, 381f, 382f
penis carcinoma photochemotherapy androgen deprivation therapy and, female patient anterior dissection in,
with, 709, 711 495–498, 499, 500, 501t, 502 382–384, 383f, 384f, 385f, 386f
PTV. See Planning target volume bladder preservation and, 391, 391t female pelvic sagittal section in, 386f
Puboprostatic ligaments radiation therapy for adenocarcinoma females with, 345
radical prostatectomy incision of, with, 477, 478, 482, 483, 485 gauze sponge withdrawing in, 376f
515–516, 517f Radiation therapy (RT). See also ICU for, 384
PUNLMP. See Papillary urothelial Brachytherapy iliac artery skeletonizing in, 375f
neoplasms of low malignant males urethral cancer with, 680 incision in, 370, 371f
potential PC staging with, 460f, 461f, 462f, 463, lateral vascular pedicle ligation in,
PUVA. See Psoralen and ultraviolet A 463f, 549 374–375, 377f
PVB. See Cisplatin, Velban and bleomycin penis carcinoma with, 705, 718 lymphadenectomy with, 345–346
PVB as prostate adenocarcinoma treated with, male patient anterior dissection in,
NSGCT treatment with, 606 477–489, 478f, 479t, 480t, 481t, 378–382, 381f, 382f
482f, 484f, 484t, 485t, 487t, 489t males with, 344–345
QLQ-C30. See Core Quality of Life ACD with, 479, 483, 485, 487–488 outcomes of, 346
Questionnaire adjuvant hormonal with, 496–498, 497f overhead pelvic view in, 372f, 373f
Quality of well-being scale (QWB) ADT and, 479, 480, 482–483, 486, 487, overview of, 343–344
HRQOL measured with, 104 488 patient positioning in, 370, 370f
QWB. See Quality of well-being scale androgen deprivation therapy with, pelvic lymphadenectomy with, 373–374,
495–498, 497f 375f, 376f
Radiation oncology ASTRO with, 479, 481 peritoneum incision in, 378f
applications of, 39–48, 40f, 41f, 42f, 43f, biochemical relapse-free survival with, posterior pedicle ligation in, 375–378,
47t 477, 478, 479–481, 480t, 481t, 482, 378f, 379f, 380f
brachytherapy in, 40–41, 42f 482f, 485–489, 485t, 487f, 489f postoperative care for, 384–385
clinical practice of, 46–47, 47t CT scan with, 478, 483, 484, 484f preoperative evaluation for, 344, 369–370
acute/chronic radiation sequelae in, 47 CTV with, 478–479, 479f counseling in, 369–370
adjuvant RT in, 46 3DCRT with, 478–479, 478f, 482, 489 patients over 50 in, 369
definitive RT in, 46 dose escalation with, 481–482, 481t preparation for, 344
palliative RT in, 46 EBRT historic perspective, 478 TCC with, 343–347
RT in, 46–47 EBRT technique for, 478–479, 479f urachal remnant excision in, 371f
Index 803

Radical cystectomy (Continued) Radical prostatectomy (Continued) Radical prostatectomy (Continued)


ureteral dissection in, 372 prostatic pedicles transection/control transperitoneal approach to, 537
vaginal incision in, 383f for, 519–522, 522f transperitoneal approach variants for,
vaginal wall dissection in, 384f, 385f puboprostatic ligaments incision for, 538
vascular pedicle isolation in, 377f 515–516, 517f ureteric injuries from, 540t, 541
Radical orchiectomy summary of, 526 urethra section in, 538
CT scan with, 642, 647, 647f surgical technique for, 515–523, 516f, urinary incontinence from, 541
general considerations for, 641–642 517f, 518f, 519f, 520f, 521f, 522f, vesicoprostatic dissection in, 538
RPLND for low-stage disease with, 523f, 524f, 525f vesicourethral anastomosis in, 538
642–647, 643f, 644f, 645f, 646f urinary continence outcome of, late complications with, 470
left-sided nerve-sparing in, 645–646, 525–526 lymph node metastases with, 472t
646f vesico-urethral anastomosis for, mortality with, 468f
postoperative management in, 646 522–523, 524f, 525f MRI assessment with, 466
right nerve-sparing in, 643–645, 645f, androgen deprivation therapy with, obturator nerve injury from, 470
646f 498–499 outcomes after, 469f, 470–474, 471f, 472f,
technique for, 643–647, 643f, 644f, biopsy Gleason sum with, 471, 472f 473f
645f, 646f blood loss during, 469, 469f patient selection for, 466
RPLND for postchemotherapy disease cancer control with, 469f, 470–473, 471f, PC natural history and, 465–466, 466t
with, 647–649, 647f, 648f, 649f 472f pelvic lymph node dissection with,
indications for, 647–648, 647f CT scan assessment with, 466 467–468
special considerations for, 649, 649f early complications with, 468–470, 468f, pretreatment risk stratification with,
technique of, 648–649, 648f 469f 466–467
RPLND with, 641–649, 643f, 644f, 645f, erectile function after, 474 PSA risk assessment with, 466–467, 470,
646f, 647f, 648f, 649f hospitalization days with, 469f 471f
summary of, 649 intraoperative complications with, PSA survival with, 469f
Radical perineal prostatectomy (RPP), 468–470, 468f, 469f rectal injury from, 469
528–534, 531f, 532f, 534f, 534t laparoscopic, 536–544, 540t, 542t, 543t remote-controlled assisted, 544
complications of, 530–531 anastomotic leaks from, 541 results of, 468
disease control with, 531–532, 531f, 532f anastomotic stenosis from, 542 risk factors with, 472f, 473f
fecal incontinence following, 533 anatomic contraindications for, 537 seminal vesicle involvement with, 472f
modification of, 530 anesthetic contraindications for, treatment rationale, 465
nerve-sparing, 530 536–537 urinary function with, 473, 473f
wide-field dissection, 530 anterior phase in, 537 urinary incontinence after, 473, 473f
patient selection for, 528–529 blood loss from, 539, 540t Radical prostatectomy (RP)
BPLND with, 528 contraindications for, 536–537 HRQOL with, 107
DRE in, 529 difficult cases for, 537 PSA with, 459, 462f
PSA levels in, 528 digestive injuries from, 541 Radiofrequency ablation
potency following, 533 extraperitoneal approach for, 538–539 RCC with, 208–209, 208t
quality of life following, 533–534 history of, 536 RAND Medical Outcomes Study (SF-36)
summary of, 534 impotence from, 541–542, 543t cutaneous urinary diversion influence in,
surgical technique for, 529–530 indications for, 536 437–438, 438t
urinary continence following, 532 insufflation/trocar positioning HRQOL measured with, 104
Radical prostatectomy. See also Radical complications for, 539 Randomized clinical trial (RTC)
perineal prostatectomy intraoperative complications for, 539 radiation therapy for adenocarcinoma
adenocarcinoma of prostate with, main operation steps for, 537–538 with, 477
465–474, 466t, 468t, 469t, 471t, material for, 537 RASS. See Renin-angiotensin-aldosterone
472t, 473t medical preparation for, 537 system
anatomic nerve-sparing retropubic, Montsouris technique in, 537 RCC. See Renal cell carcinoma
514–527, 516f, 517f, 518f, 519f, neurovascular bundles dissection in, Reabsorption syndrome
520f, 521f, 522f, 523f, 524f, 538 TUR complications with, 363–364
525f number of trocars for, 538 Reactive oxygen species (ROS)
bladder neck transection/reconstruction oncological results from, 542–544 inflammation with, 5–6
for, 522, 523f patient installation in, 537 IR with, 43–44
cancer control outcome of, 525 patient positioning complications Rectal bladder urinary diversion
complications of, 526 for, 539 cutaneous urinary diversion with,
Denonvilliers’ fascia incision for, 521f perioperative complications for, 406–407, 407f, 408f
dorsal venous ligation/transection for, 539–542, 540t taenia incision in, 407f
516–519, 518f, 519f, 520f, 521f posterior phase in, 537 technique in, 407, 407f, 408f
endopelvic fascia incision for, 515–516, postoperative complications for, 541 ureterointestinal anastomosis in, 407f
517f, 518f postoperative management for, 538 ureterosigmoidostomy in, 406–407
limited pelvic lymphadenectomy for, prostatic pedicles dissection in, 538 Reinke’s crystals
515 specimen extraction complications LCT with, 619, 621
neurovascular bundies separation for, for, 541 Renal cell carcinoma (RCC)
519, 521f specimen extraction in, 538 active immunotherapy for, 264
patient selection for, 514–515 surgical technique for, 537–538 heat shock protein with, 264
potency outcome of, 526 thromboembolic complications for, 541 VHL gene with, 264
804 Index

Renal cell carcinoma (RCC) (Continued) Renal cell carcinoma (RCC) (Continued) Renal cell carcinoma (RCC) (Continued)
adrenalectomy with, 204–205 hypertension associated with, 174t molecular genetics with, 176–177
advanced, 258–265, 259f, 260f, 262f, 263f immunotherapy for, 88, 89–92, 90t, molecular markers with, 175–176
amyloidosis associated with, 174t 95–97, 96f AgNOR, 175
anemia associated with, 174t incidence rates of, 173 CAIX protein, 176
antiangiogenic for, 264–265 inheritance of, 176–177 HIF, 176
biopsy for, 197–198 BHD in, 176, 197t MVD, 175
bone marrow transplantation for, 265 familial renal oncocytoma in, 176–177 PCNA, 175
cachexia associated with, 174t GLUT-1 in, 176 VHL protein, 176
chemotherapy for, 265 HLRC in, 177 monitoring for recurrence with, 189
classification of, 177–178, 177t, 196, 196t HPRC in, 176, 197t nephrectomy with, 206–207
Bellini’s duct, 177, 177t TGF in, 176 advanced disease with, 206
Benign neoplasms, 196t tuberous sclerosis in, 197t partial type, 206–207
chromophobe cell, 177, 177t, 196, 196t VHL in, 176, 197t surgical margin in, 207
collecting duct, 177, 177t, 196t laparoscopic partial nephrectomy for, technique in, 207
conventional type, 196t 230–235, 233f, 234f neuromyopathy associated with, 174t
metanephric adenoma, 196t AML with, 235 novel therapies for, 264–265
oncocytoma, 196t complications of, 252–283 open radical nephrectomy for, 198–199,
papillary, 177, 177t, 196t contraindications for, 249 199f, 201–203, 202t
renal medullary, 177–178, 177t CT for, 230–231 LRN v., 201–203, 202t
unclassified type, 178, 196t financial analysis of, 283 technique for, 198–199, 199f
clinical presentation with, 174–175, 174t, indications for, 249 paraneoplastic syndromes associated with,
195–196 renal hypothermia with, 250, 254f 174–175, 174t
cytokine therapy for, 88, 89–92, 90t renal parenchymal repair in, 253f passive immunotherapy for, 264
diagnosing of, 173–183, 174t, 177t, 178f, results of, 250–252, 255t TIL with, 264
179f, 180f, 181f, 182f, 182t, 183f, segmental polar, 232, 233f pathology of, 177–178, 177t
197 simple enucleation, 235 patient performance status with, 260
AML in, 179, 180, 180f in situ, 231 prognostic factors for, 210–211, 210t
Bosniak classification in, 182, 182t technique for, 249–250, 250t, 251f, TNM classification with, 210–211,
Bosniak lesions in, 181 252f, 253f 210t
CT for, 179–182, 180f, 181f, 182f, 197 transverse resection, 234–235, 234f radical nephrectomy for, 218–230, 219f,
FDG for, 183, 188–189 wedge resection, 232–234, 234f 220f, 221f, 222f, 223f, 224f, 225f,
imaging evaluation for, 178 laparoscopic radical nephrectomy for, 226f, 227f, 228f, 229f, 230t
IVP for, 178, 178f, 197 199–203, 202t anterior subcostal transperitoneal
MDCT for, 179 approach selection for, 237–238 incision in, 220, 220f, 221f
MRI for, 182, 183f, 197 complications of, 241, 243t, 244t bilateral subcostal incision in, 220
nuclear medicine and, 182–183 concomitant adrenalectomy with, CT scan for, 218
oncocytoma mimicked in, 181f 245–246 diaphragmatic incision in, 220
percutaneous biopsy in, 189, 189t contra-indications for, 237 evaluation for, 218–219
percutaneous needle aspiration cytology cytoreductive, 247–248 Gerota’s fascia in, 224f, 226, 239
in, 189, 189t financial implications of, 248 indications for, 218–219
PET for, 178, 182–183, 188–189, 197 follow-up for, 246t IVC with, 222–226, 227f, 228f
ultrasonography for, 178–179, 179f, HAL, 199, 201 left side, 220–222, 224f, 225f
197 indications for, 237 lymphadenectomy with, 222
disease progression in, 189 larger renal tumors with, 245, 247t MRI for, 218
enteropathy associated with, 174t oncologic outcome of, 244–245 renal vein in, 224f
epidemiology of, 195–196, 258–262, 259f, ORN v., 201–203, 202t, 247t right side, 222, 225f
260f RCC with, 199–203, 202t, 237–248, ring retractor with, 224f
grade with, 259–260 238f, 239f, 240f, 241f, 242t, 243t, suprahepatic vena cava with, 226–230,
histology with, 259–260 244t, 245f, 246t, 247t, 248t, 249f 228f, 229f
IVC involvement with, 261 renal vein involvement with, 246–247, surgical anatomy with, 219, 219f
patient evaluation with, 261 248t, 249f surgical incisions in, 219–220, 220f,
patient performance status with, 260 results of, 241, 242t 221f, 222f
prognosis in, 259 retroperitoneal, 199, 200–201 TEE for, 218
tumor stage in, 259, 259f specimen extraction with, 241–244, 245f thoracoabdominal approach in, 220,
UISS with, 260–261, 260f technique for, 200–201, 238–240, 238f, 222f, 223f
erythrocytosis associated with, 174t 239f, 240f, 241f thrombectomy with, 227f
etiology of, 173–174 three approaches to, 199 renal tumor ablation with, 207–210, 208t
fever associated with, 174t transperitoneal, 199, 200 cryoablation in, 207–208, 208t
FNA for, 197–198 localized, 195–212, 196t, 197t, 199f, 202t, CT with, 208, 209
grade of, 259–260 203t, 204f, 205f, 208f, 210t HIFU, 209–210
HCT for, 95–97, 96f lymphadenectomy with, 205–206, 205f MRI with, 208, 209
hepatic-dysfunction associated with, 174t metastatic, 262–263, 263f radiofrequency, 208–209, 208t
histology of, 259–260 IFN for, 262 retroperitoneal tumors, 665–666, 665t
histopathology of, 196, 196t nephrectomy in, 262–263, 263f risk factors with, 173–174, 196–197, 197t
hypercalcemia associated with, 174t SWOG study of, 262–263 screening for, 211
Index 805

Renal cell carcinoma (RCC) (Continued) Retroperitoneal technique Rhabdomyosarcoma (RMS) (Continued)
SEER reporting of, 173 laparoscopic surgery with, 164, 164f complications with, 776
serum markers with, 175 Retroperitoneal tumors evaluation of, 772–773, 772f
staging of, 183–189, 183f, 184–185t, 186f, adult urinary tract sarcoma and, 665, 665t CT scan for, 772–773
186t, 187f, 188f, 189t, 259 Agent Orange and, 652 MRI for, 772–773
CT in, 186–188, 186f benign lesions with, 652–653, 652t general approach to, 773–774
ECOG PS in, 186–187, 186t, 260–261 ganglioneuroma as, 652t chemotherapy for, 773–774
MRI in, 186f, 187–188 hemangiopericytoma as, 652t radiation therapy for, 773–774
Robson classification in, 183, 183f leiomyoma as, 652t, 653 relapse with, 774
TNM system in, 183–185, 184–185t, lipomas as, 652–653, 652t surgery for, 774
186, 186t, 259, 259f, 260 myelolipoma as, 652t, 653 XRT for, 773–774
UISS in, 186–187, 186t, 259–260, 260f Schwannoma as, 652t histologic subtypes of, 772f
Stauffer’s syndrome associated with, 174t chemotherapy for, 664–665 malignant lesions of, 653t, 656–657
summary of, 212 metastatic disease and, 664–665 molecular biology of, 771–772, 772f
surgery for, 218–235, 219f, 220f, 221f, diagnosis of, 657–658, 657f paratestis treatment for, 775–776
222f, 223f, 224f, 225f, 226f, 227f, CT scan in, 657–658, 657f chemotherapy in, 775
228f, 229f, 230t, 233f, 234f MRI in, 658 outcome for, 775–776
indications for, 198 dioxin and, 652 radiation therapy in, 775
systemic therapy for, 263–264 incidence/etiology of, 651–652 surgery in, 775
IFN in, 263 malignant lesions with, 653–657, 653t, pathology of, 771–772, 772f
IL-2 in, 263–264 654f, 655f, 656f pediatric scrotal mass and,
treatment of, 258–265, 259f, 260f, 262f, fibrosarcoma as, 653t, 656 presentation of, 772–773, 772f
263f leiomyosarcoma as, 653t, 654–655 staging of, 773, 773t
tumor biopsy for, 197–198 liposarcoma as, 653–654, 653t, 654f, summary of, 776
tumor thrombus with, 203–204, 203t, 655f vagina/uterus treatment for, 775
204f, 227f, 228f, 229, 229f malignant fibrous histiocytoma as, 653t, chemotherapy in, 775
Renal hypothermia nephrectomy 655–656, 656f outcome for, 775
Endocatch II bag in, 250, 254f malignant hemangiopericytoma as, radiation therapy in, 775
Renin-angiotensin-aldosterone system 653t, 657 Ring retractor
(RASS) rhabdomyosarcoma as, 653t, 656–657 radical nephrectomy with, 224f
hyperaldosteronism with, 137 paratesticular tumors with, 667 RMS. See Rhabdomyosarcoma
Rete testis carcinoma (RTC) pathology of, 652–657, 652t, 653t, 654f, RNASEL
clinical presentation of, 628 655f, 656f prostate cancer with, 5, 6t
evaluation of, 628–629 prostate with, 666–667 Robson classification in
management of, 628–629 radiation therapy for, 664 RCC staging with, 183, 183f
nongerm cell tumors of, 628–629, 629f IMRT, 664 ROS. See Reactive oxygen species
pathologic characteristics of, 628, 629f IORT, 664 RP. See Radical prostatectomy
Retinoblastoma gene renal sarcoma and, 665–666 RPLND. See Retroperitoneal lymph node
TCC with, 342 staging of, 658, 659t–661t disection; Retroperitoneal lymph
Retroperitoneal lymph node dissection summary of, 667–668 node dissection
(RPLND) surgery for, 658–663, 661t, 662f, 662t RPP. See Radical perineal prostatectomy
CT scan with, 642, 647, 647f operative technique in, 658–663, 661t, RT, Radiation therapy
low-stage disease treated with, 642–647, 662f, 662t RTC. See Randomized clinical trial; Rete
643f, 644f, 645f, 646f organs sacrificed during, 662t testis carcinoma
left-sided nerve-sparing in, 645–646, partial v. complete resection in, 658, RTOG. See Radiation Therapy Oncology
646f 661t Group
postoperative management in, 646 transabdominal approach in, 662f
right nerve-sparing in, 643–645, 645f, surgical outcome for, 663–664, 663f, SAMS. See Substratum adhesion
646f 664 molecules
technique for, 643–647, 643f, 644f, urinary bladder with, 666 SCA. See Sertoli cell adenoma
645f, 646f wood preservatives and, 652 SCC. See Squamous cell carcinoma
NSGCT treatment with, 600–605, 606f, Retrovirus vectors Schwannoma
607f, 608f, 609f, 610t applications of, 20–21 benign lesions of, 652t
postchemotherapy, 603–605, 606f attributes in, 20–21 Sclerosing Sertoli cell tumors (SSCT)
primary, 600–603, 606f, 607f, 608f, gene delivery in, 19 nongerm cell tumors as, 622, 623
609f, 610t gene expression in, 19–20, 19f pathologic characteristic of, 623
postchemotherapy disease treated with, gene therapy with, 19–21, 19f, 20f, SCT. See Sertoli cell tumors
647–649, 647f, 648f, 649f 21t, 26 Second malignant neoplasms (SMN)
indications for, 647–648, 647f vector production in, 19 Wilms’ tumor with, 767
special considerations for, 649, 649f Rhabdomyosarcoma (RMS) SEER. See U.S. Surveillance, Epidemiology,
technique of, 648–649, 648f bladder/prostate treatment for, 774–775 and End Results
radical orchiectomy with, 641–649, 643f, bladder primaries surgery in, 774–775 Seminal vesical biopsy (SVB)
644f, 645f, 646f, 647f, 648f, 649f chemotherapy in, 774 Seminal vesicles diagnosis of, 555, 556t
general considerations for, 641–642 outcome of, 775 Seminal vesicle invasion (SVI)
summary of, 649 prostate primaries surgery in, 775 brachytherapy and, 119
testis tumors staging with, 570, 571 radiation therapy in, 774 PC staging with, 461
806 Index

Seminal vesicles Sertoli cell tumors (SCT) Squamous cell carcinoma (SCC) (Continued)
diagnosis of, 555–557, 556f, 556t management of, 624 Mohs’ micrographic surgery for, 704, 723
Gleason score in, 556, 556t, 564f nongerm cell tumors as, 618t, 622–624, MRI with, 696
MRI in, 557 622f, 623f, 624f, 625f natural history of, 700
PSA in, 555–558, 556t, 557t, 561, 562, pathologic characteristics of, 622–624, occurrence of, 723
564f 622f, 623f, 624f, 625f partial penectomy for, 703
SVB in, 555, 556t Serum tumor markers presentation in, 696, 699–700
transrectal ultrasound in, 555, 556f AFP, 568, 597 radiation therapy for, 705
treatment options for, 557–565, 557t, HCG, 568, 597 staging in, 696–697, 701–702, 701t
559f, 560f, 561f, 562f, 563f, 564f LDH, 568, 597 staging systems for, 697, 697t
adjuvant radiation therapy, 557–558, NSE, 568 summary of carcinoma of, 707
557t NSGCT, 568 superficial carcinoma of, 699–707, 700f,
androgen deprivation in, 562 PLAP, 568, 597 701t, 702–703, 702t, 705t, 706t,
CT scan with, 558, 563f testis tumors with, 568 707f, 707t
EBRT in, 558, 560f SF-36. See RAND Medical Outcomes Study surgical excision for, 702
hormone therapy in, 562 Sickness impact profile (SIP) symptoms in, 696, 699–700
laparoscopy in, 558 HRQOL measured with, 104 TNM staging system for, 702, 702t
LHRH in, 560f Sigmoid colon conduit total penectomy for, 703–704
multimodal, 558–565, 560f, 561f, 562f, noncontinent cutaneous urinary diversion primary lesion treatment for, 715–716
563f, 564f with, 406 radiation therapy for, 718
Seminoma Silver staining nucleolar organizer regions urethral cancer pathology with, 674
clinical stage I management of, 585–588, (AgNOR) SSCT. See Sclerosing Sertoli cell tumors
586t RCC with, 175 Stauffer’s syndrome
adjuvant radiation therapy in, 587–588 Simpson-Golabi-Behmel syndrome RCC associated with, 174t
primary chemotherapy in, 588 Wilms’ tumor with, 755 Stoma stenosis
prognostic factors in, 586–587, 586t SIOP. See Société International d’Oncologie cutaneous urinary diversion with, 431
surveillance’s role in, 585–587, 586t Pédiatrique Stone formation
clinical stage II management of, 588–589 SIP. See Sickness impact profile cutaneous urinary diversion with, 434
clinical stage III management of, 589–590 SKY. See Spectral karyotyping Stromal epithelial interactions, 15–16, 15f
cryptorchid testes with, 593 SLDR. See Sublethal damage repair Sturge-Weber syndrome
early detection of, 584–585 SMN. See Second malignant neoplasms pheochromocytoma with, 139
EGCT with, 593 Société International d’Oncologie Sublethal damage repair (SLDR), 44
epidemiology of, 579–580 Pédiatrique (SIOP) Substratum adhesion molecules (SAMS), 13f
etiology of, 579 Wilms’ tumor with, 753, 762, 764–766, Suprahepatic vena cava
HIV patients with, 592 765f radical nephrectomy with, 226–230, 228f,
immunosuppression patients with, 592 cooperative group trials for, 764–766, 229f
noncompliant patients with, 593 765f Surgery
other issues with, 591–592 Southwest Oncology Group (SWOG) adrenal gland approach in, 156
pathology of, 581, 582t–584t bladder TTC study by, 325 adrenal tumors with, 139–149, 142t, 143f,
prevention of, 584–585 metastatic RCC study by, 262–263 144f, 145f, 146f, 147f, 148f,
primary surgery for, 580–581 Spectral karyotyping (SKY) 153–167, 154f, 154t, 157f, 158f,
residual masses management for, 591 cancer with, 8 159f, 160f, 161f, 161t, 162f, 163f,
spread pattern of, 581–584 SPL. See Surgical Planning Laboratory 164f, 166t
staging for, 580–581, 582t–584t Squamous cell carcinoma (SCC) anatomic nerve-sparing radical
summary of, 593 chemotherapy treatment in, 718 prostatectomy, 514–527, 516f,
symptoms of, 580 inguinal lymph node treatment for, 517f, 518f, 519f, 520f, 521f, 522f,
TIN with, 581 716–718, 716t 523f, 524f, 525f
treatment effects with, 590–591 penis with bladder neck transection/reconstruction
chronic, 590–591 brachytherapy for, 705–706 for, 522, 523f
fertility in, 591 circumcision for, 702–703, 710 Denonvilliers’ fascia incision
late gonadal toxicity in, 590 classification of, 697t for, 521f
psychologic toxicity in, 590–591 CT scan with, 696 dorsal venous ligation/transection for,
second malignancy in, 590 diagnosis of, 700–701 516–519, 518f, 519f, 520f, 521f
unusual cases management for, 591–592 diagnosis/staging of, 695–697, 697t endopelvic fascia incision for, 515–516,
bilateral tumors in, 592 differential diagnosis of, 695–696 517f, 518f
high HCG patients in, 591–592 epidemiology of, 695 limited pelvic lymphadenectomy for, 515
horseshoe/ectopic kidney in, 592 external beam radiation for, 706, 707t neurovascular bundies separation for,
IBD in, 592 grading in, 700, 701t 519, 521f
Sertoli cell adenoma (SCA) histology in, 701 patient selection for, 514–515
nongerm cell tumors as, 622, 624, 624f, HPV in, 695–696, 710 prostatic pedicles transection/control
625f incidence of, 695 for, 519–522, 522f
pathologic characteristic of, 624, 624f, inguinal node management with, puboprostatic ligaments incision for,
625f 706–707, 707f 515–516, 517f
Sertoli cell tumors (SCT) Jackson staging system for, 701t, 702 vesico-urethral anastomosis for,
clinical presentation of, 622 laboratory studies in, 696 522–523, 524f, 525f
evaluation of, 624 laser therapy for, 704–705, 705t anatomy in, 156
Index 807

Surgery (Continued) Surgery (Continued) Surgery (Continued)


approaches to, 141, 142t sigmoid colon conduit for, 406 surgical anatomy with, 219, 219f
Cushing’s syndrome in, 141, 142t T pouch, 413–414, 414f, 415f, 416f, surgical incisions in, 219–220, 220f,
cutaneous urinary diversion in 417f, 423t 221f, 222f
appendiceal approach to, 409 taenia incision in, 407f technique for, 200–201, 238–240, 238f,
Benchekroun nipple approach to, 409 transverse colon conduit for, 404–406, 239f, 240f, 241f
colocolostomy in, 405f 405f TEE for, 218
colonic conduits for, 404–406, 405f U. Miami pouch, 424t thoracoabdominal approach in, 220,
complications with, 430–437, 433t, UCLA pouch, 422t, 423t 222f, 223f
435t ureterointestinal anastomoses with, three approaches to, 199
continent, 406–427, 407f, 408f, 409f, 407f, 427–430, 429f, 430f, 431f thrombectomy with, 227f
410f, 412f, 413f, 414f, 415f, 416f, ureterosigmoidostomy in, 406–407 laparoscopic radical prostatectomy,
417f, 418f, 419f, 420f, 421f, 422f, Wallace technique for, 430, 430f 537–538
422t, 423–424t, 425f, 426f, 427f flank approach to, 145–147, 145f, 146f, anterior phase in, 537
continent catheterizable urinary 156–158, 157f main operation steps for, 537–538
diversions for, 407–409 hyperaldosteronism in, 142t medical preparation for, 537
continent mechanism in, 409–411, 410f laparoscopic partial nephrectomy Montsouris technique in, 537
continent reservoirs for, 411–427, 412f, complications of, 252–283 neurovascular bundles dissection in,
413f, 414f, 415f, 416f, 417f, contraindications for, 249 538
418–420f, 421f, 422f, 422t, financial analysis of, 283 patient installation in, 537
423–424t, 425f, 426f, 427f, 428f, indications for, 249 posterior phase in, 537
436–437 renal hypothermia with, 250, 254f prostatic pedicles dissection in, 538
continent v. noncontinent, 399 renal parenchymal repair in, 253f specimen extraction in, 538
cutaneous pyelostomy for, 400, 401f results of, 250–252, 255t transperitoneal approach to, 537
cutaneous ureterostomy for, 400–401, technique for, 249–250, 250t, 251f, urethra section in, 538
402f 252f, 253f vesicoprostatic dissection in, 538
Duke pouch, 422t, 424t laparoscopic radical nephrectomy, vesicourethral anastomosis in, 538
flap valve approach to, 409 218–230, 219f, 220f, 221f, 222f, lateral flank approach in, 156–158, 157f
Florida pouch, 422t, 424t 223f, 224f, 225f, 226f, 227f, 228f, lymphadenectomy in, 728f, 729f,
four surgical techniques for, 409 229f, 230t 730–732, 732f
gastric pouch, 424t, 426–427, 427f, anterior subcostal transperitoneal management of TCC through, 274–279,
428f, 436–437 incision in, 220, 220f, 221f 275t, 277t, 279f
hydraulic valve approach to, 409 approach selection for, 237–238 modified posterior approach to, 143–145,
ileal conduit for, 401–403, 403f, 404f bilateral subcostal incision in, 220 144f, 159, 160f
ileocecal, 411–416, 412f, 413f, 414f, complications of, 241, 243t, 244t nephrectomy with, 146, 146f
415f, 416f, 417f, 418–420f, 421f, concomitant adrenalectomy with, neuroblastoma treatment with,
422f, 422t, 423–424t 245–246 746–750
ileocecal conduit for, 405 contra-indications for, 237 open radical nephrectomy (ORN)
Indiana pouch, 416–421, 422t, CT scan for, 218 LRN v., 201–203, 202t
423–424t, 425f, 436 cytoreductive, 247–248 RCC with, 198–199, 199f, 201–203,
intraoperative pouch testing for, 411 diaphragmatic incision in, 220 202t, 247t
intussuscepted nipple valve approach evaluation for, 218–219 open surgical techniques in, 156–159,
to, 409 financial implications of, 248 158f, 159f, 160f
jejunal conduit for, 404 follow-up for, 246t orthotopic bladder substitution, 443–454,
Kock pouch, 411–413, 412f, 413f, 423t, Gerota’s fascia in, 224f, 226, 239 445f, 446f, 447f, 448f, 449f, 450f,
436 indications for, 218–219, 237 451f, 452f, 453f, 453t, 454t
laparoscopic ileal conduit for, 438, 439t IVC with, 222–226, 227f, 228f atraumatic urethra dissection as, 446
LeBag, 422t larger renal tumors with, 245, 247t bipolar electrocautery in, 446
Mainz pouch, 414–416, 418–420f, 423t, left side, 220–222, 224f, 225f bladder construction as, 447–448, 450f,
436 lymphadenectomy with, 222 451f, 453f
nasogastric tube for, 400 MRI for, 218 cystectomy as, 445–446, 445f, 446f,
noncontinent, 400–406, 401f, 402f, oncologic outcome of, 244–245 453f
403f, 404f, 405f ORN v., 201–203, 202t, 247t ileal segment resection as, 446, 447f,
novel techniques for, 438–439, 439t RCC with, 199–203, 202t, 237–248, 448f
Penn pouch, 424t, 426, 426f 238f, 239f, 240f, 241f, 242t, 243t, long-term follow-up for, 452–454, 454t
postoperative care for, 400 244t, 245f, 246t, 247t, 248t, 249f mesenteric window close in, 448f
preoperative preparation for, renal vein in, 224f metabolic acidosis in, 450, 453t
399–400 renal vein involvement with, 246–247, nerve preservation in, 445–446, 445f,
pseudoappendiceal approach to, 409 248t, 249f 446f
quality of life with, 437–438, 438t results of, 241, 242t Nesbit technique in, 449f
rectal bladder urinary diversion for, right side, 222, 225f operative technique for, 444–448, 445f,
406–407, 407f, 408f ring retractor with, 224f 446f, 447f, 448f, 449f, 450f, 451f,
rectus fascia incision in, 404f specimen extraction with, 241–244, 452f, 453f
renal pelvis incision in, 401f 245f orthotopic ileal bladder substitute as,
right colon, 416–427, 422t, 423–424t, suprahepatic vena cava with, 226–230, 446, 447f, 448f
425f, 426f, 427f, 428f, 436–437 228f, 229f patient assessment for, 443–444
808 Index

Surgery (Continued) Surgery (Continued) Surgery (Continued)


postoperative management for, patient positioning in, 370, 370f endoscopic management for, 293–295,
448–452, 453t pelvic lymphadenectomy with, 295f, 296f
preoperative patient selection for, 373–374, 375f, 376f laparoscopic nephroureterectomy in,
443–444 peritoneum incision in, 378f 278–279, 280t, 289–291, 290f,
U-shaped distal ileum in, 450f posterior pedicle ligation in, 375–378, 291f
ureteroileal anastomosis as, 446–447, 378f, 379f, 380f laparoscopic v. open
449f, 450f postoperative care for, 384–385 nephroureterectomy in, 219f, 220f,
ureters in, 446 preoperative evaluation for, 344, 280t
urethral anastomosis as, 447–448, 450f, 369–370 open nephron-sparing surgery for,
451f, 453f preparation for, 344 291–293, 292f, 293f
partial adrenalectomy in, 148–149, TCC with, 343–347 open nephroureterectomy in, 277–278,
148f urachal remnant excision in, 371f 280t, 287–289, 289f, 291f
partial nephrectomy for, 230–235, 233f, ureteral dissection in, 372 open segmental ureterectomy for,
234f vaginal incision in, 383f 292–293, 293f, 294f
AML with, 235 vaginal wall dissection in, 384f, 385f partial ureterectomy in, 276–277,
CT for, 230–231 vascular pedicle isolation in, 377f 277t
segmental polar, 232, 233f radical laproscopic, 149, 150–151f, thoracoabdominal approach to, 158, 158f
simple enucleation, 235 160–167, 161f, 161t, 162f, 163f, total penectomy for, 703–704
in situ, 231 164f, 166t complications in, 704
transverse resection, 234–235, 234f radical perineal prostatectomy, impact of, 704
wedge resection, 232–234, 234f 529–530 surgical technique in, 703
partial penectomy in, 701 radical prostatectomy, 465–474, 466t, transabdominal approach to, 147–148,
PC treatment complications with, 468t, 469t, 471t, 472t, 473t 147f, 158–159, 158f
547–552 biopsy Gleason sum with, 471, 472f TUR
pheochromocytoma in, 142t blood loss during, 469, 469f anesthesia for, 359
posterior approach to, 142–143, 143f, cancer control with, 469f, 470–473, bimanual examination for, 359, 359f
159, 159f 471f, 472f bladder cancer diagnosis with,
preoperative management with, 141, 142t, CT scan assessment with, 466 306, 338
156 early complications with, 468–470, Bladder-sparing therapy with,
procedures for penis in, 723–732, 724f, 468f, 469f 347–348
725f, 726f, 727f, 728f, 729f, erectile function after, 474 bladder tumors with, 358–365, 359f,
732f hospitalization days with, 469f 360f, 364t, 365f
radical cystectomy intraoperative complications with, complications with, 362–364
abdominal exploration in, 370 468–470, 468f, 469f equipment for, 359–360
adjuvant v. neoadjuvant, 346–347 late complications with, 470 fluoroscopic cystoscopy for, 365
bladder treated with, 368, 369–387, lymph node metastases with, 472t instrumentation for, 359–360
370f, 371f, 372f, 373f, 375f, 376f, mortality with, 468f intraoperative problem management in,
377f, 378f, 379f, 380f, 381f, 382f, MRI assessment with, 466 361–362
383f, 384f, 385f, 386f obturator nerve injury from, 470 irrigation fluid in, 361
bowel mobilization in, 370–372, 372f, outcomes after, 469f, 470–474, 471f, laser surgery for, 364–365, 364t,
373f 472f, 473f 365f
chemotherapy with, 346–347 patient selection for, 466 postoperative management in, 362
complications of, 346 PC natural history and, 465–466, preparation for, 358–359
Denonvilliers’ fascia in, 279f, 377, 378f, 466t primary muscle invasive tumor
380, 380f pelvic lymph node dissection with, treatment with, 362
discussion of, 385–387 467–468 second look resection in, 362
dorsal venous complex in, 380, 381f, pretreatment risk stratification with, TCC bladder with, 317–318
382f 466–467 TCC diagnosis with, 338
female patient anterior dissection in, PSA risk assessment with, 466–467, technique for, 360–361
382–384, 383f, 384f, 385f, 386f 470, 471f Surgical Planning Laboratory
female pelvic sagittal section in, 386f PSA survival with, 469f (SPL), 119
females with, 345 rectal injury from, 469 SVB. See Seminal vesical biopsy
gauze sponge withdrawing in, 376f results of, 468 SVI. See Seminal vesicle invasion
ICU for, 384 risk factors with, 472f, 473f SWOG. See Southwest Oncology
iliac artery skeletonizing in, 375f seminal vesicle involvement Group
incision in, 370, 371f with, 472f Systemic therapy
lateral vascular pedicle ligation in, treatment rationale, 465 IFN in, 263
374–375, 377f urinary function with, 473, 473f IL-2 in, 263–264
lymphadenectomy with, 345–346 urinary incontinence after, 473, 473f RCC with, 263–264
male patient anterior dissection in, RCC, indications for, 198
378–382, 381f, 382f RMS treated with, 774–776 T-cells
males with, 344–345 seminoma, 578–579 HCT with, 93, 96, 96f
outcomes of, 346 TCC with, 276–279, 277t, 279f, 287–296, T pouch
overhead pelvic view in, 372f, 373f 288f, 289f, 290f, 291f, 292f, 293f, complications with, 435t
overview of, 343–344 294f, 295f, 296f continent reservoirs compared to, 423t
Index 809

T pouch (Continued) Testis tumors (Continued) Toxicity


reoperation rate with, 435t miscellaneous tumors of, 627–636, androgen deprivation therapy with, 499
technique for, 413–414, 414f, 415f, 416f, 627f, 628f, 629f, 630f, 631f, 632f, TRAIL. See Tumor necrosis factor-related
417f 633f, 634f, 635f, 636f apoptosis-inducing ligand
urinary diversion with, 413–414, 414f, mixed sex cord/gonadal stromal tumors Transesophageal echocardiography (TEE)
415f, 416f, 417f, 423t, 435t as, 626–637, 627f, 628f, 629f, 630f, radical nephrectomy for, 218
Taxanes 631f, 632f, 633f, 634f, 635f, 636f Transforming growth factor (TGF)
docetaxel as, 66f, 67 ovarian surface epithelial type tumors RCC with, 176
genitourinary cancer treatment with, of, 637 TCC prognosis with, 343
65–67, 67f plasmacytoma as, 618t, 636, 636f Transitional cell carcinoma (TCC)
paclitaxel as, 65–67, 67f Reinke’s crystals in, 619, 621 biologic prognostic markers for, 321–322
TCC. See Transitional cell carcinoma Rete testis carcinoma as, 628–629, 629f BCG in, 321
TEE. See Transesophageal Sertoli cell tumors as, 618t, 622–624, blood group antigens as, 321
echocardiography 622f, 623f, 624f, 625f cytogenic markers as, 321–322
Telomerase sex cord/stromal origin with, 618–626, FISH as, 321
urologic cancers with, 16 618f, 618t, 619f, 620f, 621f, 622f, molecular genetic markers as, 321–322
Telomeric repeat amplification protocol 623f, 624f, 625f, 626f tumor cell products as, 321
(TRAP) serum tumor markers of, 568 WTG in, 321
TCC diagnosis with, 272 AFP, 568 bladder-sparing therapy for, 347–349
Temporary prostate brachytherapy HCG, 568 bladder with, 317–331, 318t, 322t, 326t,
(TPB) LDH, 568 328f, 329t, 330t
prostate adenocarcinoma treated with, NSE, 568 chemotherapy for, 279–281, 323–324
484–485, 488, 489t NSGCT, 568 CISCA regimen in, 281
BRFS with, 488, 489t PLAP, 568 CMV regimen in, 281
technique in, 484–485 staging of, 569–574, 572t, 573t, 574t doxorubicin in, 324
Teratoma, testis tumors, 781 AJCC in, 569, 571, 572t MMC in, 323–324, 326t
Testicular cancer bipedal lymphangiogram for, 570 MVAC regimen in, 281
HRQOL with, 109 CT scan for, 569–571 thiotepa in, 323
Testicular germ cell tumors (GCT). See distant metastasis in, 571, 572t valrubicin in, 324
Testis tumors IGCCCG with, 571, 573t cytologic prognostic factors for, 320–321
Testicular intraepithelial neoplasia (TIN) introduction to, 569 DNA ploidy in, 321
seminoma with, 581 modalities of, 569–571 urine cytology in, 320–321
Testis tumors PET scan for, 570 diagnosis of, 270–272, 271f, 272f, 273f
diagnosis of, 567–568 primary tumor in, 571, 572t bladder mucosa distant from tumor in,
introduction, 567 regional lymph nodes in, 571, 572t 320
presentation of, 567–568 RPLND for, 570, 571 CT scan in, 271–272, 273f, 339
imaging of, 568–569 serum tumor markers in, 571, 572t cytology in, 272, 273f
nongerm cell tumors of, 617–637, 618f, TGF. See Transforming growth factor endoscopic evaluation in, 272
618t, 619f, 620f, 621f, 622f, 623f, Therapeutic radiation (RT) MRI in, 271–272, 339
624f, 625f, 626f, 627f, 628f, 629f, adjuvant, 46 other markers in, 272
630f, 631f, 632f, 633f, 634f, 635f, definitive, 46 radiographic evaluation in, 270–272,
636f palliative, 46 271f, 272f, 273f
acquired immunodeficiency syndrome radiation oncology clinical practice and, TRAP in, 272
with, 637 46–47, 47t tumor extent evaluation in, 338–340
adenomatoid tumor of epididymis as, Thiotepa tumor metastatic evaluation in, 339
618t, 633, 633f, 634f bladder TCC with, 323 tumor stage and growth in, 338–339
carcinoid tumors of, 618t, 632–633, genitourinary cancer treatment with, 69 endoscopic management of, 274–276,
633f Three-dimensional conformal radiation 275t, 317–318, 320
epidermoid cysts as, 618t, 627–628, therapy (3DCRT) etiology in, 269–270
627f, 628f radiation therapy for adenocarcinoma evaluation of, 269–274, 271f, 272f, 273f,
generalized stroma tumors of, 636–637 with, 478–479, 478f, 482, 489 274t, 317
granulosa cell tumors as, 618t, 3D Slicer surgical simulation software, 119, grading of, 274, 318
624–626, 625f, 626f 120f histopathology of, 272–274, 319
hematologic tumors of, 618t, 633–636, 3DCRT. See Three-dimensional conformal intravesical therapy for, 322–327, 322t,
635f radiation therapy 326t
HIV and, 637 Thrombenectomy BCG in, 324–326, 326t
Klinefelter’s syndrome with, 619 radical nephrectomy with, 227f bropirimine in, 327
leukemia as, 618t, 636, 636f Thrombospondin-1 (TSP-1) chemotherapy, 323–324
Leydig cell tumors as, 618–622, 618f, TCC prognosis with, 341 combination therapy, 327
618t, 619f, 620f, 621f TIL. See Tumor-infiltrating lymphocytes doxorubicin in, 324
malignant lymphoma as, 618t, 633–636, TIN. See Testicular Intraepithelial neoplasia IFN in, 326
635f TNF. See Tumor necrosis factor immunotherapy in, 324–327
malignant mesothelioma as, 629–631, TNM. See Tumor, nodes, and metastasis KLH in, 326–327
630f, 631f Topotecan MMC in, 323–324, 326t
metastatic tumors of, 618t, 631–632, genitourinary cancer treatment with, 73f, rationale for, 322–323, 322t
632f 74 recommendations in, 327
810 Index

Transitional cell carcinoma (TCC) Transitional cell carcinoma (TCC) Transurethral resection (TUR) (Continued)
(Continued) (Continued) fluoroscopic cystoscopy for, 365
thiotepa in, 323 open nephron-sparing surgery for, instrumentation for, 359–360
valrubicin in, 324 291–293, 292f, 293f ball electrode as, 359
management of, 338–349 open nephroureterectomy in, 277–278, CIS with, 359
outcome for, 281–282 280t, 287–289, 289f, 291f intraoperative problem management in,
prognosis for, 320–322, 338–349 open segmental ureterectomy for, 361–362
angiogenic factors with, 341–342 292–293, 293f, 294f irrigation fluid in, 361
biologic markers in, 321–322 partial ureterectomy in, 276–277, 277t laser surgery for, 364–365, 364t,
cell adhesion molecules with, 341 surgical management of, 274–279, 275t, 365f
cell-cycle regulators with, 342–343 277t, 279f advantages/disadvantages with,
chromosomal abnormalities with, 340 BCG with, 276 364–365
conclusion about, 343 percutaneous resection with, 276 bladder tumor recurrence rate with,
cytologic factors in, 320–321 results of, 277, 279f 365
erb-B-2 with, 342–343 SEER on, 277, 279f techniques of, 364
genetic determinants with, 343 survival with, 279f types of, 364t
growth events with, 340–341 treatment of, 274–282, 275t, 277t, 279f, postoperative management in, 362
MDR gene with, 343 280t preparation for, 358–359
MVAC with, 343 upper urinary tract, 287–296, 288f, 289f, CT scan in, 358
MVD with, 341–342 290f, 291f, 292f, 293f, 294f, 295f, IVP in, 358
P53 with, 342 296f primary muscle invasive tumor treatment
retinoblastoma gene with, 342 endoscopic management for, 293–295, with, 362
summary of, 349 295f, 296f second look resection in, 362
TGF with, 343 initial evaluation for, 287 TCC bladder with, 317–318
treatment response with, 343 IVP for, 287 TCC diagnosis with, 338
TSP-1 with, 341 laparoscopic radical technique for, 360–361
VEGF with, 341 nephroureterectomy for, 289–291, Transverse colon conduit
prostatic urethra with, 327–328, 328f 290f, 291f noncontinent cutaneous urinary diversion
CIS only, 328f open nephron-sparing surgery for, with, 404–406, 405f
cystoprostatectomy for, 328f 291–293, 292f, 293f TRAP. See Telomeric repeat amplification
ductal, 328f open radical nephroureterectomy for, protocol
intravesical therapy for, 328f 287–289, 289f, 291f TRUS. See Transrectal US
stromal invasion, 328f open segmental ureterectomy for, TSP-1. See Thrombospondin-1
radiation therapy for, 281 292–293, 293f, 294f Tubulin modulation drugs
radical cystectomy for, 343–347 Transrectal US (TRUS) genitourinary cancer treatment with,
adjuvant v. neoadjuvant, 346–347 IGT with, 114, 117, 118, 122 64–65
chemotherapy with, 346–347 PC staging with, 461 Tumor, nodes, and metastasis (TNM)
complications of, 346 seminal vesicles diagnosis with, 555, 556f bladder cancer staging with, 308, 309
females with, 345 Transurethral resection of bladder tumor penis carcinoma staging with, 714,
lymphadenectomy with, 345–346 (TURBIT) 715t
males with, 344–345 bladder preservation, 390t, 392 Penis SCC staging with, 700, 700t
outcomes of, 346 Transurethral resection of the prostate RCC prognostic factors with, 210–211,
overview of, 343–344 (TURP) 210t
preoperative evaluation for, 344 brachytherapy and, 119 RCC staging with, 183–185, 184–185t,
preparation for, 344 PC staging with, 549 186, 186t, 259, 259f
renal pelvis and ureter with, 269–282, Transurethral resection (TUR) TCC staging with, 274, 274t
271f, 272f, 273f, 274t, 275t, 277t, anesthesia for, 359 Tumor-infiltrating lymphocytes (TIL)
279f, 280t bimanual examination for, 359, 359f immunotherapy with, 93, 94f, 264
staging of, 274, 274t, 319–320 bladder cancer diagnosis with, 306, 338 Tumor necrosis factor-related apoptosis-
AJCC in, 274t, 319 bladder preservation with, 389, 390t, 394f inducing ligand (TRAIL), 92
distant metastasis in, 274t Bladder-sparing therapy with, 347–348 Tumor necrosis factor (TNF), 12
lymph nodes in, 274t bladder tumors with, 358–365, 359f, 360f, Tumor thrombus
primary tumor in, 274t 364t, 365f RCC with, 203–204, 203t, 204f, 227f,
PUNLMP in, 274 complications with, 362–364 228f, 229, 229f
reproducibility of, 319–320 bladder explosion in, 364 Tunneled ureterointestinal anstomoses, 430
TNM in, 274, 274t bladder perforation in, 363 TUR. See Transurethral resection
surgical approaches to, 276–279, 277t, hemorrhage in, 362–363 TURBIT. See Transurethral resection
279f, 287–296, 288f, 289f, 290f, obturator nerve reflex in, 363 of bladder tumor
291f, 292f, 293f, 294f, 295f, 296f reabsorption syndrome in, 363–364 TURP. See Transurethral resection of the
endoscopic management for, 293–295, ureteral orifice injury in, 363 prostate
295f, 296f, 317–318 urinary infection in, 363
laparoscopic nephroureterectomy in, urinary stricture in, 363 U. Miami pouch
278–279, 280t, 289–291, 290f, 291f equipment for, 359–360 complications with, 435t
laparoscopic v. open cold-cup biopsy in, 360 continent reservoirs compared to, 424t
nephroureterectomy in, 219f, 220f, flexible cystoscope in, 360 reoperation rate with, 435t
280t video endoscopy in, 360 urinary diversion with, 424t, 435t
Index 811

UCLA Integrated Staging System (UISS) Ureterointestinal anastomotic (Continued) Urethrectomy (Continued)
RCC grade with, 259 small bowel, 427–430, 429f total male anterior urethrectomy in,
RCC staging with, 186–187, 186t, 260, 260f tunneled, 430 688–690
UCLA PCI. See University of California, ureterocolonic, 430, 431f summary of, 692
Los Angeles Prostate Cancer Index Wallace technique for, 430, 430f Urethrovesical anastomosis
UCLA pouch Ureterosigmoidostomy RRP with, 529
complications with, 435t cutaneous urinary diversion complications Urinary infection
continent colon pouches compared to, 422t with, 434–436 TUR complications with, 363
continent reservoirs compared to, 423t Ureterosignoidostomy Urinary stricture
reoperation rate with, 435t rectal bladder urinary diversion with, TUR complications with, 363
urinary diversion with, 422t, 423t, 435t 406–407 Urologic cancers
UICC. See Union Internationale Contre le Ureters adenovirus vectors gene therapy for,
Cancer orthotopic bladder substitution with, 446 21–22, 21t, 22f, 26t
UISS. See UCLA Integrated Staging System Urethral anastomosis aging/telomerase with, 16
Ultrasound (US). See also Focused orthotopic bladder substitution with, aptamers with, 16–17
ultrasound surgery 447–448, 450f, 451f, 453f bulbourethral with, 3, 4f
brachytherapy guided by, 118–119 Urethral cancer cancer genes and, 8–10
AUR with, 119 evaluation of, 675, 676t carcinogens in, 4–5
outcome of, 119 females with, 680–682, 680f, 681t, 682f cell cycle with, 11–12, 12f
procedure for, 118–119 fossa navicularis, 675, 677t cell signaling with, 12f, 14–15, 14f
incidentaloma with, 133 males with, 673–680, 674f, 676t, 677f, cellular biology of, 3–17, 4f, 6t, 10f, 12f,
NSGCT diagnosis with, 597, 598f, 599f, 677t, 678f, 679f 13f, 14f, 15f, 17f
600f multimodal therapy for, 680 CGH with, 8
prostate cancer surgery focused with, 122 operative techniques for, 676–680, enigmas of, 3, 4f
RCC diagnosing with, 178–179, 179f, 197 677f, 678f, 679f epigenetic effects in, 3–4, 4f
renal tumor ablation with, 209–210 radiation therapy for, 680 families and, 7–8
Wilms’ tumor evaluation with, 756, 756f urethrectomy with cystectomy for, 680 gene therapy for, 18–34, 19f, 20f, 21t,
Union Internationale Contre le Cancer natural history of, 675–676 22f, 23f, 24f, 25f, 26t, 27f, 31f,
(UICC), 196, 196t, 259 partial/total urethrectomy for, 676–680, 32f, 33f
University of California, Los Angeles Prostate 677f, 678f, 679f gene therapy molecular targets of, 25–33,
Cancer Index (UCLA PCI) pathology of, 673–680, 674f, 676t, 677f, 26t, 27f, 31f, 32f
HRQOL measured with, 105 677t, 678f, 679f gene transfer vectors for, 18–19
Upper urinary tract penile urethra, 675, 677t inflammation with, 5–7, 6t
transitional cell carcinoma in prostatic urethral cancer as, 675–676, liposomal gene transfer for, 21t, 24–25,
endoscopic management for, 293–295, 677t 24f, 25f, 26t
295f, 296f staging of, 675, 676t microarrays with, 10–11
initial evaluation for, 287 treatment of, 675, 677t modified vector tropism in, 33–34
IVP for, 287 Urethral recurrence molecular biology of, 3–17, 4f, 6t, 10f,
laparoscopic radical orthotopic bladder substitution with, 12f, 13f, 14f, 15f, 17f
nephroureterectomy for, 289–291, 444 nature v. nurture in, 4
290f, 291f Urethrectomy nonviral vectors gene therapy for, 21t,
open nephron-sparing surgery for, cystectomy with, 680 24–25, 24f, 25f, 26t
291–293, 292f, 293f females urethral cancer with, 681–682, oncolytic virotherapy for, 30
open radical nephroureterectomy for, 682f poxvirus vectors therapy for, 21t, 22–24,
287–289, 289f, 291f indications for, 686 23f, 26t
open segmental ureterectomy for, male urethral cancer with, 676–680, 677f, prostate with, 3, 4f
292–293, 293f, 294f 678f, 679f proteomics with, 10–11
Ureteral orifice injury operative technique for, 686–692, 687f, restricted transgene expression
TUR complications with, 363 688f, 689f, 690f, 691f, 692f, 693f, in, 34
Ureteral-small bowel anstomoses, 427–430, 694f retrovirus vectors therapy for, 19–21, 19f,
429f bulbar urethral arteries in, 691f 20f, 21t, 26t
Ureterectomy bulbocavernous muscle division in, 689f ribozyme construct approach to, 30–31,
partial cystoprostatectomy with, 686–688, 31f
TCC with, 276–277, 277t 687f, 688f, 689f, 690f, 691f, 692f, SKY with, 8
Ureterocolonic anstomoses, 430, 431f 693f stromal epithelial interactions with,
Ureteroileal anastomosis electrocautery in, 689f 15–16, 15f
orthotopic bladder substitution with, female urethrectomy in, 690 target cell death with, 26t, 29–30
446–447, 449f, 450f larger distal tumors in, 691–692 targeting vector specificity in,
Ureterointestinal anastomotic patient position in, 687f 33–34, 33f
cutaneous urinary diversion complications pelvic dissection in, 686–687, 687f, transcriptional targeting gene therapy
with, 431, 432t 688f for, 34
cutaneous urinary diversion with, perineal dissection in, 687, 688, 689f, tumor cell heterogeneity in, 16–17,
427–430, 429f, 430f, 431f 690f, 691f, 692f, 693f 17f
direct type, 427–430, 429f proximal tumors in, 691–692 tumor suppressor gene restoration for,
rectal bladder urinary diversion with, 407f small distal tumors in, 690–691, 693f, 31–33, 32f
refluxing v. nonrefluxing, 427 694f US. See Ultrasound
812 Index

U.S. Surveillance, Epidemiology, and End Von Hippel-Lindau (VHL) HA with, 761
Results (SEER) gene, 31 HASA with, 761
RCC reported by, 173 immunotherapy with, 264 hTERT with, 760
TCC surgical results from, 277, 279f pheochromocytoma with, 139, 140t ILNR with, 758–759, 758f
RCC with, 176, 197t, 264 NSE with, 761
Valrubicin Vysion PLNR with, 758–759, 758f
bladder TCC with, 324 bladder cancer diagnosis with, 305 precursor lesions in, 758–759, 758f,
Vascular endocrine growth factor (VEGF) 758t, 760t
Wilms’ tumor pathology with, 761 WAGR syndrome. See Wilms’ tumor and VEGF with, 761
Vascular endothelial growth factor (VEGF) AGR syndrome postoperative evaluation of, 755–757,
immunotherapy with, 98 Wallace technique 756f
RCC antiangiogenic with, 264–265 ureterointestinal anastomoses with, 430, CT scan for, 756, 756f, 758f
TCC prognosis with, 341 430f MRI for, 756
VEGF. See Vascular endocrine growth Watchful waiting (WW) ultrasound for, 756, 756f
factor; Vascular endothelial growth HRQOL with, 107 prognostic consideration for, 761, 761t
factor WHO. See World Health Organization relapse treatment for, 766–767
Verrucous carcinoma Wilms’ tumor Simpson-Golabi-Behmel syndrome with,
penis carcinoma with, 712 AGR syndrome with, 754, 754t, 759t 754
Vesico-urethral anastomosis bilateral Wilms’ tumor and, 766 SMN with, 767
radical prostatectomy with, 522–523, BWS syndrome with, 754, 754t, 759t surgical management of, 761–762
524f, 525f clinical presentation of, 755 WAGR syndrome with, 754, 758, 759t
Vesicoprostatic dissection cooperative group trials for, 762–766, Wilms’ tumor and AGR syndrome (WAGR
laparoscopic radical prostatectomy with, 764t, 765f syndrome)
538 NWTS-1, 763 Wilms’ tumor with, 754, 758, 759t
Vesicourethral anastomosis NWTS-2, 763 Wilms’ tumor gene (WTG)
laparoscopic radical prostatectomy with, NWTS-3, 763 TCC prognostic markers with, 321
538 NWTS-4, 763 Wood preservatives
VHL. See Von Hippel-Lindau NWTS-5, 763–764, 764t retroperitoneal tumors from, 652
Video endoscopy SIOP in, 764–766, 765f World Health Organization (WHO), 196,
TUR equipment including, 360 Denys-Drash syndrome with, 753, 758, 302
Vinblastine 759t WTG. See Wilms’ tumor gene
bladder preservation with, 391t differential diagnosis of, 757, 758f WW. See Watchful waiting
Vinca alkaloids epidemiology of, 753–754, 754t
genitourinary cancer treatment with, genetics of, 754–755 XRT. See External beam radiation therapy
64–65 late effects of treatment for, 767
Vomiting pathology of, 757–761, 758f, 758t, 760t
orthotopic bladder substitution with, 453t biologic parameters in, 759–761

Vous aimerez peut-être aussi