Vous êtes sur la page 1sur 8

Article

Clinical outcomes of dogs with transitional cell carcinoma receiving


medical therapy, with and without partial cystectomy
Marcus L. Bradbury, Christine M. Mullin, Shaban D. Gillian, Chick Weisse, Philip J. Bergman,
Michelle A. Morges, Lauren R. May, David M. Vail, Craig A. Clifford

Abstract — The objective of this retrospective study was to evaluate the effects of surgery on outcome for dogs
with naturally occurring urinary bladder transitional cell carcinoma. Forty-seven dogs met the inclusion criteria.
Thirty-one dogs (Group A) were treated with partial cystectomy and adjunctive medical therapy and 16 dogs
(Group B) were treated with medical therapy alone. Overall survival was greater in dogs treated with partial
cystectomy and adjunctive medical therapy (498 days for Group A versus 335 days for Group B, hazard
ratio 2.5; 95% confidence interval: 1.1 to 5.7; P = 0.026). Progression-free survival was not different between
groups (85 days for Group A versus 83 days for Group B; P = 0.663). No prognostic factors were identified for
progression-free survival. Due to the many cases in Group A that were lost to follow-up, time-to-event survival
analysis was performed. No significant difference in overall survival was noted, and no prognostic factors were
identified in the time-to-event analysis. Prospective, randomized studies are needed to determine the role of partial
cystectomy in the treatment of transitional cell carcinoma.

Résumé — Résultats cliniques des chiens atteints d’un carcinome à cellules transitionnelles recevant un
traitement médical, avec et sans cystectomie partielle. L’objectif de cette étude rétrospective était d’évaluer les
effets de la chirurgie sur les résultats chez des chiens atteints d’un carcinome à cellules transitionnelles de la vessie
d’origine naturelle. Quarante-sept chiens répondaient aux critères d’inclusion. Trente et un chiens (Groupe A) ont
été traités par cystectomie partielle et traitement médical d’appoint et 16 chiens (Groupe B) ont été traités par
thérapie médicale seule. La survie globale était plus élevée chez les chiens traités par cystectomie partielle et
traitement médical d’appoint (498 jours pour le Groupe A contre 335 jours pour le Groupe B, rapport de risque
de 2,5; intervalle de confiance à 95 % : 1,1 à 5,7; P = 0,026). La survie sans progression n’était pas différente entre
les groupes (85 jours pour le Groupe A contre 83 jours pour le Groupe B; P = 0,663). Aucun facteur pronostique
n’a été identifié pour la survie sans progression. En raison des nombreux cas dans le Groupe A qui ont été perdus
de vue lors du suivi, une analyse du temps de survie a été realisée. Aucune différence significative dans la survie
globale n’a été notée et aucun facteur pronostique n’a été identifié dans l’analyse du temps de survive. Des études
prospectives randomisées sont nécessaires pour déterminer le rôle de la cystectomie partielle dans le traitement du
carcinome à cellules transitionnelles.
(Traduit par Dr Serge Messier)
Can Vet J 2021;62:133–140

Hope Veterinary Specialists, Surgery Department, 40 Three Tun Road, Malvern, Pennsylvania 19355, USA (Bradbury, Mullin, May,
Clifford); Koret School of Veterinary Medicine, The Hebrew University in Jerusalem, Yehoshua Hankin St 21, Rehovot 76100, Israel
(Gillian); The Animal Medical Center, Interventional Radiology and Endoscopy, 510 East 62nd Street, New York, New York 10065,
USA (Weisse); Katonah Bedford Veterinary Center, Medical Oncology Department, 546 Bedford Road, Bedford Hills, New York
10507, USA (Bergman); Red Bank Veterinary Hospital, Medical Oncology Department, 197 Hance Avenue, Tinton Falls, New
Jersey 07724, USA (Morges); Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison,
2015 Linden Drive, Madison, Wisconsin 53706, USA (Vail).
Address all correspondence to Dr. Marcus Bradbury; e-mail: mlb0021@auburn.edu
Dr. Bradbury’s current address is Coastal Empire Veterinary Surgery, 355 Stephenson Avenue, Savannah, Georgia 31405, USA.
Use of this article is limited to a single copy for personal study. Anyone interested in obtaining reprints should contact the CVMA
office (hbroughton@cvma-acmv.org) for additional copies or permission to use this material elsewhere.

CVJ / VOL 62 / FEBRUARY 2021 133


Introduction treated with partial cystectomy had aimproved MST (217 d

U
with surgery compared to 133 d without surgery, P = 0.02) but
rinary bladder transitional cell carcinoma (TCC) is an
not progression-free survival (PFS) (18). A more recent study
important neoplasm in dogs, with the most common
by Marvel et al (19) evaluated 37 dogs with TCC treated with
clinical signs being hematuria, dysuria, and pollakiuria (1–2).
partial cystectomy and COX inhibitors, with or without other
Additionally, canine urinary bladder TCC has been used as a
medical therapies. The MST for all dogs was 348 d, but dogs
model to study the muscle-invasive form of urothelial carcinoma
with non-trigonal bladder TCC treated with full-thickness
in human patients (3–4). The invasive nature of TCC and its
A R T I C LE

partial cystectomy and daily piroxicam (6 chemotherapy) had


propensity to develop in the trigone of the bladder contribute
improved survival (772 d, P # 0.0001) (19). Although these
to the historically poor prognosis associated with this cancer in
latter results are promising, it is still unclear whether initiating
dogs (3). Lymph node metastasis is reported in 16% of dogs
surgery before medical therapy improves disease-free survival
with TCC at the time of diagnosis (4). Distant metastasis to the
compared to medical therapy alone.
lung or liver is reported in 14% of dogs at the time of diagnosis
The goal of the current retrospective study was to compare
and in 50% by the time of death (4).
the outcomes of dogs with urinary bladder TCC treated with
Treatments for dogs with urinary bladder TCC include medi-
partial cystectomy and adjuvant chemotherapy and COX inhibi-
cal therapy, radiation therapy, and surgery (4). The most often
tors versus those treated with chemotherapy and COX inhibitors
used medical therapies for canine TCC involve cyclooxygenase alone. A secondary aim was to identify prognostic factors for
(COX) inhibitors, systemic chemotherapeutic agents, or some PFS and overall survival (OS) for the same patient groups.
combination thereof (4). Previous studies have evaluated overall
response rates, remission duration, and survival times for vari- Materials and methods
ous medical therapies (5–7). It is the authors’ opinion that no Case selection
consensus exists as to the “gold standard” of therapy. Medical records of dogs that were presented over a 9-year
Although beneficial in local control, the high incidence of interval (September 2003 through November 2012) to 3 multi­
severe late radiation-associated toxicoses associated with con- disciplinary referral hospitals (Red Bank Veterinary Hospital,
ventional definitive radiation protocols has precluded radia- Tinton Falls, New Jersey, USA; The Animal Medical Center,
tion from being included as standard of care treatment (8–10). New York, New York, USA; and The Koret School of Veterinary
However, intensity-modulated radiation therapy allows for Medicine of the Hebrew University of Jerusalem, Israel) were
conformity of radiation dose and use of smaller treatment fields retrospectively reviewed to identify dogs diagnosed with urinary
to limit exposure to normal tissue and ultimately decrease the bladder TCC. Dogs were included if they were diagnosed with
adverse events previously noted (11). Further investigation is TCC by either histopathology (surgical or endoscopic biopsies)
needed, but this has promise in the treatment of TCC in dogs. or by cytology (obtained via traumatic catheterization or fine-
In humans, radical cystectomy for muscle-invasive urothelial needle aspiration) and were treated with systemic injectable
carcinoma improves outcomes but requires urinary diver- chemotherapy and COX inhibitors. Dogs were then grouped
sion techniques (12). Although complete cystectomy provides to compare those that were also treated with partial cystectomy
a surgical solution to canine tumors affecting the trigone, to those that received medical therapy alone. Partial cystectomy
few veterinary studies have examined the feasibility of total was defined as removal of any portion of the urinary bladder
cystectomy with ureteral diversion, given the postoperative with the intent of tumor cytoreduction. This was further cat-
challenges associated with home management after complete egorized into removal of all visible disease or partial resection
cystectomy (13–15). of visible disease as noted at the time of surgery. Dogs were
Common sequelae of complete cystectomy in the dog are excluded if they were not followed with sequential abdominal
permanent urinary incontinence, pyelonephritis, and azote- ultrasound after the initial 2 cycles of chemotherapy and then
mia (13–15). Thus, if confirmed to improve outcome, partial every 2 to 4 cycles thereafter. Dogs were also excluded if infor-
cystectomy and/or cytoreductive surgeries would be considered mation in regard to abdominal ultrasonography findings, type
desirable options. and/or dose of chemotherapy, and type of COX inhibitor used
Several studies have suggested cytoreductive surgeries may was missing. Data collected from the medical records included
benefit dogs with TCC (16–18). In 1 report, a benefit was breed age, gender, weight, date of diagnosis, staging tests per-
noted following complete excision of non-metastatic bladder formed, date of surgery, tumor location in bladder based on
tumors, with a median survival time (MST) of 365 d following abdominal imaging, surgical procedure (removal of all visible
surgery alone (16). Extent of tumor excision was prognostic, disease versus partial resection of visible disease), histopathol-
with an MST of 120 d when . 50% but , 100% of the ogy results, adjuvant therapy (chemotherapy, COX inhibitors),
tumor was removed, and 75 d when , 50% of the tumor was date of recurrence, date and cause of death, or loss to follow-up.
removed (16). A separate study noted 6/10 dogs with urinary Tumor location was based on the site of attachment or origin
bladder TCC treated with partial cystectomy, with or without of the tumor and was assigned to 1 of 4 anatomic regions of
ureteral re-implantation, survived over 1 y with or without the bladder: apex, body, trigone, or neck. The apex was defined
chemotherapy and/or radiation therapy (17). A retrospec- as the region immediately surrounding the previous urachal
tive study evaluating the combination of doxorubicin and attachment site at the cranial/proximal 25% of the bladder.
piroxicam for canine TCC noted that the subset of dogs first The body was defined as the region between the apex and the

134 CVJ / VOL 62 / FEBRUARY 2021


trigione. The trigione was defined as the triangular area, near using a 2-sample t-test or Mann-Whitney test, depending on
the neck, on the dorsal surface of the bladder immediately sur- normality of the data. Categorical variables were compared
rounding the implantation site of the ureters. The neck was between cohorts using a Fisher’s Exact test. The Kaplan-Meier
defined as the region ventral and distal to the trigione and was method was used to estimate and display the distribution of
site of bladder termination and urethral origin. Ureteral involve- PFS and OS. Differences between potential prognostic subsets
ment was defined as visualization of tumor within one or both and between study arms were compared using the log-rank
ureters at the time of initial ultrasonographic examination or test. All reported P-values are 2-sided and P , 0.05 was used

A R T I C LE
at the time of surgery. Prostatic involvement was determined to determine statistical significance. All statistical analyses were
based on ultrasonographic evidence of tumor extension into the completed using a commercial software package (Prism v. 6.0b;
prostate, with prostate enlargement and mineralization. After GraphPad Software, La Jolla, California, USA).
initiation of treatment, dogs were periodically reassessed with
physical examinations, clinical laboratory data, and imaging. Results
Urinary tract ultrasonography was employed to determine tumor Forty-seven dogs met the inclusion criteria (Group A; n = 31
response and remission status, using World Health Organization and Group B; n = 16). Information regarding signalment is
(WHO) criteria. Tumor size was calculated as the product of presented in Table 1. Clinical signs included; hematuria (n = 32;
the longest diameter and the greatest perpendicular diameter. 68%), pollakiuria (n = 23; 49%), stranguria (n = 21; 45%), dys-
Complete remission was defined as disappearance of all measur- uria (n = 10; 21%), polydipsia (n = 3; 6%), 2 cases each of leth-
able disease, partial remission was defined as . 50% decrease argy, urine dribbling, and polyuria, and 1 case each of urinary
in tumor size from baseline measurements, stable disease was incontinence, tenesmus, kyphosis, and recurrent lower urinary
defined as , 25% increase in tumor size from baseline measure- tract infection. In 3 dogs (Group A, n = 2; Group B, n = 1) the
ment, and progressive disease was defined as . 25% increase in diagnosis was incidental, and the dogs had no clinical signs.
tumor size, based on baseline measurement or the appearance Twenty-four (51%) dogs had a urinalysis and 11 (23%)
of new lesions (either locally or at a distant site). dogs had a urine culture completed before initiation of treat-
Dogs were assigned to 1 of 2 groups: Group A — dogs that ment. Urinalysis revealed atypical transitional cells in 6 of the
underwent a partial cystectomy, followed by treatment with 24 dogs and bacterial culture was positive in 5 of the 11 dogs.
systemic chemotherapy and COX inhibitors; and Group B — Bacteria isolated included Escherichia coli, Proteus mirabilis,
dogs receiving only medical therapy with chemotherapy and and Staphylococcus intermedius. All dogs underwent an initial
COX inhibitors. Median PFS and median OS were compared abdominal ultrasonographic examination that identified a
between the 2 groups. Data were also evaluated for prognostic bladder mass and 91% (n = 43) had a detailed description
factors across both groups. Dogs in Group A were restaged using of the mass location available for review. The most common
ultrasonography for detection of tumor recurrence following location was the trigone (Group A, n = 8; Group B, n = 13),
surgery and chemotherapy initiation; whereas, dogs in Group B followed by apical (Group A, n = 11; Group B, n = 1), and neck
were monitored for change in tumor status from baseline follow- (Group A, n = 2; Group B, n = 1). Ten dogs (Group A, n = 7;
ing initiation of chemotherapy. Ultrasonography was done after Group B, n = 3) had multifocal disease. Three dogs (Group A,
the initial 2 cycles of chemotherapy, then every 2 to 4 cycles n = 1; Group B, n = 2) had ultrasonographic evidence of ure-
thereafter. Progressive disease (PD) was determined in both teral involvement, 10 dogs (Group B, n = 10) had evidence of
Groups A and B by ultrasonographic evaluation. urethral involvement, and 5 dogs (Group A, n = 2; Group B,
n = 3) had evidence of prostatic involvement. No dogs had ultra-
Histology sonographic evidence of metastasis to abdominal organs outside
All biopsy samples were reviewed and the diagnosis of TCC the urinary tract. No dogs had evidence of regional lymph node
confirmed by Board-certified anatomic pathologists. Histologic involvement at the time of initial diagnosis. Baseline thoracic
margins were classified as complete or incomplete. radiographs were done for 60% of the dogs (n = 28). All dogs
in Group A and 3 dogs (19%) in Group B had diagnosis
Cytology confirmed via histopathology (surgically retrieved samples for
All cytology samples were reviewed and the diagnosis of TCC Group A and cystoscopic biopsies for Group B). The diagnosis
was confirmed by Board-certified clinical pathologists. of TCC was confirmed in the remaining Group B dogs (n =13;
81%) by evaluation of cytology samples collected via traumatic
Statistical analysis catheterization (n = 10; 63%) or fine-needle aspiration (n = 3;
Continuous data were expressed as median and range, and cat- 19%). Five dogs (38%) diagnosed via cytology had evidence of
egorical data as frequencies and percentages. Progression-free concurrent lower urinary tract infection.
survival and OS were calculated from the date of first chemo- Thirty-one dogs (66%; Group A) were treated with partial
therapy to the date of PD or death from any cause, and death, cystectomy, followed by systemic chemotherapy and COX inhibi-
respectively. Dogs were censored if they had not developed PD tors. Sixteen dogs (34%; Group B) were treated with systemic
or died at the time of data analysis, or if they were lost to follow- chemotherapy and COX inhibitors alone. One dog had a left
up. The cases that were lost to follow-up were then uncensored neocystoureterostomy, due to left ureteral involvement. No dogs
and a time-to-event (TTE) comparison was done for OS. with urethral involvement were in Group A. Except for 1 case,
Continuous variables were compared between groups of patients dogs with prostatic extension and those with trigonal tumors had

CVJ / VOL 62 / FEBRUARY 2021 135


Table 1. Characteristics of dogs in Groups A and B.
Group A Group B
Parameter (31 dogs) (16 dogs) P-value
Age (y)
Median (range) 10.3 (5.3 to 15.3) 11.1 (5.6 to 15.6) 0.256a
Gender
Spayed female 18 (58%) 8 (50%) 0.718b
A R T I C LE

Neutered male 9 (29%) 7 (44%)


Intact male 3 (10%) 0 (0%)
Intact female 1 (3%) 1 (6%)
Weight (kg)
Median (range) 14.2 (2.3 to 80) 12.7 (7.1 to 46.4) 0.953a
Breed
Mixed breed 7 (23%) 3 (19%) 0.761b
Beagle 6 (19%) 3 (19%)
West highland white terrier 2 (6%) 1 (6%)
Other (n = 1 each breed) 16 (52%) 9 (56%)
a t-test.
b Chi-square test.

Table 2. Summary of medical therapy protocols for dogs in Groups A and B.


Group A Group B
Medical therapy protocols (31 dogs) (16 dogs)
Single agent 20 (65%) 9 (56%)
Doxorubicin 13 (42%) 4 (25%)
Mitoxantrone 4 (13%) 2 (13%)
Vinblastine 3 (10%) 1 (6%)
Carboplatin 0 2 (13%)
Multi agent 11 (35%) 7 (44%)
Vinblastine/doxorubicin 3 (10%) 2 (13%)
Mitoxantrone/doxorubicin/carboplatin 3 (10%) 1 (6%)
Mitoxantrone/doxorubicin 2 (6%) 3 (19%)
Vinblastine/mitoxatrone 1 (3%) 0
Doxorubicin/carboplatin 0 1 (6%)
Doxorubicin/vinblastine/mitoxantrone/carboplatin 1 (3%) 0
Cisplatin/carboplatin/gemcitabine/cyclophosphamide 1 (3%) 0
Rescue protocol 2 (6%) 1 (6%)
Cyclophosphamide 2 (6%) 0
Cyclophosphamide/gemcitabine 0 1 (6%)
COX inhibitor
derocoxib 16 (52%) 6 (38%)
piroxicam 9 (29%) 7 (44%)
meloxicam 1 (3%) 0
carprofen 0 1 (6%)
multiple 5 (16%) 2 (13%)

partial thickness cytoreductive surgeries that did not disrupt the All study dogs were treated with a COX inhibitor at stan-
ureterovesicular junction. Based upon the surgical report, complete dard anti-inflammatory doses. The COX inhibitors used for
excision of all gross disease was achieved in 16 dogs (52%) and par- both treatment groups are summarized in Table 2. Twenty-two
tial excision of gross disease achieved in 15 (48%). Histopathology dogs (47%) received deracoxib only, 16 (34%) received piroxi-
reports were available for review for all 31 cases and margins were cam only, 1 (2%) received carprofen only, and 1 (2%) received
deemed complete in 6 dogs (19%), incomplete in 21 dogs (68%), meloxicam only. Seven dogs (15%) were switched from one
and histologic margins were not noted in the histopathology report COX inhibitor to another at varying times throughout therapy,
in 4 cases (13%). One dog had regional lymph node biopsy per- due to patient intolerance to a specific drug. The most com-
formed at the time of cystectomy and no evidence of metastatic monly intolerable drug was piroxicam and the most common
disease was detected histologically. Specific information on peri- undesirable side effects were vomiting, diarrhea, and azotemia.
operative surgical complications was not available, but all dogs Three dogs (6%) received sequential piroxicam then deracoxib,
survived to discharge. One dog (2.7%) developed tumor seeding to and 1 (2%) each received sequential piroxicam then meloxicam,
the abdominal wall associated with the surgical scar 95 d after sur- piroxicam then carprofen, piroxicam then firocoxib, and piroxi-
gery, following a diagnosis via fine-needle aspiration and cytology. cam then carprofen then deraxocib.

136 CVJ / VOL 62 / FEBRUARY 2021


Table 3. Prognostic factors evaluated for progression-free survival (PFS) and overall survival (OS) for
dogs treated with partial cystectomy and medical therapy or medical therapy alone.
PFS OS OS TTE
Variable (days) P-value (days) P-value (days) P-value
Surgery 0.026
Yes 85 0.663 498 HR 2.5 276 0.200
No 83 335 95% CI (1.1–5.7) 306

A R T I C LE
Age
Above median 121 0.499 363 0.341 306 0.434
Below median 79 424 302
Gender
Male 122 0.117 484 0.738 329 0.620
Female 61 424 210
Body weight
Above median 85 0.331 424 0.784 363 0.989
Below median 61 276 216
Multifocal
Yes 67 0.414 535 0.134 461 0.112
No 85 306 220
Trigonal
Yes 57 0.149 363 0.360 215 0.267
No 104 374 329
Diffuse 60 0.204 484 0.115 309 0.117
Polypoid 146 616 498
Ureteral involvement
Yes 168 168
No 79 0.819 432 0.266
Urethral involvement
Yes 125 363 363
No 77 0.895 484 0.066 306 0.407
Prostate involvement
Yes 125 329 0.257 270
No 77 0.873 484 363 0.377
Complete gross excision 85 424 276
Gross cytoreduction 72 0.379 616 0.487 287 0.948
Margins
Complete 70 0.558 329 0.340 248 0.296
Incomplete 57 484 242
Deracoxib 56 276 226 0.465
Piroxicam 104 0.521 424 0.793 364
HR — Hazard ratio; CI — Confidence interval.

All study dogs received chemotherapy using various agents as sequential doses every 21 d for 2 or 3 cycles (n = 4; 9%).
and protocols. The chemotherapy protocols used for both treat- Vinblastine/mitoxantrone with vinblastine was given on week 1,
ment groups are summarized in Table 2. First-line protocols mitoxantrone on week 2, and week 3 was off. This cycle was
were recorded for all 47 dogs. Twenty-nine dogs (62%) received repeated for 2 cycles (n = 1, 2%). Doxorubicin/carboplatin was
a single agent protocol consisting of either doxorubicin given given as sequential doses every 21 d for 3 cycles (n = 1, 2%).
once every 21 d for 4 to 6 treatments (n = 17; 36%), mito- Doxorubicin/­vinblastine/carboplatin/mitoxantrone was given as
xantrone given once every 21 d for 4 to 6 treatments (n = 6; sequential doses every 21 d for 6 cycles (n = 1, 2%). Cisplatin,
13%), vinblastine given once every 7 d for 5 or 6 treatments carboplatin, and gemcitabine/cyclophosphamide were admin-
(n = 4; 9%), or carboplatin given once every 21 d for 5 or istered with cisplatin and carboplatin given sequentially, then
6 treatments (n = 2; 4%). Eighteen dogs (38%) received multia- gemcitabine and oral cyclophosphamide given concurrently.
gent therapy. Combination mitoxantrone/doxorubicin was given These were administered every 21 d for 3 cycles (n = 1, 2%).
as alternating doses every 21 d for 2 or 3 cycles (n = 5; 11%). Upon confirmation of progressive disease, rescue protocols
Combination vinblastine/doxorubicin with vinblastine was utilizing oral cyclophosphamide (n = 2, 4%) and combination
given on week 1, doxorubicin on week 2, and week 3 was off. gemcitabine oral cyclophosphamide (n = 1, 2%) were instituted.
This cycle was repeated for a total of 4 to 6 cycles (n = 5; 11%). Twelve dogs (39%) in Group A and 2 dogs (13%) in Group B
Combination mitoxantrone/doxorubicin/carboplatin was given were censored in OS analysis due to being lost to follow-up.

CVJ / VOL 62 / FEBRUARY 2021 137


100 Surgery
90 Median OS = 498 days
100
Progression-free probability
80 Surgery No surgery
90
70 Median PFS = 85 days Median OS = 335 days
80

Survival probability
60 70 P = 0.026 HR = 2.5
No surgery 60
50 95% CI = 1.1–5.7
Median PFS = 83 days 50
40
40
30 P = 0.663
A R T I C LE

30
20 20
10 10
0 0
0 100 200 300 400 500 600 0 100 200 300 400 500 600
Days following treatment initiation Days following treatment initiation

Figure 1. Kaplan–Meier curve comparing the progression-free Figure 2. Kaplan–Meier curve comparing the overall survival
survival for dogs treated with partial cystectomy and medical for dogs treated with partial cystectomy and medical therapy
therapy (Surgery — solid line) versus those treated with medical (Surgery — solid line) versus those treated with medical therapy
therapy alone (No surgery — dashed line). Vertical tick marks alone (No surgery — dashed line). Hazard ratio (HR); 95% CI
indicate censored cases. (confidence interval). Vertical tick marks indicate censored cases.

Only 1 dog in Group A (3%) and no dogs in Group B were for dogs undergoing cytoreductive surgery (Group A) and dogs
lost to follow-up with respect to date of progression. The mean not undergoing surgery (Group B) were not different (85 d
and median times to censoring for dogs lost to follow-up in versus 83 d, P = 0.663; Figure 1). No variables were prognostic
Group A were 491 d (mean) and 364 d (median); and for for PFS (Table 2). The median OS for dogs undergoing cyto-
Group B were 447 d (mean) and 447 d (median). Twenty-seven reductive surgery (Group A) was greater than that for dogs not
dogs (87%) in Group A and all dogs in Group B died or were undergoing surgery (Group B) when lost to follow-up cases were
euthanized due to progression of disease. Dogs that were lost to censored (498 d versus 335 d, hazard ratio 2.5; 95% CI: 1.1 to
follow-up were presumed to have died from progressive disease 5.7; P = 0.026; Figure 2). There was no difference in PFS or OS
if tumor progression was confirmed at the date of last contact. between dogs with histologically complete versus histologically
Four dogs (13%) in Group A and no dogs in Group B died incomplete tumor resection (PFS 70 d for complete versus 57 d
or were euthanized due to causes unrelated to TCC. Causes for incomplete, P = 0.558 and OS 329 d for complete versus
of death in these 4 Group A dogs included acute collapse and 484 d for incomplete, P = 0.340; Table 3). No other prognostic
cardiopulmonary arrest during exercise, progression of dilated factors were identified.
cardiomyopathy, euthanasia due to progression of chronic renal Table 3 also reports OS time to event (TTE) with lost to
failure and abscessation of a cutaneous plasmocytoma, and follow-up (LTF) cases uncensored due to the high numbers of
severe cervical pain suspected to be secondary to intervertebral LTF in dogs in the surgery group (Group A). In this scenario,
disc disease. The dog that died of acute cardiopulmonary arrest the median OS for dogs undergoing cytoreductive surgery
was treated with doxorubicin; however, the total cumulative (Group A) was not different from that for dogs not undergoing
dose was 150 mm/m2. The dog that died of progressive dilated surgery (Group B) (276 d versus 306 d, P = 0.200; Figure 2).
cardiomyopathy had this diagnosis before being diagnosed with In Group B dogs, stable disease was noted in 10 dogs (63%)
TCC but was managed medically and was stable at the time of and progressive disease in 6 dogs (38%). No dogs in Group B
TCC diagnosis. This dog was not treated with doxorubicin but had a complete or partial response to therapy.
rather vinblastine. The dog that was euthanized due to progres-
sive chronic renal failure and abscessation of cutaneous plasmo- Discussion
cytoma was treated with the COX inhibitor piroxicam. At the Previous veterinary studies have formally evaluated effects of
time the cutaneous abscess was diagnosed, the owner of this dog surgery on outcomes for dogs with naturally occurring urinary
elected euthanasia rather than surgical treatment of the cutane- bladder TCC (13–19). However, to the authors’ knowledge,
ous abscess. All 6 dogs that had histologically complete tumor there are no studies comparing the outcomes of canine patients
excision died or were euthanized due to progression of disease. with urinary bladder TCC receiving surgery and adjunctive
medical therapy, compared to those receiving medical therapy
Outcome and prognostic factors alone, with case controls from the same time interval and
Disease progression was documented in all 47 dogs. Progression ­institutions. Our study population was similar with respect to
was confirmed via abdominal ultrasonography and characterized breed, gender, and clinical signs to previously reported popula-
by recurrence within the bladder lumen (n = 41), evidence of tions of dogs with urinary bladder TCC (2,3,19,20).
metastasis to sub-lumbar lymph nodes (n = 1), local extension In the study reported here, the median OS for dogs treated
into the prostate or urethra (n = 1), and abdominal wall seed- with partial cystectomy and adjuvant systemic chemotherapy/
ing (n = 1). COX inhibitors (Group A), was 498 d compared to 335 d for
The outcomes and potential prognostic variables for both dogs treated with systemic chemotherapy/COX inhibitors alone
treatment groups are summarized in Table 3. The median PFS (Group B) (P = 0.026). The median PFS of 83 d and median

138 CVJ / VOL 62 / FEBRUARY 2021


OS of 335 d for dogs in Group B (medical therapy only) were and functional impact differences in where the tumor arises
comparable to previously published survival times for dogs (and recurs). This latter alternative is supported by the fact that
treated with medical therapies alone (2,5,7,21). The OS of surgery did not improve PFS in our study. This implies, and is
498 d for dogs in Group A, seemed greater than the reported intuitive, that progression away from the trigone (and therefore
median OS of approximately 1 y for dogs undergoing partial away from ureters and urethra) is less likely to affect quality of
cystectomy, with or without adjuvant chemotherapy in other life and indeed, life-threatening outflow problems. In such a
reports (16,18,19). scenario, PFS duration that was not improved by surgery may

A R T I C LE
Knowing the substantial number of LTF cases in Group A not have as grave an impact on survival based on an anatomically
may have influenced outcomes, a TTE analysis was performed. more favorable location and therefore, that patient would live
In this analysis, median OS was not significantly different longer despite, not because of, surgery.
between the 2 groups. While censoring LTF cases likely arti- Similar to other reports (18,19), complete histologic excision
ficially prolonged the median OS for Group A, uncensoring was not prognostic in our study. This repeatable finding may
these cases for TTE calculation may have underestimated the be explained by the field cancerization effect within the bladder
median OS calculation. Although the exact dates of death were lining and/or the likelihood of TCC cell exfoliation and implan-
unknown, 11 (92%) of the dogs that were LTF in Group A had tation in other locations within the urinary tract.
ultrasonographic evidence of progressive disease and were alive Comparison of benefit achieved with the various chemo-
at the date of last contact, with median time for LTF of 491 d. therapy protocols and COX inhibitors used in this study was
This approaches the censored median OS of 498 d and it is not possible, as the agents used, dose intensity, and duration
reasonable to presume these patients subsequently died or were were not uniform. Given the findings supporting the theory of
euthanized due to their disease shortly thereafter. field cancerization effect, the exfoliative nature of this tumor,
In our study, the impact of extent of surgery was unclear, as locoregionally aggressive behavior, and considerable risk of
surgical dose (complete visual excision versus gross debulking) and metastasis with TCC, adjuvant chemotherapy should be con-
histologic completeness of excision were not significantly associ- sidered, even in cases of complete excision with no evidence of
ated with outcome. This finding was in contrast to that of Marvel metastasis (4). This recommendation is supported by the fact
et al (19), in which dogs that underwent full thickness cystectomy that 43 dogs (91%) in this study died or were euthanized due
had significantly improved PFI and median OS compared to dogs to progressive disease.
that had partial thickness tumor excision. That surgical dose is Serial abdominal ultrasound was used to monitor response
prognostic is also suggested by data reported by Knapp et al (4), to therapy and assess for progressive disease. In a recent study,
who noted improved median OS for patients under­going cyto- experienced observers using a standardized protocol (WHO
reductive surgery and adjuvant medical therapy (4). The lack of or RECIST criteria) reliably determined urinary bladder TCC
significance of surgical dose in the current study could be attrib- dimensions using 2D ultrasonography (22).
uted to inadequate power due to low case numbers. Intra-operative complications were not specifically reported in
Tumor location away from the trigone was not associated this study, but all dogs survived to discharge, and no dogs died
with an improvement in median OS (374 d, P = 0.360) (using due to peri-operative surgical or anesthetic complications. One
both the censored and uncensored data, Table 3). This was in dog developed tumor seeding to the abdominal wall associated
contrast to the study by Marvel et al (19) in which tumor loca- with the surgical scar; this represented 2.7% of cases undergoing
tion away from the trigone was a prognostic factor that signifi- partial cystectomy in our study, considerably lower than the 10
cantly improved survival. In our study, dogs with non-trigonal to 11% incidence reported by others (19,23)
tumors, were in fact, more likely to undergo surgery prior to Limitations of this study were primarily due to its retrospec-
medical therapy (74% versus 38%). The propensity for patients tive nature. These include the relatively small number of cases, a
with non-trigonal tumors to undergo surgery more commonly protracted case accrual period, lack of review by one pathologist,
than patients with trigonal tumors was likely explained by non-standardized follow-up schedule, and non-uniform report-
surgical decision-making, as full-thickness partial cystectomy ing of data in the medical record precluding accurate determina-
or total cystectomy for trigonal tumors is a more challenging tion of TNM stage and verification of tumor response using the
surgical procedure with higher morbidity than partial cystec- cRECIST criteria. The retrospective nature also did not allow for
tomy for tumors at non-trigonal locations. In a pilot study, total standardization of systemic therapy protocols. A further limita-
cystectomy with ureterocolonic anastomosis in 10 dogs with tion was the potential for case selection bias towards surgery for
TCC was associated with a poor prognosis and adverse effects dogs with non-trigonal tumors. This potential bias could mean
related to systemic uremia (15). Total cystectomy with vaginal these cases with non-trigonal tumors benefited the most from
or preputial ureteral diversion in 10 dogs yielded an MST of surgical treatment. Given the anatomy of the trigone region,
385 d and cutaneous ureteral diversion in 4 dogs was associated elimination of this bias in future studies would require investi-
with an MST of 279 d (13,14). These patients were persistently gation of non-trigonal tumors separate from trigonal tumors,
incontinent, making at home management more difficult and or further development of surgical techniques for neobladder
it is the authors’ opinion that these surgical approaches would in the dog.
be less acceptable to owners. In summary, dogs treated with partial cystectomy, systemic
An alternative explanation would be that surgery does chemotherapeutics, and daily COX inhibitors had an improved
not improve outcome, rather it is purely a result of anatomic median OS of 498 d compared to 335 d for dogs receiving

CVJ / VOL 62 / FEBRUARY 2021 139


medical therapy alone [hazard ratio: 2.5; 95% CI: 1.1 to 5.7; 12. Lee RK, Abol-Enein H, Artibani W, et al. Urinary diversion after radical
cystectomy for bladder cancer: Options, patient selection, and outcomes.
P = 0.026]. No prognostic factors were identified for PFS. The
BJU Int 2014;113:11–23.
findings of our study supported the recommendation for partial 13. Ricardo Huppes R, Crivellenti LZ, Barboza De Nardi A, et al. Radical
cystectomy with systemic chemotherapeutics and daily COX cystectomy and cutaneous ureterostomy in 4 dogs with trigonal tran-
sitional cell carcinoma: Description of technique and case series. Vet
inhibitors in the treatment of this disease. The current study also
Surg 2016;46:111–119.
supported the need for prospective, randomized controlled trials 14. Saeki K, Fujita A, Fujita N, Nakagawa T, Nishimura R. Total cystec-
evaluating dogs with primary urinary bladder TCC to determine tomy and subsequent urinary diversion to the prepuce or vagina in
A R T I C LE

dogs with transitional cell carcinoma of the trigone area: A report of


the most favorable protocol for adjuvant chemotherapy and the
10 cases (2005–2011). Can Vet J 2015;56:73–80.
role of partial cystectomy for the treatment of TCC. CVJ 15. Stone EA, Withrow SJ, Page RL, Schwarz PD, Wheeler SL, Seim 3rd
HB. Ureterocolonic anastomosis in ten dogs with transitional cell car-
References cinoma. Vet Surg 1988;17:147–153.
16. Norris AM, Laing EJ, Valli VE, et al. Canine bladder and urethral
1. Henry CJ. Management of transitional cell carcinoma. Vet Clin North
tumors: A retrospective study of 115 cases (1980–1985). J Vet Intern
Am Small Anim Pract 2003;33:597–613.
Med 1992;6:145–153.
2. Mutsaers AJ, Widmer WR, Knapp DW. Canine transitional cell carci-
17. Stone EA, George TF, Gilson SD, Page RL. Partial cystectomy for
noma. J Vet Intern Med 2003;17:136–144.
urinary bladder neoplasia: Surgical technique and outcome in 11 dogs.
3. Knapp DW, Ramos-Vara JA, Moore GE, Dhawan D, Bonney PL, Young
J Small Anim Pract 1996;37:1–6.
KE. Urinary bladder cancer in dogs, a naturally occurring model for
18. Robat C, Burton J, Thamm D, Vail D. Retrospective evaluation of
cancer biology and drug development. ILAR Journal 2014;55:100–118.
doxorubicin-piroxicam combination for treatment of transitional cell
4. Knapp DW, Glickman NW, DeNicola DB, Bonney PL, Lin TL,
carcinoma in dogs. J Small Anim Pract 2013;54:67–74.
Glickman LT. Naturally-occurring canine transitional cell carcinoma
19. Marvel SJ, Séguin B, Dailey DD, Thamm DH. Clinical outcome of
of the urinary bladder: A relevant model of human invasive bladder
partial cystectomy for transitional cell carcinoma of the canine bladder.
cancer. Uro Oncol 2000;5:47–59.
Vet Comp Oncol 2017;15:1417–1427.
5. Arnold EJ, Childress MO, Fourez LM, et al. Clinical trail of vinblastine
20. Knapp DW, McMillian SK. Tumors of the urinary system. In: Withrow
in dogs with transitional cell carcinoma of the urinary bladder. J Vet
SJ, Vail DM, Page RL, eds. Withrow and MacEwen’s Small Animal
Intern Med 2011;25:1385–1390.
Clinical Oncology. 5th ed. St. Louis, Missouri: Elsevier, 2013:572–582.
6. Boria PA, Glickman NW, Schmidt BR, et al. Carboplatin and piroxicam
21. Mcmillan SK, Boria P, Moore GE, Widmer WR, Bonney PL, Knapp
therapy in 31 dogs with transitional cell carcinoma of the urinary blad-
DW. Antitumor effects of deracoxib treatment in 26 dogs with transi-
der. Vet Comp Oncol 2005;3:73–80.
tional cell carcinoma of the urinary bladder. J Am Vet Med Assoc 2011;
7. Henry CJ, McCaw DL, Turnquist SE, et al. Clinical evaluation of mito-
239:13–18.
xantrone and piroxicam in a canine model of human invasive urinary
22. Honkisz SI, Naughton JF, Weng HY, Fourez LM, Knapp DW.
bladder carcinoma. Clin Cancer Res 2003;9:906–911.
Evaluation of two-dimensional ultrasonography and computed tomog-
8. Anderson CR, McNeil EA, Gillette EL, Powers BE, LaRue SM. Late
raphy in the mapping and measuring of canine urinary bladder tumors.
complications of pelvic irradiation in 16 dogs. Vet Radiol Ultrasound
Vet J 2018;232:23–26.
2002;43:187–192.
23. Higuchi T, Burcham GN, Childress MO, et al. Characterization and
9. Walker M, Breider M. Intraoperative radiotherapy of canine bladder
treatment of transitional cell carcinoma of the abdominal wall in dogs:
cancer. Vet Radiol Ultrasound 1987;28:200–204.
24 cases (1985–2010). J Am Vet Med Assoc 2013;242:499–506.
10. Withrow SJ, Gillette EL, Hoopes PJ, McChesney SL. Intraoperative
irradiation of 16 spontaneously occurring canine neoplasms. Vet Surg
1989;18:7–11.
11. Nolan MW, Kogan L, Griffin LR, et al. Intensity-modulated and image-
guided radiation therapy of treatment of genitourinary carcinomas in
dogs. J Vet Intern Med 2012;26:987–995.

140 CVJ / VOL 62 / FEBRUARY 2021

Vous aimerez peut-être aussi