Vous êtes sur la page 1sur 27

Annals of Diagnostic Pathology 10 (2006) 39 – 65

Review article
Advances in the pathologic diagnosis and biology of acute
myeloid leukemia
Sergej Konoplev, MD, PhD, Carlos E. Bueso-Ramos, MD, PhD4
Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4095, USA

Abstract In a general surgical pathology practice, cases of acute myeloid leukemia (AML), including myeloid
sarcoma, are relatively rare; the diagnosis is very often difficult, however, and consequences of a
missed or improper diagnosis compromise patient care. Currently, accurate diagnosis of every case of
AML requires integration of the morphological features and results of cytochemical and
immunohistochemical stains, flow cytometric immunophenotyping, cytogenetics, and molecular
studies. This review focuses on a practical approach to diagnosis of AML according to current
standard of practice and discusses some of recent changes in the field of AML.
D 2006 Elsevier Inc. All rights reserved.
Keywords: Acute myeloid leukemia; Myeloid sarcoma; Cytogenetics; Molecular

1. Introduction the very beginning, the diagnosis is delayed and


appropriate therapy is compromised.
1.1. Why should a surgical pathologist care about acute
3. The disease is encountered relatively rarely in
myeloid leukemia?
extramedullary sites compared with other entities,
Acute myeloid leukemia (AML), including extramedul- and there is a tendency to forget about the possibility
lary tumor referred to as myeloid (granulocytic) sarcoma of AML when dealing with a specimen outside the
(MS), represents a serious problem for a surgical pathologist bone marrow.
for several reasons, including the following: 4. Acute myeloid leukemia demonstrates a wide range
of morphological features, competing in extramedul-
1. Acute myeloid leukemia is a medical emergency,
lary sites with melanoma for the title bgreat
and delay in making a proper diagnosis jeopardizes a
mimicker.Q Therefore, the bclassical pictureQ of MS
patient’s life.
may not be present, and thus the diagnosis of MS is
2. The therapy for AML is not universal; specific
missed in most cases.
therapies are effective for one AML subtype and
totally inappropriate for another, such as all-trans- 1.2. Practical approach
retionic acid (ATRA) for t(15;17), and thus it is not
To assure the best patient care possible, the practical
sufficient to diagnose bAML.Q Acute myeloid
approach to pathologic diagnosis of AML is summarized in
leukemias with recurrent cytogenetic abnormalities
Fig. 1 and discussed here. This discussion is followed by a
such as t(15;17), t(8;21), or inv(16) present not
detailed description of the current World Health Organiza-
uncommonly as extramedullary tumors. Many
tion (WHO) classification of AML [1,2].
modalities used for precise classification (eg,
1. Consider MS in the differential diagnosis of poorly
cytogenetics, flow cytometry) require fresh tissue,
differentiated or undifferentiated tumors. The first step in
and if a possibility of AML is not considered from
recognizing any entity is to remember its possibility, and this
rule applies very well to MS. Good pathology practice advises
4 Corresponding author. Division of Pathology and Laboratory that MS, like melanoma, be put in the differential diagnosis
Medicine, Hematopathology Program, The University of Texas MD
Anderson Cancer Center, Houston, TX 77030-4095, USA. Tel.: +1 713
of every surgical case with poorly differentiated morphology.
792 6328; fax: +1 713 794 1800. 2. Review the patient’s medical history. Knowing that a
E-mail address: cbuesora@mdanderson.org (C.E. Bueso-Ramos). patient has AML usually alerts a pathologist to the
1092-9134/$ – see front matter D 2006 Elsevier Inc. All rights reserved.
doi:10.1016/j.anndiagpath.2005.10.001
40 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

possibility of MS, but no history of AML by no means specific for Ewing sarcoma; CD99 expression was detected
allows ruling out MS, as it may precede bone marrow in as many as 55% of MS cases and 43% of AML [3].
involvement. On the other hand, a report of hypoplastic 5. Be aware of hematopoietic growth factors. Hemato-
bone marrow (which may be diagnosed as aplastic anemia poietic growth factors (granulocyte colony-stimulating
elsewhere) gives an important hint, as MS may coexist with factor, granulocyte macrophage colony-stimulating factor,
hypoplastic bone marrow. erythropoietin, thrombopoietin) are potent stimulants of
3. Always make touch imprints. Touch imprint technique immature and mature hematopoietic cells, and changes
does not require much time or resources. Acute myeloid induced by these factors may be indistinguishable from a
leukemia blasts are recognized as such on Wright-Giemsa– picture of AML [4-6]. Be extremely cautious in diagnosing
stained slides based on the fine chromatin, distinct nucleoli, AML if hematopoietic growth factors were given to a
and cytoplasmic azurophilic granules that are positive for patient in the last 4 weeks. Moreover, tumors can produce
myeloperoxidase (MPO) or nonspecific esterase. Granulo- hematopoietic cytokines and mimic leukemias. Speak to the
cytic precursors, megakaryocytes, and normoblasts are also clinician, and if the patient’s clinical condition allows,
easily identified on touch imprints. Very often it is possible require another sample after a period without growth
to identify some unique morphological features of AML factor(s). It is also important to be aware of generalized
with recurrent cytogenetic abnormalities, which allows infections that might cause leukocytosis and a prominent
confirmatory ancillary studies to be expedited. Moreover, left shift, a clinical picture referred to as leukemoid reaction.
unstained touch imprint slides may be used for ancillary Information regarding prior chemotherapy is crucial for
studies such as cytochemical stains, fluorescence in situ making a diagnosis of therapy-related AML.
hybridization (FISH), or polymerase chain reaction (PCR). 6. Review the peripheral blood smear. A report by an
Pay attention to immature eosinophils and maturing outside facility cannot substitute for internal review. The
granulocytes because their presence is an important diag- degree of dysplasia can be subtle and is best identified on
nostic clue pointing toward MS. peripheral blood smear. The presence and percentage of blasts
4. Do not rely on a limited panel of immunostains. may be underestimated, especially in cases in which immature
Practically all immunostains have cross-reactivity with cells are blast equivalents rather than classic blasts. In these
different entities, and no tumor reacts with all antibodies cases, monoblasts and promonocytes can be misclassified as
with which it is said to react; therefore, a panel of antibodies monocytes and neoplastic progranulocytes as myelocytes.
is needed to confirm the diagnosis of AML. This is 7. Use a systematic approach and secure material for
particularly true for MS. The panel we use includes MPO, ancillary studies. See Fig. 1 for more information.
lysozyme, terminal deoxynucleotidyl transferase (TdT), 8. Use a systematic approach to the bone marrow sample.
CD34, CD43, CD45 (LCA), CD68, and CD117. Because Three important checkpoints include cellularity of aspirate
Ewing sarcoma enters the differential diagnosis of cases of smear, percentage of erythroid precursors, and percentage of
MS, it is important to realize that CD99 expression is not blasts (or blast equivalents). We routinely make several

Touch imprints1 Gross exam 10 buffered


(at least 6: formalin2
Wright-Giemsa,
5 unstained)

Cell suspension Cell suspension Frozen tissue at Fresh tissue for


in 10 RPMI in 10 RPMI –80oC (at least molecular
medium for medium for 0.5x0.5x 0.5cm)5 study5
Flow cytometry3 cytogenetics4

Fig. 1. Diagram of workflow of a specimen suspected to be AML. 1Touch imprints can be used for cytochemical (MPO, butyrate), immunofluorescence
(TdT, POD test for AML M3), FISH, and molecular studies (cell scraping); 2Formalin fixed, paraffin-embedded tissue can be used for
immunohistochemical, DNA- and RNA-based molecular studies (T-cell receptor beta and gamma, IgH, JAK-2), and FISH; 3Some institutions have
adopted a process and hold policy when cells are processed immediately, but the actual immunophenotypical analysis is held waiting for a pathologist
decision. This approach secures a specimen and avoids unjustified flow cytometric studies; 4Conventional cytogenetics is the only way to obtain a full
karyotype, while all other techniques simply answer the question of whether a particular abnormality is present or absent. It does require viable cells; 5Fresh
and frozen tissue may be used for RNA and DNA extraction. RNA is used for RT-PCR tests for the t(15;17), t(8;21), inv(16), and t(9;22) abnormalities;
DNA for FLT3 and RAS mutation studies, T-cell receptor beta and gamma, IgH, and JAK-2. Some institutions prefer paraffin-embedded tissue to fresh or
frozen tissue for molecular studies.
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 41

touch imprints of a bone marrow biopsy specimen and use after their exposure [15,16]. Therapeutic radiations appear
them for differential count if the aspirate smear is hypo- to impart minimal risk, but the risk may be significant if it
cellular or of poor quality. If erythroid precursors dominate was administered with concurrent chemotherapy with an
( N50% of all nucleated cells), the blast percentage is alkylating agent.
calculated on the basis of all non-erythroid cells. A similar There are two types of chemotherapy-related AML. The
approach is used in rare cases of bone marrow involvement bclassicQ alkylating agent type (eg, cyclophosphamide,
by another malignant process (eg, chronic lymphocytic melphalan, nitrogen mustard) has a latency period of 4 to
leukemia), and blast percentage is reported as a fraction of 8 years and often is associated with abnormalities of
cells not involved in the known process. Otherwise, blast chromosome 5 and/or chromosome 7 [17,18]. The other
count is reported as a fraction of all nucleated cells. type, associated with exposure to agents that inhibit the DNA
repair enzyme topoisomerase II (eg, etoposide, teniposide),
1.3. Background
has a shorter latency period, usually 1 to 3 years, and is
Acute leukemias are clonal malignant disorders resulting associated with chromosome 11q23 abnormalities [19].
from genetic alterations in hematopoietic stem cells [7]. Drugs such as chloramphenicol, phenylbutazone, chlo-
These alterations limit their ability to differentiate into roquine, and methoxypsoralen can result in bone marrow
erythrocytes, granulocytes, and platelets and lead to the damage that may later evolve into AML. Exposure to
proliferation of bleukemicQ cells or bblasts.Q Acute myeloid benzene, a ubiquitous solvent used in several industries,
leukemia, also referred to as acute nonlymphocytic leuke- cigarettes, dyes, herbicides, and pesticides, has been
mia, is a heterogeneous group of disorders [1,2]. implicated as another potential risk factor [20,21].
Approximately 11 000 cases of AML are diagnosed Acute myeloid leukemia may also be secondary to the
annually in the United States. The overall annual incidence progression of a myelodysplastic process or due to
of AML is 3.4 per 100 000 populations [8]. Although progression of a chronic bone marrow bstem cellQ disorder,
leukemia is the most common malignancy of children such as polycythemia vera, chronic myelogenous leukemia,
15 years and younger, most cases are diagnosed in older or paroxysmal nocturnal hemoglobinuria.
adults. The median age at presentation for AML is 68 years
1.4. Clinical features
[8]. The incidence of AML increases with age being less
than 1 per 100 000 per year for persons younger than The proliferation of abnormal leukemic cells and their
30 years and 17 per 100 000 for persons 75 years of age [8]. impact on normal hematopoiesis lead to the symptoms and
The incidence of AML is higher in males than in females signs seen in AML.
and in whites than in African Americans [8,9]. The Fatigue, bruising or bleeding, fever, and infection reflect
incidence of AML is increasing in the elderly. Several a state of bone marrow failure. Although the term leukemia
factors including improved diagnosis and longer life may suggest marked elevations in white blood cells counts,
expectancy, resulting in increased environmental exposures, pancytopenia is more common. More than 95% of patients
are probably responsible for this increase [10]. Acute in whom AML is diagnosed have a hemoglobin level of less
myeloid leukemia accounts for less than 15% of cases of than 12 g/dL. Only 10% of patients with newly diagnosed
leukemia in children younger than 10 years and 25% to 30% AML present with leukocyte count greater than 100 000/lL
of cases between 10 and 15 years of age [8,11]; in adults, [22] and are at higher risk of tumor lysis syndrome and
AML accounts for 80% to 90% of cases of acute leukemia leukostasis. Tumor lysis syndrome can be due to spontane-
[8]. The incidence of leukemia, along with myelodysplasia, ous or treatment-mediated cell destruction [23,24]. It is
appears to be rising, particularly in the population older than characterized by hyperuricemia, renal failure, acidosis,
60 years [10]. In adults, AML is by far the most common hypocalcemia, and hyperphosphatemia [23,24]. Leukostasis
type of acute leukemia. The incidence of AML is slightly may manifest as dyspnea, chest pain, headaches, altered
higher in males and in populations of European descent [9]. mental status, cranial nerve palsies, or priapism [25,26].
Recent reports indicate that acute promyelocytic leukemia Leukostasis and tumor lysis syndrome are both oncologic
(APL), a distinct subtype of AML, is more common among emergencies and require prompt recognition and manage-
certain populations of Hispanic background [12]. ment. Disseminated intravascular coagulation is most
An increased incidence of AML is seen in patients common in cases with the t(15;17) [27]. Disseminated
involving certain disorders with excessive chromatin fragil- intravascular coagulation is typically seen at presentation or
ity, such as Bloom syndrome, Fanconi anemia, and during induction chemotherapy [28,29] and manifested by
Kostmann syndrome, and in individuals with Wiskott- diffuse oozing of blood with thrombocytopenia, hypofibri-
Aldrich syndrome or ataxia-telangiectasia. Other syndromes nogenemia, elevated fibrin split products, and deficiency of
such as Down (chromosome 21 trisomy), Klinefelter (XXY coagulation factors [30].
and variants), and Patau (chromosome 13 trisomy) have also Physical findings other than bleeding and related to
been associated with AML [13,14]. infection may include organomegaly, sternal tenderness,
Survivors of the atomic bombs in Japan had an increased retinal hemorrhages, and infiltration of gingivae, skin, soft
incidence of myeloid leukemias that peaked 5 to 7 years tissues, or meninges (more common with monocytic
42 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

Neutropenia is common in patients with MS; the marrow


may be hypocellular and often, but not invariably, reveals
increased blasts (Fig. 2). Myeloid sarcoma may be present at
diagnosis of AML or may precede the diagnosis. In
children, the presence of MS involving sites other than skin
was an independent favorable prognostic factor compared

Fig. 2. Hypocellular bone marrow. A patient presented with pancytopenia,


and a bone marrow specimen was hypocellular with no evidence of AML.
The patient was treated with antithymocyte globulin, cyclosporine,
granulocyte colony-stimulating factor, and erythropoietin without re-
sponse. One year later, a mass was detected in the psoas and iliac
region, and biopsy revealed MS. A bone marrow biopsy (shown here)
was obtained at that time; extensive workup including immunohisto-
chemical staining with MPO, lysozyme, CD34, and CD117 demonstrated
no evidence of AML.

variants M4 or M5 French-American British Cooperative


Group [FAB] classification) [31].
Myeloid sarcoma occurs in 2% to 14% of cases of AML,
more frequently in children than in adults [32- 40]. Myeloid
sarcoma most often involves soft tissues, skin, bone
(including periosteum), lymph nodes, and/or the orbit and
paranasal sinuses, but it has been reported in virtually all
anatomic locations [32-40]. Testicular mass is less common
than in ALL; to date only 37 cases have been reported [36].

Table 1
WHO classification of Acute Myeloid Leukemia (AML)
1. AML with recurrent cytogenetic abnormalities
AML with t(8;21)(q22;q22), (AML1/ETO)
AML with inv(16)(p13q22) or t(16;16)(p13;q22), (CBFb/MYH11)
APL (AML with t(15;17)(q22;q21), (PML/RARa and variants)
AML with 11q23 (MLL) abnormalities
2. AML with multilineage dysplasia
With prior MDS
Without prior MDS
3. AML and MDS, therapy related
Alkylating agent related
Topoisomerase II inhibitor related
4. AML, not otherwise categorized
AML, minimally differentiated
AML without maturation
AML with maturation
Acute myelomonocytic leukemia
Acute monoblastic/acute monocytic leukemia
Acute erythroid leukemia (erythroid/myeloid and pure erythroleukemia)
Acute megakaryoblastic leukemia Fig. 3. Myeloid sarcoma involving the myometrium. Note that
Acute basophilic leukemia neoplastic cells are streaming along muscle fascicles. (A) Hematoxylin
Acute panmyelosis with myelofibrosis and eosin (H&E); (B) CD117 immunohistochemical stain; (C) lysozyme
Myeloid sarcoma immunostain.
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 43

with patients with MS involving skin or patients without collection. If it is not possible to extract DNA and RNA
MS [39]. In adult patient population, patients with MS fare immediately, fresh tissue (at least 0.5  0.5  0.5 cm) should
worse compared with patients without MS [38]. be frozen for DNA and RNA studies.
Bone marrow trephine biopsy tissues are most com-
1.5. Handling of the bone marrow or peripheral blood
monly fixed in 10% buffered formalin, decalcified in 5%
specimen
formic acid for 12 hours, embedded in paraffin, and cut
Specimens of every case of AML must be handled into sections 2 lm thick. If the bone marrow aspiration is
properly to optimize the diagnostic yield of the ancillary not successful (most often because of packed or fibrotic
tests. The specimens are most commonly peripheral blood bone marrow), certain alternative approaches may be taken
or bone marrow aspirates. A bone marrow core biopsy with to optimize the results. Touch imprints of the core biopsy
touch imprints and bone marrow aspirate clots also should are invaluable for cytological examination in this setting
be obtained in all newly diagnosed cases. Specimens and are also a useful source of materials for cytochemical,
intended for flow cytometry should be kept in test tubes immunocytochemical, and FISH studies. A variety of
containing EDTA. A sterile sample for cytogenetic analysis cytogenetic defects can be evaluated by loss of heterozy-
is collected with sodium heparin as the anticoagulant. gosity studies. These studies have an advantage over
Samples for cytogenetics are cultured immediately or kept conventional cytogenetic studies in that archival tissues
at 48C for culture within 24 hours of collection. Specimens may be used. Allelic losses are more common in extra-
kept at room temperature are acceptable for flow cytometric medullary sites (eg, spleen) than in bone marrow [42].
analysis for up to 3 days. Immunohistochemical analysis applied to a paraffin-
For molecular studies, a variety of specimens can be used, embedded biopsy specimen is a useful alternative immuno-
depending on which specific molecular tests are required. phenotyping technique because many cell lineage–specific
For instance, detection of monoclonal rearrangements of the markers that are resistant to routine fixation and processing
T-cell receptor gene and the immunoglobulin heavy chain have become available. These markers are described in
gene through PCR assays can be achieved with fresh tissues, detail later in this article.
air-dried smears, Wright-Giemsa–stained smears, and for-
1.6. Classification of AML
malin-fixed, paraffin-embedded tissues. Because heparin
interferes with the results of PCR-based molecular studies, The current WHO classification of acute leukemias and
EDTA is preferred in this situation and the samples should be myelodysplastic syndrome (MDS) has evolved away from
kept at 48C. RNA must be extracted within 24 hours after the FAB classification, which is based on morphology, to

Fig. 4. Acute myeloid leukemia involving the uterine cervix. (A) Low power demonstrates an ill-defined infiltrate extending to the surface of a vaginal cuff. (B)
High power shows blasts with monocytoid nuclei and mitoses. (C) CD68 staining. (D) Lysozyme immunostain.
44 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

include not only morphology, but also clinical, immuno-


phenotypic, and cytogenetic features [1,2,43]. Besides
evaluation of percentage of immature cells and their
morphological, cytochemical, and immunophenotypic char-
acteristics, it is necessary to evaluate residual hematopoiesis
for the presence and degree of dysplasia, obtain the patient’s
history of chemotherapy, and learn from cytogenetic and/or

Fig. 6. Myeloid sarcoma involving the skin. The patient presented with a
scalp lesion diagnosed as MS. Extensive workup, including a bone
marrow biopsy, reveals no evidence of disease. The patient opted to wait
and, 3 months later, developed full-blown AML M5. (A) Myeloid
sarcoma in scalp (H&E, 20); (B) blasts showing monocytoid nuclei and
cytoplasm (H&E, 4000); (C) lysozyme immunostain (40).

molecular studies whether a patient has a recurrent


Fig. 5. Acute myeloid leukemia with inv(16) presented as lymphadenop- cytogenetic abnormality.
athy. (A-B) A lymph node ([A] 20; [B] 1000). Note the interfollicular
pattern of involvement by blasts and prominent mitotic activity with tingle-
The current WHO classification of AML includes
body macrophages creating a starry-sky pattern. (C) Bone marrow (400) 4 categories: AML with recurrent cytogenetic abnormalities,
demonstrating sheets of blasts admixed with eosinophils. AML with multilineage dysplasia, therapy-related AML and
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 45

MDS, and AML not otherwise specified (NOS). Acute


myeloid leukemia NOS categories are usually reported in
terms of the FAB AML classification (Table 1).

2. Morphological features of AML


Acute myeloid leukemia comprises a group of diseases
whose cell morphology sometimes differs significantly from
one group to another. Therefore, common morphological

Fig. 8. Acute myeloid leukemia without maturation (AML M1 by FAB).


(A and B) hypercellular bone marrow with increased blasts; (C) immature
blasts with fine chromatin and high nucleus-cytoplasm ratio.

features of AML and special features of particular subtypes


are discussed here.
Fig. 7. Acute myeloid leukemia appearing as sarcoma in liver and lung. (A) 2.1. Myeloid sarcoma
Hypercellular bone marrow with sheets of immature cells and residual
hemopoiesis (H&E, 400); (B-C) liver biopsy with neoplastic spindle- The tumor usually represents a diffuse infiltrate
shaped cells ([B] H&E, 40; [C] H&E, 400). composed of a relatively uniform population of immature
46 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

cells, which may include all stages of myeloid differen- as 40 per 10 high-power fields. In cases with high
tiation or be predominantly blastic such that few or no mitotic activity, tingle-body macrophages may be seen
cells have recognizable granulocytic differentiation. In giving a bstarry-skyQ appearance to a section (Fig. 5).
some anatomic sites, such as myometrium, neoplastic Single-cell necrosis (apoptotic bodies) may be identified in
cells may stream along muscle fibers (Fig. 3), whereas in poorly differentiated and blastic neoplasms. Geographic
other sites, neoplastic cells may invade local structures areas of necrosis may be observed in some cases. In some
such as glands (Fig. 4). The presence of immature cases, marked desmoplastic reaction together with promi-
eosinophils and maturing granulocytes is an important nent cellular atypia gives a sarcoma-like appearance to
diagnostic clue, especially in patients with no known AML. MS (Fig. 7).
Neoplastic cells are of moderate size with irregularly In the past, MS was subclassified on the basis of degree
shaped, angulated nuclei, irregular nuclear contours, of differentiation into blastic, immature, and differentiated
and fine chromatin (Fig. 5). The neoplastic cells may have [33,41]. It is important to keep in mind that, in cases of MS
folded nuclear contours with smaller nucleoli and more with bone marrow involvement, the morphology of the
delicate chromatin suggestive of monocytic differentiation blasts composing MS may differ from that of the blasts in
(Figs. 4, 6). Mitotic figures may be sparse or as frequent bone marrow.

Fig. 9. Morphological features of AML with the t(8;21) abnormality. (A) Hypercellular bone marrow with residual hematopoiesis. (B) Sheets of immature cells
with fine chromatin and 2 to 3 nucleoli form a solid sheet in the center and are intermixed with residual mature cells (left upper corner). (C-D) Immature blasts
with fine chromatin, 2 to 3 nucleoli, and high nucleus-cytoplasm ratio are intermixed with mature granulocytes. Long, slender Auer rod is present. (E)
Myeloperoxidase is positive in some blasts (MPO stain with Wright-Giemsa counterstain).
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 47

2.2. Peripheral blood, bone marrow aspirate, and touch similar to type I with the addition of up to 20 delicate
imprint azurophilic granules in the cytoplasm (Fig. 9). The type III
myeloblast has numerous azurophilic granules in the
2.2.1. Blasts cytoplasm. Distinction between the type III blast and
By the FAB criteria, acute leukemia is diagnosed when promyelocyte is somewhat arbitrary and may be impossible.
a 200-cell differential reveals the presence of more than One characteristic feature of myeloblasts in AML is the
30% blasts in a marrow aspirate. The WHO changed this presence of Auer rods, which are seen in 60% to 70 % of all
minimum criterion to 20%. cases and represent abnormal azurophilic granules. In some
In addition, patients with the clonal, recurring cytoge- cases, leukemic cells contain numerous Auer rods and are
netic abnormalities t(8;21)(q22;q22), inv(16)(p13q22) or referred to as bfaggot cellsQ (Fig. 10). The faggot cells are
t(16;16)(p13;q22), and t(15;17)(q22;q21) should be consid- typical, but not unique, for cases with the recurring
ered to have AML regardless of the blast percentage. If the cytogenetic abnormality t(15;17)(q22;q21). The long, slen-
marrow contains more than 50% normoblasts and pronor- der Auer rods with tapered ends are typical for cases with the
moblasts, the blast percentage is based only on the non- recurring cytogenetic abnormality t(8;21)(q22;q22) (Fig. 9).
erythroid cells; in these cases, the diagnosis is typically The monoblast has a round, sometimes folded nucleus,
acute erythroid leukemia, which can be confirmed if finely dispersed nuclear chromatin, variably prominent
glycophorin A is expressed on the blasts’ surface. nucleoli, abundant slightly basophilic cytoplasm with fine
In AML, blasts are classified as myeloblasts, monoblasts, granulation, and occasional vacuoles (Figs. 11, 12).
erythroblasts, or megakaryoblasts. The erythroblast has a round nucleus, slightly condensed
Three major types of myeloid blast are reported in the nuclear chromatin, variably prominent nucleoli, and a
literature. The type I myeloblast has fine nuclear chromatin, moderate amount of deeply basophilic cytoplasm (Fig. 13).
2 to 4 distinct nucleoli, and a moderate rim of pale to The megakaryoblasts vary significantly in their appear-
basophilic cytoplasm without azurophilic granules (Fig. 8). ance from one case to another, ranging from undifferentiated
The type II myeloblast has nuclear and cytoplasmic features blasts to mature-appearing megakaryocytes (Fig. 14). In

Fig. 10. Acute myeloid leukemia with the (15;17). (A) Hypercellular bone marrow. (B) High-power microscopy demonstrates immature cells with fine
chromatin and abundant pink cytoplasm. (C) Cell with numerous Auer rods referred to as faggot cell. (D) Myeloperoxidase is positive in all neoplastic
progranulocytes (compare with MPO positivity of AML M2).
48 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

Fig. 11. Morphological features of AML M4. (A) Hypercellular bone marrow. (B-C) Mixture of typical myeloblasts with monocytoid cells with twisted nuclei
and more abundant cytoplasm. (D-E) Some neoplastic cells are positive for MPO and some are positive for butyrate esterase, respectively.

some cases, the full range is present, whereas in others, there commonly in cases of AML with monocytic differentia-
is a predominance of a particular type. Cells are of moderate tion, especially cases with the translocation t(8;16). Some
size, with dispersed nuclear chromatin, distinct nucleoli, and myeloblasts have prominent vacuolization with an appear-
a moderate amount of light basophilic cytoplasm, with very ance similar to the L3 blasts of ALL (Fig. 15).
fine azurophilic granules frequently located to the perinu-
clear zone. The cytoplasm may be irregular and show 2.2.2. Blast equivalents
pseudopod formation (Fig. 14). To fulfill the diagnostic criterion of 20% blasts set
In some cases of AML, blasts exhibit any of several for AML, in some cases it is necessary to combine brealQ
unusual morphological features. Marked nuclear lobulation blasts with so-called blast equivalents. The term bblast
is seen in cases with the recurring cytogenetic abnormality equivalentsQ is specific to a particular type of AML. The
t(15;17)(q22;q21). Multinucleation is seen in many types neoplastic promyelocyte considered a blast equivalent
of blasts, especially erythroblasts and megakaryoblasts. in cases with the recurring cytogenetic abnormality
Pseudo-Chédiak-Higashi-type giant granules result from t(15;17)(q22;q21), or AML M3, has an eccentric, often
fusion of primary granules and are occasionally seen in folded and lobulated nucleus with slightly condensed
leukemic blasts and granulocytic precursors, especially in nuclear chromatin, intense cytoplasmic granularity, and an
cases with t(8;21). Erythrophagocytosis is seen most apparent Golgi zone (Fig. 10). The promonocyte considered
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 49

Fig. 12. Morphological features of AML M5. (A) Hypercellular bone marrow with osteosclerosis. (B-C) Neoplastic cells have twisted nuclear shapes with
irregular nuclear contour, indentations and grooves, and abundant eosinophilic cytoplasm. (D-E) Cells are negative for MPO and strongly positive for
butyrate esterase, respectively.

a blast equivalent in cases of AML with monocytic present in at least 50% of cells of at least 2 lines. The
differentiation (Fig. 12). criteria for dysplasia are as follows:
! granulocytic dysplasia: agranular or hypogranular
2.2.3. Dysplasia in myeloid cells
polymorphonuclear cells, hyposegmented nuclei
In addition to evaluating blasts and blasts equivalents,
(pseudo Pelger-Huet), or pseudo-Chédiak-Higashi-
pay close attention to cells appearing as mature or
type giant granules;
maturing for several reasons. These cells may be part of
! megakaryocytic dysplasia: micromegakaryocytes,
a neoplastic clone and demonstrate unusual morphological
multiple separated nuclei, or large mononuclear forms;
features, such as giant neutrophils or giant platelets, or
! erythroid dysplasia: karyorrhexis, megaloblastoid
contain Auer rods. Chunky basophilic granules in maturing
aspect, multinuclearity, nuclear fragments, nuclear
eosinophilic precursors give a very important hint of AML
budding, internuclear bridging, cytoplasmic vacuoli-
with recurring cytogenetic abnormalities inv(16)(p13q22)
zation, or aberrant periodic acid–Schiff (PAS) posi-
or t(16;16)(p13;q22) (Fig. 16). Significant dysplasia may
tivity (Fig. 16C).
be a hint of a history of previous chemotherapy and, in the
absence of such a history, warrants classification of the 2.2.4. Dysplasia using flow cytometry immunophenotyping
case as AML with multilineage dysplasia (Fig. 17). Dysplastic features can be assessed by using flow
Dysplastic features are also seen in cases of therapy- cytometry immunophenotyping [44]. The proposed criteria
related AML (Fig. 18). By definition, the dysplasia in for myeloid cells are abnormal granularity, abnormal decrease
AML with multilineage dysplasia must be severe, that is, in CD45 expression, presence of HLA-DR or lack of CD11b,
50 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

Fig. 13. Morphological features of AML M6. (A) Hypercellular bone marrow with marked dysplastic erythroid and megakaryocytic hyperplasia; (B) aspirate
smear. Note deep blue cytoplasm and presence of cytoplasmic vacuoles. (C) PAS positivity in neoplastic erythroid precursors. (D) Iron stain shows a ringed
sideroblast.

abnormal relationship between CD13 and CD16, expression granular appearance (Fig. 10). In some cases with recurring
of CD56 on a subpopulation of myeloid cells, lack of CD33 cytogenetic abnormality inv(16)(p13q22) or t(16;16)
expression, asynchronous shift to the left, presence of (p13;q22), increased and/or abnormal eosinophilic precur-
lymphoid antigens, or CD34 on myeloid cells [44]. For sors are apparent on bone marrow biopsy or clot sections
monocytic cells, the abnormalities include abnormal granu- (Fig. 16). In cases with monocytic differentiation, blasts are
larity, abnormal CD11b or HLA-DR expression, loss of CD13 large and have abundant amphophilic to lightly eosinophilic
or CD16, presence of CD56, lack of CD33 or CD14, presence cytoplasm and round to oval, sometimes very convoluted
of CD34, or lymphoid antigen expression on monocytes [44]. (bmonocytoidQ) nuclei with single or multiple, usually
eosinophilic nucleoli, which may be very prominent (Fig.
2.3. Bone marrow biopsy and clot section
12). In cases with erythroid differentiation, erythroblasts
The bone marrow sections in AML are generally have abundant basophilic cytoplasm and nuclei with distinct
hypercellular and in most cases are extensively replaced nucleoli and nuclear chromatin, which is slightly coarser
by leukemic cells. Approximately 5% of adult AML cases than that of myeloblasts. In cases with megakaryocytic
present with hypocellular bone marrow, which may pose a differentiation, megakaryoblasts may demonstrate abnor-
diagnostic challenge. Another diagnostic challenge is focal mal differentiation, forming micromegakaryocytes in small
involvement by leukemia with a significant proportion of clusters or broad sheets (Fig. 14). Micromegakaryocytes
sections spared. Immunohistochemical stains for MPO, show nuclear cytoplasmic asynchrony characterized by fine
CD34, and CD117 are helpful to highlight the few foci of nuclear chromatin and a rim of eosinophilic cytoplasm.
myeloblasts (see bImmunohistochemistryQ section). There may be increased reticulin fibers.
Myeloblasts may be distributed uniformly throughout the
bone marrow interstitium or may be present in a large focal
3. Cytochemical features of AML
aggregate. In most cases, blasts are relatively uniform and
have a round to oval, sometimes indented nucleus, 1 to 4 An important aspect of the classification of acute
variably prominent nucleoli, and a moderate amount of leukemia is the cytochemical reactivity pattern of the blasts.
cytoplasm. In some cases, as in those with the recurring Cytochemical stains include MPO (or, to less extent, Sudan
cytogenetic abnormality t(15;17)(q22;q21), blasts have black B), nonspecific esterase (a-naphthyl butyrate or
abundant, deeply eosinophilic cytoplasm with distinctly a-naphthyl acetate), and PAS. Some laboratories also use
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 51

Fig. 14. Morphological features of AML M7. (A-B) Bone marrow biopsy demonstrates hypercellular marrow with a mixture of immature cells and increased,
clustering megakaryocytes at different stage of differentiation ([A] 200; [B] 500). (C-D) Blasts are medium sized with basophilic cytoplasm forming
characteristic projections. (E) Electron micrographs showing cytoplasmic projections. Note the platelet peroxidase reaction product deposited in the perinuclear
envelope of the blast.

naphthol ASD chloroacetate esterase. If 3% or more of the 4. Ancillary laboratory investigation


blasts stain positive for MPO or Sudan black B SBB, the
4.1. Flow cytometry
diagnosis is AML. If the blasts are MPO negative, but stain
for butyrate or nonspecific esterase, acute monocytic Immunophenotyping by use of flow cytometry has
leukemia (a variant of AML) is diagnosed. dramatically improved the accuracy of diagnosis of AML.
A minority of MPO-negative and Sudan black B– It is strongly recommended, however, that immunologic
negative cases are AML, and may include minimally marker study results be interpreted in conjunction with
differentiated leukemia (M0), acute monocytic leukemia morphological studies. Only the typical immunophenotypic
(M5), and megakaryocytic leukemias (M7) that require features of AML and some of the pertinent information
electron microscopy study (Figs. 14, 19) or flow cytometry related to the differential diagnosis of AML are presented
immunophenotypic analyses (Figs. 20, 21) for characteriza- here. Various T-cell, B-cell, and myeloid-associated markers
tion [45]. are available for flow cytometric immunophenotypic anal-
52 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

Fig. 15. Acute myeloid leukemia M4 with blasts containing numerous vacuoles. (A) Peripheral blood smear; (B-D) bone marrow aspirate smear.

ysis. Table 2 summarizes the immunophenotypic profile of 3. erythroid-associated markers: hemoglobin A, glyco-
immature myeloid cells. Table 3 shows the European Group phorin A;
for the Immunological Classification of Leukemias (EGIL) 4. megakaryocyte-associated markers: factor VIII,
scoring system of lineage specificity for different markers CD41, CD61;
proposed in 1995, later revised in 1998 [46,47]. For 5. T cell–associated markers: CD3, CD5, CD4, CD8;
instance, two markers, CD3 and cytoplasmic MPO, are 6. B cell–associated markers: CD20, CD22, CD79a,
highly specific for T-cell and myeloid-cell lineages, respec- Pax-5, immunoglobulin kappa and lambda, CD10.
tively. Myeloid antigens are CD13, CD33, CD117, CD14,
In cases of MS without previous or concurrent AML,
CD64 (the latter two are monocytic markers), glycophorin A
CD45 antibody is useful to establish the hematopoietic
(an erythroid marker), and CD41 (a megakaryocytic
origin of the tumor.
marker). Nevertheless, the specificity of these markers is
not absolute.
4.1.2. Electron microscopy
Flow immunophenotypic displays of two typical AML
With improvements in laboratory investigations, the role
cases are illustrated in Figs. 20, 21. For more details on flow
of electron microscopy has diminished. Demonstration of
cytometric immunophenotyping in leukemia, please refer to
platelet peroxidase is useful in documenting the diagnosis of
appropriate review articles [48-50].
acute megakaryocytic leukemia (M7). Characteristically, the
platelet peroxidase reaction product is deposited in the
4.1.1. Immunohistochemistry
perinuclear envelope and endoplasmic reticulum, but not in
The antigen retrieval technique allows routinely fixed,
the Golgi regions (Figs. 14, 19).
paraffin-embedded bone marrow biopsy sections to be
used for antigenic characterization. When fresh bone 4.2. Cytogenetic and molecular markers
marrow aspirate and peripheral blood samples are not
Table 4 summarizes frequency of recurrent chromo-
available, this method is useful in classifying acute
somal abnormalities in pediatric and adult patient pop-
leukemias. The following markers are often included in
ulations. Molecular study of many recurring cytogenetic
the panel:
abnormalities has revealed genes that may be involved in
1. progenitor-associated markers: CD34, TdT; leukemogenesis. A well-accepted concept states that, for
2. myeloid-associated markers: MPO, lysozyme; an acute leukemia to develop, at least two broad mutations
CD117, CD68, CD15, CD33; are necessary, referred to as class I and class II mutations.
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 53

fusion of the CBFb (CBFB) gene on the q arm and the


myosin heavy chain (MYH11) gene on the p arm. The 8;21
translocation involves the CBFa (CBFA) gene on chromo-
some 21, called the AML1 (also RUNX1) gene, joining the
ETO gene on chromosome 8. Like the t(15;17) gene
product, the AML1/ETO protein acts to block transcription
of CBFA-CBFB–controlled genes.
Another example of class II mutation is involvement of
the myeloid-lymphoid (or MLL) gene located on 11q23. The
MLL gene has two regions that encompass multiple zinc
fingers and at least two additional potential DNA-binding
motifs. Abnormalities in the MLL gene are relatively common
in patients with AML who do not have 11q23 rearrangements
on cytogenetic analysis. MLL gene locus at 11q23 is
involved in more than 30 leukemia-associated translocations
[52,53]. The most common translocation is t(4;11)
(q21;q23), which gives rise to a chimeric MLL/AF4 fusion
gene. Prognosis of AML with 11q23 rearrangements is poor.
The t(15;17) translocation does not fit readily into the
class I-class II mutation concept, as it has features of both. It
encodes a chimeric protein, PML/RARa, which is formed
by the fusion of the retinoic acid receptor-a (RARa) gene
from chromosome 17 and the promyelocytic leukemia
(PML) gene from chromosome 15. The RARa gene encodes
a member of the nuclear hormone receptor family of
transcription factors. After binding retinoic acid, RARa
can promote expression of a variety of genes. The t(15;17)
translocation juxtaposes PML with RARa in a head-to-tail
configuration that is under the transcriptional control of
PML. The PML-RARa fusion protein tends to suppress
gene transcription and blocks differentiation of the cells.
Pharmacological doses of the RARa ligand ATRA (tretinoin)
relieve the block and promote differentiation.
The presence of the t(15;17) also may be detected by a
unique immunofluorescence method using antibody specific
for promyelocytic leukemia (PML) protein. Normally, PML
is compactly located at multiple nuclear domains known as
PML oncogenic domains (PODs), whose structures are dyna-
mically regulated and disrupted in t(15;17) cases (Fig. 22).
Besides conventional cytogenetic studies, FISH may
be used to detect the above-mentioned abnormalities
(Figs. 23, 24), as may reverse transcriptase–PCR. Both
techniques may use archival formalin-fixed, paraffin-
embedded tissue; however, FISH studies using paraffin-
Fig. 16. Morphological features of AML with inv(16). (A-B) Hypercellular embedded tissue are more technically difficult and the
bone marrow with numerous blasts and immature eosinophils. (C)
results not straightforward to interpret. Please see review
Myeloblasts intermixed with characteristic immature eosinophils with
chunky basophilic granules. articles for more details [54-57].

4.2.1. Gene expression profiling


Class I mutations give a proliferative and/or survival Oligonucleotide or complementary DNA microarray
advantage; these include RAS and FLT3 mutations. Class technology is being established as an alternative and
II mutations complement class I mutation and impair extension to conventional karyotyping and FISH in the
myeloid differentiation. diagnosis of leukemia subtypes. Several groups have
Both inv(16) and t(8;21) belong to class II mutations and demonstrated that the sensitivity and specificity of micro-
involve subunits of the transcription factor complex core- array technology is as high as 100% for some types of AML
binding factor (CBF ). The inv(16) abnormality results in with recurrent cytogenetic abnormalities and more than 90%
54 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

Fig. 17. Morphological features of AML with multilineage dysplasia. (A) Sheets of blasts; (B-F) marked dysplasia in all 3 lineages. Note that MPO stain is
absent in mature granulocytes, another sign of dysplasia (F).

for other types [58]. It appears unlikely, however, that or MLL [61,62] gene have considerably poorer prognoses
microarray technology will completely replace other tech- than those described above. The approximately 10% of
niques in the near future. patients who have a normal karyotype and a mutation in the
CEBPA gene and no FLT3 duplication have a prognosis
4.3. Prognostic indicators
similar to that of patients with CBF AML [63]. Increased
The prognosis of an individual with AML depends on expression of the BAALC gene has been shown to worsen
several parameters, including age, cytogenetic findings, prognosis in patients with a normal karyotype [64].
preceding MDS and/or exposure to chemotherapeutic Identification of such genes will provide new therapeutic
agents, and coexisting morbidity. Acute myeloid leukemia btargetsQ as well as further refinement of prognoses. Once
with recurrent cytogenetic abnormalities t(8;21), inv(16), or covariates such as those noted above are specified, neither
t(15;17) is associated with a better prognosis, whereas AML the presence of dysplasia [65] nor morphological diagnosis
with 11q23, complex karyotype, preceding MDS, or (eg, AML RAEB-t or RAEB or, among AML patients, FAB
previous chemotherapy is associated with a worse prognosis. subtype) affects prognosis [66].
Tools for further discrimination of prognosis in this
patient group are becoming available. It has been estab- 4.3.1. Minimal residual disease
lished, for example, that patients who have a normal Since disease recurs in most patients who are in complete
karyotype but also have duplication of the FLT3 [59,60] remission (CR), detection of minimal residual disease in
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 55

detection is, in principle, relevant to all patients.


However, the inability to establish an aberrant
immunophenotype in every patient might be prob-
lematic. Furthermore, because it is not logistically
feasible to do a complete screen for an aberrant
immunophenotype at every time point, screening
once treatment begins focuses on the aberrant
immunophenotype detected at diagnosis. Hence, the
possibility that the immunophenotype could change
between diagnosis and relapse could also limit
applicability [71].
3. Molecular. Polymerase chain reaction screening to
detect minimal residual disease is currently applica-
ble to APL, as described in that section. On the other
hand, the applicability of such analysis to CBF AML
is unresolved [73].
For more details on methods for detecting MDR in AML,
please see the comprehensive review by Raanani and Ben-
Bassat [74].

5. Differential diagnosis
In a general surgical pathology practice, cases of AML,
including MS, are relatively rare, but the diagnosis is very
often difficult.
5.1. Hematologic malignancies

5.1.1. Acute lymphoblastic leukemia


Distinguishing between AML and ALL on morpholog-
ical basis only is not reliable, especially in cases of AML
Fig. 18. Morphological features of therapy-related AML. (A) Bone marrow
biopsy showing foci of immature cells and dysplastic megakaryocytes. (B)
Bone marrow aspirate smear showing blasts and dysplastic features in
erythroid and granulocytic lineages.

patients who appear to be approaching CR or who are in CR


has received considerable attention. Detection methods may
be grouped as follows:
1. Cytogenetic. It has been found that among patients
presenting with abnormal cytogenetic findings,
cytogenetic analysis 21 days after beginning therapy
is predictive of relapse-free survival, independent of
the proportion of blasts in the marrow on day 21 or
initial cytogenetic results [67]. Others have noted
that the presence of residual abnormal metaphases at
the time of CR is predictive of shorter relapse-free
survival time [68,69].
2. Immunophenotypic. Aberrant immunophenotypes
(eg, asynchronous surface antigen expression or
lymphoid-associated markers such as CD2, CD5, or
CD19) suggestive of the presence of morphologically
Fig. 19. Electron micrograph of a myeloblast. In this case, less than 3% of
undetectable AML may be useful either during the blasts demonstrate weak MPO positivity by cytochemistry. Electron
first course of therapy or in CR [70-72]. Unlike cyto- microscopy demonstrated MPO positivity in blasts. Note dense-staining
genetic or molecular methods, immunophenotypic peroxidase-positive primary granules.
56 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

Fig. 20. Typical flow cytometry immunophenotype of a case with t(8;21). Neoplastic cells are positive for CD34, CD45, CD117, and MPO. Note coexpression
of a lymphoid marker, CD19, on CD34-positive cells.

with blasts showing little or no differentiation. To complicate 5.1.2. Biphenotypic leukemia


the issue, the leukemic cells in AML M0 as well as AML M7 Biphenotypic acute leukemias have been described in the
are negative for MPO, some AML cases demonstrate literature as mixed lineage or hybrid acute leukemia. The
aberrant expression of TdT and other lymphoid-associated EGIL summarized the defining criteria, which are based on
markers, and some cases of ALL demonstrate aberrant the number and degree of specificity of the markers
expression of myeloid-associated markers. It is therefore (lymphoid and myeloid) expressed by the leukemic cells
mandatory to use a panel of basic histochemical and (Table 3) [46].
immunophenotypic markers in all cases of acute leukemia Cases of acute leukemia with two distinct populations of
to assign an appropriate lineage. The components of such a cells expressing different markers are called bbilineage
panel vary from one institution to another. At MD Anderson leukemiaQ or bmixed-lineage leukemia.Q For cases with a
Cancer Center, we routinely use MPO, a-naphthyl butyrate single population of cells expressing the markers of two
esterase, PAS, naphthol ASD chloroacetate esterase (by lineages, the term bbiphenotypic leukemiaQ is used. It is
histochemical methods), POD (by an immunofluorescence possible sometimes to see two distinct (by morphological
method), and, by flow cytometry, a panel of markers criteria) blast populations in the same case, one of small size
that includes CD3, CD4, CD5, CD7, CD8, CD10, CD13, with a high nucleus-cytoplasm ratio (resembling lympho-
CD14, CD19, CD33, CD34, CD64, CD117, MPO, TdT, and blasts) and the other larger with more abundant cytoplasm
HLA-DR. The use of this panel allows us to assign a lineage with or without granulation (resembling myeloblasts). The
of differentiation (and thus make a definitive diagnosis of following cases have been found to have a high incidence of
AML or ALL) in most cases and recognize cases of structural chromosome abnormalities: the Ph chromosome,
ambiguous leukemia. rearrangements involving 11q23, and complex cytogenetic
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 57

Fig. 21. Flow cytometric immunophenotype of an AML M5 case. Neoplastic cells are positive for CD13, CD33, CD45, CD117, and MPO and negative for
CD34 and HLA-DR.

abnormalities [75]. A flow cytometric panel with an optimal diagnosis of AML for cases of AML with Ph and/or
number of markers and the use of well-defined criteria for BCR/ABL fusion gene transcripts, raising the possibility of
the diagnosis of biphenotypic acute leukemia are critical in CML blast phase in a comment. In our opinion, the presence
making the distinction from AML. of typical micromegakaryocytes and increases in the number
of eosinophils and/or basophils increase the chances of
5.1.3. Blast phase of chronic myeloid leukemia CML, but do not make it certain.
The blast phase may be an initial presentation of chronic
5.2. CD4+CD56+ hematodermic neoplasm
myeloid leukemia, and distinguishing it from AML may be
difficult. The presence of Ph chromosome and/or BCR/ABL The CD4+CD56+ hematodermic neoplasm, also known
fusion gene transcripts is not a reliable criterion, as some as blastic natural killer (NK) cell lymphoma and designated
cases of AML with these features have been reported [76], as such in a current WHO classification, represents a
including cases in which BCR/ABL fusion gene transcripts relatively recently described entity with aggressive behavior.
were acquired by an MDS clone [77]. To complicate the Originally thought to belong to the NK lineage, the
issue, the blast karyotype in blast phase is often very neoplastic cells derive from plasmacytoid dendritic cells
complex. Some authors believe that the only reliable way to [78]. The disease may be limited to skin involvement only,
distinguish these entities is to wait until CR and then test may spread to a regional lymph node, or may be systemic
maturing hematopoietic elements for the BCR/ABL fusion with involvement of the bone marrow and peripheral blood
gene transcript, which would be present in CML and absent [78]. Histological analysis reveals that medium-sized
in AML. Fortunately, the short-term approach to these two lymphoid-appearing cells with scant cytoplasm, a slightly
entities is rather similar. At MD Anderson, we make a irregular-shaped nucleus with fine chromatin, and one to
58 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

Table 2 Table 4
Immunophenotypic profiles of immature myeloid elements Recurrent cytogenetic abnormalities in children and adults with AML [51]
Cell type Characteristic immunophenotypic features/comments Abnormalities Fusion genes Frequency (%)
Myeloblast wCD45, CD34, CD33a, CD15, CD13a, vCD11c, Children Adults
CD4, HLA-DR, MPO t(8;21)(q22;q22) AML1-ETO 10-15 8-12
Promyelocyte CDllc, CD13, CD15, CD33, CD45, MPO, loss of inv(16)(p13q22) CBFB-MYH11 6-12 8-12
HLA-DR, and acquisition of strong CD15 and t(15;17)(q22;q21) PML-RARa 8-15 8-10
CD11c associated with maturation. Gradual loss
of CD33 also characterizes successive maturation
stages. include small cell lung carcinoma, Merkel cell tumor, and
Monoblast wCD45, vCD34, CD33, CD15, CD13, CD11c, rhabdomyosarcoma in adults and Ewing sarcoma/primitive
vCD4, HLA-DR
neuroectodermal tumor (PNET) and rhabdomyosarcoma in
Promonocyte HLA-DR, CD13, CD33, CD14, CD4, CD15,
CD11c, CD45 children. The morphological distinction between AML and
Erythroblast Glycophorin A, hemoglobin A, CD45 all other types of small round cell tumors is difficult when
Megakaryoblastb HLA-DR, vCD34, CD41b, CD13, vCD33, the neoplasms are initially detected in the bone marrow.
CD61, CD45, CD31. Progressive maturation Routine Wright-Giemsa–stained preparations may show
characterized by loss of CD34 and acquisition of
overlapping cytological features, although these cases will
CD42 and von Willebrand factor
be negative for MPO and TdT.
v indicates variable antigen expression; w, weak antigen expression.
a As a rule, cytomorphology alone is not sufficient to
Rare AML lacks surface CD13 and CD33.
b
False-positive CD41 expression by flow cytometry may be secondary distinguish these entities, but in conjunction with relevant
to platelet adherence to blasts. clinical information (eg, patient’s age), morphology is often
helpful in predicting the type and the primary site of the
several medium or small nucleoli infiltrate the dermis. tumor. Immunohistochemical, ultrastructural, and genetic
Occasionally, large and small cells and/or an associated and molecular studies are also helpful when the primary site
inflammatory infiltrate can be seen in the background. is not evident from radiological studies or when laboratory
Mitotic figures are rare. The epidermis is always spared. studies (eg, ectopic secretion of calcitonin by small cell
In lymph nodes, the involvement starts in the medulla carcinoma or other amine and peptide hormones) are not
and advances to the sinuses and cortical interfollicular areas. sufficient for making a diagnosis.
The neoplastic cells are positive for CD4, CD43, CD45, Usually, little stromal fibrotic reaction is induced by
CD45RA, and CD56 and, in about 50% of cases, for CD68 nonhematopoietic small round cell tumors metastatic to the
[78]. The cells are negative for markers of B-cell lineage bone marrow before initiation of chemotherapy or irradia-
(CD19, CD20, CD23, CD24, CD79A, and immunoglobu- tion. Thus, the marrow involved by these tumors is easily
lin), T-cell lineage (CD2, CD3, CD5, CD7, CD8, b-F1, and aspirated. A bone marrow core biopsy and imprints made
d-TCR1), NK cell lineage (CD16 and CD57), and myelo- from needle biopsy should accompany all aspirates because
monocytic cell lineage (CD13, CD14, CD15, CD33, and these two marrow-sampling techniques, when performed
CD117), with rare exceptions [78]. together; maximize the odds of detecting metastases. In
addition, the histological appearance of the tumor in the
5.3. Nonhematologic malignancies biopsy core is sometimes sufficient to distinguish between
In addition to lymphoma and acute leukemia, pathologic acute leukemia and a metastatic process.
entities characterized as small round cell tumors may As discussed previously, the diagnosis of AML requires
well-prepared and stained aspirate smears, along with an
Table 3 appropriate workup with various ancillary laboratory
Scoring system for defining acute biphenotypic leukemiasa [46] studies. Care should be exercised when interpreting
Scoring points B cell T cell Myeloid nonspecific esterase results because some carcinomas or
2 CytCD79a (mb-1) CD3 (m/cyt) MPOb rhabdomyosarcomas may give positive reactions. Although
CytCD22 Anti-TCR metastatic small cell tumors tend to present as cell
CytIgM aggregates, diffuse noncohesive cells are common on bone
1 CD19 CD2 CD117
CD20 CD5 CD13
marrow aspirate smears.
CD10 CD8 CD33
CD10 CD65 5.3.1. Melanoma
0.5 TdT TdT CD14 It is well known that the morphology of metastatic
CD24 CD7 CD15 melanoma varies significantly and that metastases may
CD1a CD64
present without any history of primary skin involvement.
Cyt indicates cytoplasmic; m, membrane; TCR, T-cell receptor.
a This also applies to bone marrow. Cells are usually
Biphenotypic acute leukemia is established when the score from two
separate lineages is greater than 2. moderately pleomorphic with large nuclei, focally promi-
b
Myeloperoxidase demonstrated by cytochemical or flow cytometric nent nucleoli, and abundant eosinophilic cytoplasm that
studies. contains fine granular brown pigment. Cells usually grow
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 59

Fig. 22. Promyelocytic leukemia protein oncogenic domain test, also referred to as POD. This is an immunofluorescence technique using antibody specific for
PML. Normally, PML is compactly located at multiple nuclear domains known as PODs, whose structures are dynamically regulated and disrupted in t(15;17)
cases. (A) Normal coarse staining pattern. (B) A fine, dispersed staining pattern in a case with the t(15;17).

diffusely or in nodular aggregates. Occasionally, small cells One recently published case of metastatic melanoma
with scant cytoplasm or spindly cells may be present or may presenting as pancytopenia in a 3-year-old boy brought out
even be predominant. In some cases, cells contain no an important point, as melanoma cells were positive for CD56
pigment (Fig. 25). Growth in a discrete rounded aggregate and CD117 on flow cytometric immunophenotype analysis
with a nevoid appearance is helpful. Melanin may be [79]. That observation stresses again the importance of not
misinterpreted as lipochrome pigment or hemosiderin. relying on a limited panel of immunophenotypic markers.
S-100 antibodies are considered more sensitive than
HMB-45 for detection of melanoma cells. Other useful 5.3.2. Rhabdomyosarcoma
markers include melan-A (Mart-1), tyrosinase, and neuron- Rhabdomyosarcoma may present as disseminated disease
specific enolase (NSE). with no certain primary lesion and in some cases is restricted

Fig. 24. The LSI CBFB (Core Binding Factor Beta subunit) Dual Color,
Fig. 23. The LSI PML/RARa Dual Color Translocation Probe. A normal Break Apart Rearrangement Probe. Nucleus without inv(16) will have two
cell has a two orange and two green signal pattern. In an abnormal cell fused red/green (yellow) signals. The inv(16) results in separate red and
containing a PML/RARa fusion, one green (RARa), one magenta (PML), green signals appearing on opposite arms of the inverted 16 chromosome
and one closely adjacent or fused green/magenta (yellow) signal pattern is creating an adjacent or fused red/green signal on the q arm of one of the
observed. Fluorescence in situ hybridization can be performed on an 16 chromosomes and a green signal on the other arm of 16, while the
archive smear as shown in this picture. 16 chromosome homolog will only contain the red signal on one arm.
60 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

Fig. 25. Involvement of bone marrow in a patient with malignant melanoma. (A) Bone marrow clot packed by immature cells with a similarity to blasts
or immature plasma cells. (B-D) Neoplastic cells in the bone marrow aspirate. Although cells are very similar to blasts, note that nuclear chromatin is
more condensed that nuclear chromatin of a typical myeloblast. Binucleation is also not typical for AML, but is very typical for multiple myeloma. (E)
HMB-45 stain.

to bone marrow [80]. Almost all reported cases of of rhabdomyosarcoma may be nonimmunoreactive with all
rhabdomyosarcoma with bone marrow involvement, includ- markers but vimentin. Molecular and cytogenetic studies are
ing cases presenting in bone marrow, were alveolar helpful. Most alveolar rhabdomyosarcoma cases have the
rhabdomyosarcoma [80]. The diagnosis of acute leukemia translocation t(2;13)(q35;q14); a smaller subset have the
is suspected because of the presence of undifferentiated cells translocation t(1;13)(p36;q14) [82]. Most cases of embryo-
in the bone marrow and signs of acute systemic process nal rhabdomyosarcoma have allelic loss in chromosomal
(fever, anorexia, disseminated intravascular coagulation). In region 11p15 [83].
fact, in one report, 5 of 10 cases of rhabdomyosarcoma with
bone marrow involvement had a referring diagnosis of acute 5.3.3. Ewing sarcoma/PNET
leukemia [81]. By morphology, malignant cells may be The pelvic bones are the presenting sites of Ewing
indistinguishable from blasts (Fig. 26). Immunohistochem- sarcoma/PNET in 12% to 18% of patients. As such, a bone
ical studies for the muscle markers (desmin, myoglobin, marrow aspirate and biopsy may be the first evidence of this
myosin, muscle-specific actin [HHF-35], and more specific tumor in a patient. The cytology of the tumor cells on the
MyoD1 and myogenin) usually aid in the diagnosis. It is aspirate smear and biopsy section is usually not sufficient
important to bear in mind that some undifferentiated cases for a definite diagnosis. The use of immunoperoxidase
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 61

detected by immunologic methods. Such bone marrow


involvement seems to be associated with poorer patient
survival rates [85]. On marrow aspirate smear, these
tumor cells present as clusters of large tumor cells with
immature chromatin and mitosis. The malignant tumor
cells show immunoreactivity with antibodies against NSE,
neurofilaments, chromogranin, synaptophysin, low-molec-
ular-weight cytokeratin, CD57, epithelial membrane anti-
gen (EMA), TTF-1, and Bcl-2 and are negative for CD99
and 34b12.

5.3.5. Germ cell tumor


Occasionally, germ cell tumors enter the differential
diagnosis for MS. The morphology can be extremely similar
(Fig. 27), and to complicate the issue, some cases of
seminoma are positive for CD117 and CD57 and negative
for cytokeratin. CD45 is usually negative, and immunore-
activity for placental alkaline phosphatase (PLAP) and
a-fetoprotein often helps distinguish seminoma from MS.
In addition to PLAP and CD57, embryonal sarcomas are
usually positive for CD30 and cytokeratin.

Fig. 26. (A-B) Rhabdomyosarcoma involving bone marrow. Note the


different staining characteristics of cytoplasm of malignant cells, while the
nuclear chromatin is very similar to that of myeloblasts.

stains for leukocyte common antigen (CD45), TdT, CD43,


CD34, CD99, MPO, NSE, and lysozyme is critical in this
situation. Ewing sarcoma/PNET is positive for vimentin,
CD99, NSE, and, in some cases, cytokeratin and negative for
other markers. Molecular and cytogenetic studies are useful
to demonstrate the fusion of the EWS gene, located on 22q12,
with one of the members of the ETS family. Two recurrent
translocations, t(11;22)(q24;q12) and t(21;22)(q22;q12),
account for more than 95% of all cases [84], whereas other
translocations such as t(7;22), t(17;22), and t(2;22) and
inversion 22 are rare. Some authors believe that virtually all
cases of Ewing sarcoma/PNET have some form of EWS/ETS
gene fusion [84].
It is very important to realize that the expression of CD99
(MIC2) is by no means specific for Ewing sarcoma. In
addition to Ewing sarcoma/PNET and ALL, CD99 expres-
sion has been detected in as many as 55% of MS cases and
43% of AML [3].
Fig. 27. Seminoma involving a lymph node. (A) Medium power
5.3.4. Small cell lung cancer (400). Sheets of neoplastic cells with residual lymphoid tissue. (B) High
Fifty to seventy percent of patients with small cell power (1000). Neoplastic cells are indistinguishable morphologically
cancer of the lung have bone marrow involvement from monoblasts.
62 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

5.3.6. Poorly differentiated carcinoma


Poorly differentiated carcinoma enters the differential
diagnosis for MS (Fig. 28). The morphology of immature
cells assessed on touch imprint preparation could be
extremely helpful. Histochemical and immunoperoxidase
studies for markers of myeloid or monocytic differentiation
(MPO, naphthol ASD chloroacetate esterase, lysozyme,
CD68) and for cytokeratin are useful in distinguishing these
two entities. For male patients, immunohistochemical stains
for prostate-specific antigen, prostatic acid phosphatase,
CD57, a-methylacyl-CoA racemase (P504S), EMA, carci-
noembryonic antigen (CEA), B72.3, and 34bE12 are useful
in the diagnosis of metastatic prostate carcinoma. For female
patients, a-lactalbumin, low-molecular-weight cytokeratin,
EMA, CEA, B72.3, and BCA-225 are used to rule out breast
carcinoma. TTF-1, surfactant apoprotein A, and low- and
high-molecular-weight cytokeratins are useful for lung
primary tumors.
5.4. Megaloblastic anemia
Rarely, megaloblastic anemia can be morphologically
indistinguishable from AML M6. Both conditions share
hypercellular bone marrow and bizarre morphology of

Fig. 29. Megaloblastic anemia due to severe vitamin B12 deficiency. (A)
Hypercellular bone marrow. (B) Aspirate smear shows sheets of immature
erythroblasts. Compare the morphology of erythroid precursors in this case
with AML M6b (Fig. 13).

erythroid precursors (Fig. 29). Increased percentage of


myeloblasts helps in establishing the diagnosis of AML
M6a, but this feature is absent by definition in AML M6b
(so-called pure erythroid leukemia). The presence of
hypersegmented granulocytes and a range of megaloblastic
erythroid precursors with nuclear/cytoplasmic asynchrony is
a typical feature of megaloblastic anemia, but in severe
cases, there is no maturation in both myeloid and erythroid
series. The serum level of folic acid and vitamin B12 and
pertinent clinical history including exposure to several
medications are crucial.

6. Summary
With our present knowledge, the optimal management
of individual cases of AML requires that all cases be
studied by morphological, cytochemical, cytogenetic,
immunologic, and molecular techniques. Other techniques,
such as gene expression profiling in AML, are important
in the advancement of knowledge but have not yet had a
major impact on patient management. Future studies on the
clinical importance of the expression of various markers
Fig. 28. Metastatic involvement of bone marrow in a patient with cervical will further aid clinicians in treating AML and in
carcinoma. (A-B) Note the similarity of neoplastic cells to AML. preventing refractory disease.
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 63

References [23] O’Regan S, Carson S, Chesney RW, et al. Electrolyte and acid-
base disturbances in the management of leukemia. Blood 1977;49:
[1] Brunning RDME, Harris NL, Flandrin G, Vardiman J, Bennett J, 345 - 53.
Head D. Acute myeloid leukemia. In: Jaffe ES, Harris NL, Stein H, [24] Wiernik P. Diagnosis and treatment of adult acute myelocytic
Vardiman JW, editors. World Health Organization Classification of leukemia. In: Wiernik PH, Canellos GP, Dutcher JP, Kyle RA,
Tumours: Pathology and Genetics of Tumors of Haematopoietic and editors. Neoplastic Diseases of the Blood. New York7 Churchill
Lymphoid Tissues. Lyon7 ARC Press; 2000. p. 75 - 107. Livingstone; 1996. p. 331 - 51.
[2] Vardiman JW, Harris NL, Brunning RD. The World Health [25] Lichtman MA, Rowe JM. Hyperleukocytic leukemias: rheological,
Organization (WHO) classification of the myeloid neoplasms. Blood clinical, and therapeutic considerations. Blood 1982;60:279 - 83.
2002;100:2292 - 302. [26] Cuttner J. Hyperleukocytosis in adult leukemias. In: Bloomfield CD,
[3] Zhang PJ, Barcos M, Stewart CC, et al. Immunoreactivity of MIC2 editor. Chronic and Acute Leukemias in Adults. Boston7 Martinus
(CD99) in acute myelogenous leukemia and related diseases. Mod Nijhoff; 1985. p. 263-7.
Pathol 2000;13:452 - 8. [27] Gralnick HR, Marchesi S, Givelber H. Intravascular coagulation in
[4] Dicke KA, Hood DL, Arneson M, et al. Effects of short-term in vivo acute leukemia: clinical and subclinical abnormalities. Blood
administration of G-CSF on bone marrow prior to harvesting. Exp 1972;40:709 - 18.
Hematol 1997;25:34 - 8. [28] Goad KE, Gralnick HR. Coagulation disorders in cancer. Hematol
[5] Ganser A, Ottmann OG, Seipelt G, et al. Effect of long-term treatment Oncol Clin North Am 1996;10:457 - 84.
with recombinant human interleukin-3 in patients with myelodys- [29] Kubota T, Andoh K, Sadakata H, et al. Tissue factor released from
plastic syndromes. Leukemia 1993;7:696 - 701. leukemic cells. Thromb Haemost 1991;65:59 - 63.
[6] Naeim F, Champlin R, Nimer S. Bone marrow changes in patients [30] Ribeiro RC, Pui CH. The clinical and biological correlates of
with refractory aplastic anemia treated by recombinant GM-CSF. coagulopathy in children with acute leukemia. J Clin Oncol
Hematol Pathol 1990;4:79 - 85. 1986;4:1212 - 8.
[7] Fialkow PJ, Singer JW, Adamson JW, et al. Acute nonlympho- [31] Scott CS, Stark AN, Limbert HJ, et al. Diagnostic and prognostic
cytic leukemia: heterogeneity of stem cell origin. Blood 1981;57: factors in acute monocytic leukaemia: an analysis of 51 cases. Br J
1068 - 73. Haematol 1988;69:247 - 52.
[8] Ries LAG, Eisner MKC. SEER Cancer statistics review, 1973-1999. [32] Muller S, Sangster G, Crocker J, et al. An immunohistochemical and
Bethesda (Md): National Cancer Institute; 2002. clinicopathological study of granulocytic sarcoma (dchloromaT).
[9] Hernandez JA, Land KJ, McKenna RW. Leukemias, myeloma, and Hematol Oncol 1986;4:101 - 12.
other lymphoreticular neoplasms. Cancer 1995;75:381 - 94. [33] Neiman RS, Barcos M, Berard C, et al. Granulocytic sarcoma: a
[10] Kinlen LJ. Leukaemia. Cancer Surv 1994;19-20:475 - 91. clinicopathologic study of 61 biopsied cases. Cancer 1981;48:
[11] Linet MS, Devessa SS. Epidemiology of leukemia: overview and 1426 - 37.
patterns of epidemiology of leukemia: overview and patterns of [34] Abe R, Umezu H, Uchida T, et al. Myeloblastoma with an 8;21
occurrence. In: Henderson ES, Lister TA, Greaves MF, editors. chromosome translocation in acute myeloblastic leukemia. Cancer
Leukemia. Philadelphia7 WB Saunders; 2002. p. 131 - 51. 1986;58:1260 - 4.
[12] Douer D, Preston-Martin S, Chang E, et al. High frequency of acute [35] Welch P, Grossi C, Carroll A, et al. Granulocytic sarcoma with an
promyelocytic leukemia among Latinos with acute myeloid leukemia. indolent course and destructive skeletal disease. Tumor characteriza-
Blood 1996;87:308 - 13. tion with immunologic markers, electron microscopy, cytochemistry,
[13] Wertelecki W, Shapiro JR. 45,XO Turner’s syndrome and leukaemia. and cytogenetic studies. Cancer 1986;57:1005 - 10.
Lancet 1970;1:789 - 90. [36] Valbuena JR, Admirand JH, Lin P, et al. Myeloid sarcoma involving
[14] Schwartz CL, Cohen HJ. Preleukemic syndromes and other the testis. Am J Clin Pathol 2005;124:445 - 52.
syndromes predisposing to leukemia. Pediatr Clin North Am [37] Shea B, Reddy V, Abbitt P, et al. Granulocytic sarcoma (chloroma) of
1988;35:853 - 71. the breast: a diagnostic dilemma and review of the literature. Breast J
[15] Kato H, Brown CC, Hoel DG, et al. Studies of the mortality of A- 2004;10:48 - 53.
bomb survivors. Report 7. Mortality, 1950-1978: Part II. Mortality [38] Tsimberidou AM, Kantarjian HM, Estey E, et al. Outcome in patients
from causes other than cancer and mortality in early entrants. Radiat with nonleukemic granulocytic sarcoma treated with chemotherapy
Res 1982;91:243 - 64. with or without radiotherapy. Leukemia 2003;17:1100 - 3.
[16] Kodama K, Mabuchi K, Shigematsu I. A long-term cohort study of the [39] Dusenbery KE, Howells WB, Arthur DC, et al. Extramedullary
atomic-bomb survivors. J Epidemiol 1996;6:S95-S105. leukemia in children with newly diagnosed acute myeloid leukemia: a
[17] Ellis M, Ravid M, Lishner M. A comparative analysis of alkylating report from the Children’s Cancer Group. J Pediatr Hematol Oncol
agent and epipodophyllotoxin-related leukemias. Leuk Lymphoma 2003;25:760 - 8.
1993;11:9 - 13. [40] Wiernik PH, De Bellis R, Muxi P, et al. Extramedullary acute
[18] Michels SD, McKenna RW, Arthur DC, et al. Therapy-related acute promyelocytic leukemia. Cancer 1996;78:2510 - 4.
myeloid leukemia and myelodysplastic syndrome: a clinical and [41] Meis JM, Butler JJ, Osborne BM, et al. Granulocytic sarcoma in
morphologic study of 65 cases. Blood 1985;65:1364 - 72. nonleukemic patients. Cancer 1986;58:2697 - 709.
[19] Pui CH, Relling MV, Rivera GK, et al. Epipodophyllotoxin-related [42] O’Malley DP, Orazi A, Wang M, et al. Analysis of loss of
acute myeloid leukemia: a study of 35 cases. Leukemia 1995;9: heterozygosity and X chromosome inactivation in spleens with
1990 - 6. myeloproliferative disorders and acute myeloid leukemia. Mod
[20] Mills PK, Newell GR, Beeson WL, et al. History of cigarette smoking Pathol 2005;18:1562-8.
and risk of leukemia and myeloma: results from the Adventist health [43] Bennett JM, Catovsky D, Daniel MT. Proposed revised criteria for the
study. J Natl Cancer Inst 1990;82:1832 - 6. classification of acute myeloid leukemia. A report of the French-
[21] Sullivan A. Classification, pathogenesis, and etiology of neoplastic American-British Cooperative Group. Ann Intern Med 1985;103:
diseases of the hematopoietic system. In: Lee GR, Bithell TC, Foerster 620 - 5.
J, et al., editors. Wintrobe’s clinical hematology. Philadelphia: Lea & [44] Wells DA, Benesch M, Loken MR, et al. Myeloid and monocytic
Febiger; 1993. p. 1725-91. dyspoiesis as determined by flow cytometric scoring in myelo-
[22] Boggs DR, Wintrobe MM, Cartwright GE. The acute leukemias. dysplastic syndrome correlates with the IPSS and with out-
Analysis of 322 cases and review of the literature. Medicine come after hematopoietic stem cell transplantation. Blood 2003;
(Baltimore) 1962;41:163 - 225. 102:394 - 403.
64 S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65

[45] Bennett JM, Catovsky D, Daniel MT, et al. Criteria for the diagnosis with normal cytogenetics: a Cancer and Leukemia Group B Study.
of acute leukemia of megakaryocyte lineage (M7). A report of the Blood 2003;102:1613 - 8.
French-American-British Cooperative Group. Ann Intern Med [65] Haferlach T, Schoch C, Loffler H, et al. Morphologic dysplasia in de
1985;103:460 - 2. novo acute myeloid leukemia (AML) is related to unfavorable
[46] EGIL. The value of c-kit in the diagnosis of biophenotypic acute cytogenetics but has no independent prognostic relevance under the
leukemia (European Group for the Immunological Classification of conditions of intensive induction therapy: results of a multiparameter
Leukemias). Leukemia 1998;12:2038. analysis from the German AML Cooperative Group studies. J Clin
[47] Bene MC, Castoldi G, Knapp W, et al. Proposals for the immunolog- Oncol 2003;21:256 - 65.
ical classification of acute leukemias. European Group for the [66] Estey E, Thall P, Beran M, et al. Effect of diagnosis (refractory anemia
Immunological Characterization of Leukemias (EGIL). Leukemia with excess blasts, refractory anemia with excess blasts in transfor-
1995;9:1783 - 6. mation, or acute myeloid leukemia [AML]) on outcome of AML-type
[48] Weir EG, Borowitz MJ. Flow cytometry in the diagnosis of acute chemotherapy. Blood 1997;90:2969 - 77.
leukemia. Semin Hematol 2001;38:124 - 38. [67] Konopleva M, Cheng SC, Cortes JE, et al. Independent prognostic
[49] Kussick SJ, Wood BL. Using 4-color flow cytometry to identify significance of day 21 cytogenetic findings in newly-diagnosed acute
abnormal myeloid populations. Arch Pathol Lab Med 2003;127: myeloid leukemia or refractory anemia with excess blasts. Haemato-
1140 - 7. logica 2003;88:733 - 66.
[50] Kussick SJ, Fromm JR, Rossini A, et al. Four-color flow cytometry [68] Freireich EJ, Cork A, Stass SA, et al. Cytogenetics for detection of
shows strong concordance with bone marrow morphology and minimal residual disease in acute myeloblastic leukemia. Leukemia
cytogenetics in the evaluation for myelodysplasia. Am J Clin Pathol 1992;6:500 - 6.
2005;124:170 - 81. [69] Marcucci G, Mrozek K, Ruppert AS, et al. Abnormal cytogenetics at
[51] Haferlach T, Schnittger S, Kern W, et al. Genetic classification of acute date of morphologic complete remission predicts short overall and
myeloid leukemia (AML). Ann Hematol 2004;83(Suppl 1):S97-S100. disease-free survival, and higher relapse rate in adult acute myeloid
[52] Czuczman MS, Dodge RK, Stewart CC, et al. Value of leukemia: results from Cancer and Leukemia Group B Study 8461.
immunophenotype in intensively treated adult acute lymphoblastic J Clin Oncol 2004;22:2410 - 8.
leukemia: Cancer and Leukemia Group B Study 8364. Blood [70] Sievers EL, Lange BJ, Alonzo TA, et al. Immunophenotypic evidence
1999;93:3931 - 9. of leukemia after induction therapy predicts relapse: results from a
[53] Huret JL, Dessen P, Bernheim A. An atlas of chromosomes in prospective Children’s Cancer Group study of 252 patients with acute
hematological malignancies example: 11q23 and MLL partners. myeloid leukemia. Blood 2003;101:3398 - 406.
Leukemia 2001;15:987 - 9. [71] Baer MR, Stewart CC, Dodge RK, et al. High frequency of
[54] Mrozek K, Heerema NA, Bloomfield CD. Cytogenetics in acute immunophenotype changes in acute myeloid leukemia at relapse:
leukemia. Blood Rev 2004;18:115 - 36. implications for residual disease detection (Cancer and Leukemia
[55] Giles FJ, Keating A, Goldstone AH, et al. Acute myeloid leukemia. Group B Study 8361). Blood 2001;97:3574 - 80.
Hematology (Am Soc Hematol Educ Program) 2002;73 - 110. [72] San Miguel JF, Vidriales MB, Lopez-Berges C, et al. Early
[56] McKenzie SB. Advances in understanding the biology and genetics of immunophenotypical evaluation of minimal residual disease in acute
acute myelocytic leukemia. Clin Lab Sci 2005;18:28 - 37. myeloid leukemia identifies different patient risk groups and may
[57] Smith SM, Le Beau MM, Huo D, et al. Clinical-cytogenetic
contribute to postinduction treatment stratification. Blood 2001;98:
associations in 306 patients with therapy-related myelodysplasia and
1746 - 51.
myeloid leukemia: the University of Chicago series. Blood
[73] Marcucci G, Caligiuri MA, Bloomfield CD. Core binding factor
2003;102:43 - 52.
(CBF) acute myeloid leukemia: is molecular monitoring by RT-PCR
[58] Haferlach T, Kohlmann A, Schnittger S, et al. Global approach to the
useful clinically? Eur J Haematol 2003;71:143 - 54.
diagnosis of leukemia using gene expression profiling. Blood
2005;106:1189 - 98. [74] Raanani P, Ben-Bassat I. Detection of minimal residual disease in
[59] Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 acute myelogenous leukemia. Acta Haematol 2004;112:40 - 54.
internal tandem duplication in patients with acute myeloid leukemia [75] Carbonell F, Swansbury J, Min T, et al. Cytogenetic findings in acute
(AML) adds important prognostic information to cytogenetic risk biphenotypic leukaemia. Leukemia 1996;10:1283 - 7.
group and response to the first cycle of chemotherapy: analysis of 854 [76] Paietta E, Racevskis J, Bennett JM, et al. Biologic heterogeneity in
patients from the United Kingdom Medical Research Council AML Philadelphia chromosome–positive acute leukemia with myeloid
10 and 12 trials. Blood 2001;98:1752 - 9. morphology: the Eastern Cooperative Oncology Group experience.
[60] Whitman SP, Archer KJ, Feng L, et al. Absence of the wild-type Leukemia 1998;12:1881 - 5.
allele predicts poor prognosis in adult de novo acute myeloid [77] Katsuno M, Yamashita S, Sadamura S, et al. Late-appearing
leukemia with normal cytogenetics and the internal tandem duplica- Philadelphia chromosome in a patient with acute nonlymphocytic
tion of FLT3: a Cancer and Leukemia Group B Study. Cancer Res leukaemia derived from myelodysplastic syndrome: detection of
2001;61:7233 - 9. P210- and P190-type bcr/abl fusion gene transcripts at the leukaemic
[61] Caligiuri MA, Strout MP, Lawrence D, et al. Rearrangement of ALL1 stage. Br J Haematol 1994;87:51 - 6.
(MLL) in acute myeloid leukemia with normal cytogenetics. Cancer [78] Petrella T, Bagot M, Willemze R, et al. Blastic NK-cell lymphomas
Res 1998;58:55 - 9. (agranular CD4+CD56+ hematodermic neoplasms): a review. Am J
[62] Dohner K, Tobis K, Ulrich R, et al. Prognostic significance of partial Clin Pathol 2005;123:662 - 75.
tandem duplications of the MLL gene in adult patients 16 to 60 years [79] Spiller SE, Hawkins DS, Finn LS, et al. Metastatic malignant
old with acute myeloid leukemia and normal cytogenetics: a study of melanoma presenting as pancytopenia in a three-year-old boy. Pediatr
the Acute Myeloid Leukemia Study Group Ulm. J Clin Oncol Blood Cancer 2005;45:60 - 3.
2002;20:3254 - 61. [80] Sandberg AA, Stone JF, Czarnecki L, et al. Hematologic masquerade
[63] Preudhomme C, Sagot C, Boissel N, et al. Favorable prognostic of rhabdomyosarcoma. Am J Hematol 2001;68:51 - 7.
significance of CEBPA mutations in patients with de novo acute [81] Etcubanas E, Peiper S, Stass S, et al. Rhabdomyosarcoma, presenting as
myeloid leukemia: a study from the Acute Leukemia French disseminated malignancy from an unknown primary site: a retrospective
Association (ALFA). Blood 2002;100:2717 - 23. study of ten pediatric cases. Med Pediatr Oncol 1989;17:39 - 44.
[64] Baldus CD, Tanner SM, Ruppert AS, et al. BAALC expression [82] Barr FG. Molecular genetics and pathogenesis of rhabdomyosarcoma.
predicts clinical outcome of de novo acute myeloid leukemia patients J Pediatr Hematol Oncol 1997;19:483 - 91.
S. Konoplev, C.E. Bueso-Ramos / Annals of Diagnostic Pathology 10 (2006) 39 – 65 65

[83] Scrable HJ, Witte DP, Lampkin BC, et al. Chromosomal localization [85] Pasini F, Cetto GL, Pelosi G. Does bone marrow involvement affect
of the human rhabdomyosarcoma locus by mitotic recombination prognosis in small-cell lung cancer? Ann Oncol 1998;9:247 - 50.
mapping. Nature 1987;329:645 - 7.
[84] Delattre O, Zucman J, Melot T, et al. The Ewing family of tumors—a
subgroup of small-round-cell tumors defined by specific chimeric
transcripts. N Engl J Med 1994;331:294 - 9.

Vous aimerez peut-être aussi