TO THE EDITOR:

The International Consensus Classification of Myeloid and Lymphoid Neoplasms is the result of the findings of a clinical advisory committee (CAC) meeting with involvement by an international group of hematologists, oncologists, pathologists, and geneticists.1,2 The formal CAC meeting, however, did not address potential changes to the classification of acute leukemias of ambiguous lineage (ALAL); a CAC subgroup was created to address this issue (Table 1). ALAL include biologically diverse leukemias that fail to show commitment to either the myeloid or lymphoid lineages or show evidence of commitment to more than 1 lineage.1-3 Cases in the former group are referred to as acute undifferentiated leukemias (AUL), whereas those in the latter are identified as mixed-phenotype acute leukemias (MPAL). One of the major challenges in the subclassification of ALAL in general and MPAL in particular is that classification based primarily on driver mutations does not explain the phenotypic diversity seen in these leukemias. Although we anticipate that future generations of leukemia classification will recognize that these are better considered as single entities with variable expression of differentiation-related markers, because the highest-level organizing principle of current classification is phenotypic, and because current clinical protocols still require distinction between MPAL and single-lineage leukemias, distinguishing MPAL from leukemia of a single lineage with the same genetic driver requires an immunophenotypic definition.

Table 1.

Acute leukemia of ambiguous lineage entities

Acute leukemia of ambiguous lineage
MPAL with defining genetic alterations 
MPAL with BCR::ABL1 
MPAL with t(v;11q23.3); KMT2A rearranged 
MPAL with ZNF384 rearrangement 
MPAL with BCL11B activation 
MPAL with defining immunophenotypic changes 
B/myeloid MPAL 
T/myeloid MPAL 
B/T/myeloid MPAL 
B/T MPAL 
Acute undifferentiated leukemia (AUL)  
ALAL, NOS  
Acute leukemia of ambiguous lineage
MPAL with defining genetic alterations 
MPAL with BCR::ABL1 
MPAL with t(v;11q23.3); KMT2A rearranged 
MPAL with ZNF384 rearrangement 
MPAL with BCL11B activation 
MPAL with defining immunophenotypic changes 
B/myeloid MPAL 
T/myeloid MPAL 
B/T/myeloid MPAL 
B/T MPAL 
Acute undifferentiated leukemia (AUL)  
ALAL, NOS  

NOS, not otherwise specified.

AUL is defined by a lack of lineage-specific markers (as outlined in Table 2). Based on a recent multiinstitutional study, cases with a partial or single full myeloid marker can be considered as AUL.11 Cases with immunophenotype of AUL but with cytogenetic abnormalities and molecular findings diagnostic of AML with myelodysplasia-related cytogenetic abnormalities or AML with myelodysplasia-related mutation should be diagnosed as AML.

Rare cases may fulfill neither the criteria for MPAL nor acute leukemia of a single lineage or may lack all the lineage-specific markers of Table 2. Such cases are best referred to as ALAL, NOS.

All ALAL categories require that the total number of blasts in blood or bone marrow must be >20%. MPAL cases can contain 2 or more completely separable single-lineage leukemia populations or more than 1 lineage-defining marker in a single blast population.4,5 In cases with separate blast populations, each population needs to be classified according to acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) criteria.1 In cases in which small aberrant clones of divergent lineage are identified (>5%), a diagnosis of MPAL could be rendered if clear immunophenotypic aberrancies are identified. Identification of such aberrancies is essential to differentiate a small bilineal blast population from normal myeloid or B-cell precursors. For cases with aberrant clones, which represent <5% of all cells, a diagnosis should be based on the major leukemic population with a descriptive modifier, for example, “predominantly ALL with a small leukemic population of myeloid lineage detected of uncertain significance.” Additional details should be provided in the body of the report.

Similar to the criteria published in the revised fourth edition of the World Health Organization classification of these disorders, B-lineage designation in MPAL requires strong expression of CD19, along with at least 1 other B-cell marker.6 If CD19 expression is weak, 2 other B-lineage markers are needed (Table 2). T-lineage assignment requires a fraction of blasts to show strong cytoplasmic expression of CD3 (cCD3), with the brightest expression comparable to that of background mature T cells (Table 2). The negative predictive value of dim, rather than strong, cCD3 expression for T lineage is still unknown, and it often disappears during treatment or at relapse.7,8 MPO has been considered the most specific marker for myeloid lineage in prior classification systems. At this time, no specific threshold is set for MPO positivity, but care should be taken in instances when <10% MPO+ cells are seen in flow cytometry.9 Additionally, extra care should be taken to discriminate small MPO populations from background nonneoplastic myeloid progenitors.10 Isolated MPO expression can also be seen in B-ALL cases with homogeneous expression of typical B lineage markers, and this alone should not change the diagnosis. AUL is defined by lack of lineage-specific markers (Table 2). A recent multiinstitutional study showed that only a small subset of AUL cases (25%) lacked all myeloid markers, whereas 62% showed partial or full expression of 1 myeloid marker.11 

Table 2.

Markers used in lineage assignment

LineageMarkers
B lineage  
Strong CD19 and ≥1 marker expression CD10, CD22, or CD79a 
Weak CD19 and ≥2 strongly expressed: CD10, CD22, CD79a 
Consider immunohistochemical stains for B lineage PAX5, OCT2, BOB1 
T lineage  
CD3 (surface or cytoplasmic) — 
Myeloid lineage  
MPO or — 
Monocytic differentiation NSE, CD64, CD11c, CD14, or lysozyme 
LineageMarkers
B lineage  
Strong CD19 and ≥1 marker expression CD10, CD22, or CD79a 
Weak CD19 and ≥2 strongly expressed: CD10, CD22, CD79a 
Consider immunohistochemical stains for B lineage PAX5, OCT2, BOB1 
T lineage  
CD3 (surface or cytoplasmic) — 
Myeloid lineage  
MPO or — 
Monocytic differentiation NSE, CD64, CD11c, CD14, or lysozyme 

The immunophenotypic criteria described here are for cases of suspected MPAL and are not required for straightforward cases of AML or ALL.

Expression should be at least similar to that seen in stage I B-cell precursors or mature B cells.

Genetically defined subtypes of ALAL

Studies have shown that distinct subsets of MPALs are classified based on recurring genetic alterations. In addition to MPAL with BCR::ABL1 and MPAL with KMT2A gene rearrangements, 2 new molecularly defined entities have been described, B/myeloid MPAL with ZNF384 rearrangements and T/myeloid MPAL with BCL11B activation (Table 1). As with BCR::ABL1 and KMT2A, these 2 newer entities can manifest as ALAL or as single-lineage leukemias. In childhood B/myeloid MPAL, fusion genes involving the ZNF384 transcription factor gene were identified in nearly 50% of cases.12 ALAL with rearrangements leading to deregulation of BCL11B (BCL11B-activated) comprise 10% to 15% of MPAL and are further enriched in patients with MPAL, T/myeloid cases, representing up to one-third of this population.13-15 Unlike the BCL11B alterations described in typical T-ALL, including loss-of-function sequence mutations, deletions, and rearrangements deregulating oncogenes such as TLX3, the 14q32 structural variants identified in ambiguous-lineage leukemias result in deregulation of BCL11B expression through juxtaposition of hematopoietic stem cell enhancers or formation of neo-enhancers that also deregulated BCL11B expression.15 

Other genetic alterations

The majority (64% to 87%) of MPAL cases have an abnormal karyotype. Cytogenetic abnormalities and molecular findings diagnostic of AML with myelodysplasia-related cytogenetic abnormalities or AML with myelodysplasia-related mutations1 are currently classified as AML, even if there is also evidence of concurrent lymphoid antigen expression or AUL immunophenotype, although such a feature could be annotated to highlight the immunophenotype.

Gene mutations found within MPAL are a mixture of those commonly seen in both AML and ALL and as such cannot be used to define a lineage.12 A study of 115 pediatric MPAL/ALAL cases showed 158 recurrently mutated genes, including the AML-associated genes FLT3, RUNX1, and CEBPA and the ALL-associated genes CDKN2A and ETV6. Most of the MPAL cases harbored mutually exclusive mutations of either WT1, ETV6, RUNX1, or CEBPA.16,ZNF384 fusions are also commonly seen in B-ALL and have an overlapping gene-expression profile with ZNF384-rearranged B/myeloid MPALs.12 AUL cases are characterized by frequent PHF6 mutations and trisomy 13.11 

T/myeloid MPAL cases possess a more heterogeneous group of driver mutations, with fusions that overlap with T-ALL such as ZEB2::BCL11B, NUP214::ABL1 and fusions with ETV6 involving multiple different partners.16 Early T-cell precursor (ETP) ALL, characterized by expression of CD7, weak or absent CD5, and 1 or more myeloid/stem cell markers, has been shown to have a gene-expression profile that is intermediate between T/myeloid MPALs and T-ALL.17 The coexpression of myeloid markers and CD3 makes distinguishing ETP-ALL from T/myeloid MPAL problematic; molecular data blur the increasingly artificial distinction between these entities. The BCL11B cases present with absence of CD5 and cCD3 or MPO expression, resulting in variable classification. Moreover, emerging work is showing that the biology of immature leukemias is explained by diverse genomic alterations converging on a conserved set of stem cell genes showing phenotypic ambiguity: ETP, MPAL, or neither. This further suggests that genetic alterations rather than phenotypic expression may, in the future, be a better way of classifying these leukemias.16 

The currently recommended treatment approach for MPAL in both pediatric and adult patients is induction therapy with an ALL regimen.17-19 Patients with MPAL with BCR::ABL1 benefit from addition of a tyrosine kinase inhibitor.19 Immunophenotypic properties of MPAL are not stable over time: both variable proportions of different lineages and phenotypic alterations are known as lineage switch. Most cases of lineage transformation involve a switch from ALL to AML, and a retrospective review can often reveal the presence of an unrecognized or underappreciated malignant subclonal population present in the initial specimen, which displayed more chemoresistance and emerged after eradication of the chemosensitive dominant clone.20 The selective pressure brought on by specific therapies can influence these transformations, especially in MPAL (eg, resultant AML after the use of B-cell–specific therapy such as blinatumomab). However, frank ALL to AML lineage switch must be distinguished from the B-monocytic transition commonly seen with ALL with DUX4-rearrangements and other alterations early in therapy,21 in which such changes do not affect prognosis with continued ALL therapy.

Contribution: O.K.W. drafted the first version of the manuscript based on discussions of a clinical advisory committee meeting; and all other authors participated in the classification draft and reviewed and edited the final manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Olga K. Weinberg, UT Southwestern Medical Center BioCenter, 2230 Inwood Rd, EB03.220G, Dallas, TX 75235; e-mail: olga.weinberg@utsouthwestern.edu.

1.
Arber
DA
,
Orazi
A
,
Hasserjian
RP
, et al
.
International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: integrating morphologic, clinical, and genomic data
.
Blood
.
2022
;
140
(
11
):
1200
-
1228
.
2.
Campo
E
,
Jaffe
ES
,
Cook
JR
, et al
.
The International Consensus Classification of Mature Lymphoid Neoplasms: a report from the Clinical Advisory Committee
.
Blood
.
2022
;
140
(
11
):
1229
-
1253
.
3.
Khoury
JD
,
Solary
E
,
Abla
O
, et al
.
The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms
.
Leukemia
.
2022
;
36
(
7
):
1703
-
1719
.
4.
Aggarwal
N
,
Weinberg
OK
.
Update on acute leukemias of ambiguous lineage
.
Clin Lab Med
.
2021
;
41
(
3
):
453
-
466
.
5.
Alexander
TB
,
Orgel
E
.
Mixed phenotype acute leukemia: current approaches to diagnosis and treatment
.
Curr Oncol Rep
.
2021
;
23
(
2
):
22
.
6.
World Health Organization
. Classification of Tumours of Haematopoietic and Lymphoid Tissues.
IARC
;
2008
.
7.
Toba
K
,
Hanawa
H
,
Fuse
I
, et al
.
Difference in CD22 molecules in human B cells and basophils
.
Exp Hematol
.
2002
;
30
(
3
):
205
-
211
.
8.
Lewis
RE
,
Cruse
JM
,
Sanders
CM
,
Webb
RN
,
Suggs
JL
.
Aberrant expression of T-cell markers in acute myeloid leukemia
.
Exp Mol Pathol
.
2007
;
83
(
3
):
462
-
463
.
9.
van den Ancker
W
,
Westers
TM
,
de Leeuw
DC
, et al
.
A threshold of 10% for myeloperoxidase by flow cytometry is valid to classify acute leukemia of ambiguous and myeloid origin
.
Cytometry B Clin Cytom
.
2013
;
84
(
2
):
114
-
118
.
10.
Borowitz
MJ
.
Mixed phenotype acuteleukemia
.
Cytometry B Clin Cytom
.
2014
;
86
(
3
):
152
-
153
.
11.
Weinberg
OK
,
Hasserjian
RP
,
Baraban
E
, et al
.
Clinical, immunophenotypic, and genomic findings of acute undifferentiated leukemia and comparison to acute myeloid leukemia with minimal differentiation: a study from the bone marrow pathology group
.
Mod Pathol
.
2019
;
32
(
9
):
1373
-
1385
.
12.
Alexander
TB
,
Gu
Z
,
Iacobucci
I
, et al
.
The genetic basis and cell of origin of mixed phenotype acute leukaemia
.
Nature
.
2018
;
562
(
7727
):
373
-
379
.
13.
Giacomo
DD
,
La Starza
R
,
Gorello
P
, et al
.
14q32 rearrangements deregulating BCL11B mark a distinct subgroup of T-lymphoid and myeloid immature acute leukemia
.
Blood
.
2021
;
138
(
9
):
773
-
784
.
14.
Wang
W
,
Beird
H
,
Kroll
CJ
, et al
.
T(6;14)(q25;q32) involves BCL11B and is highly associated with mixed-phenotype acute leukemia, T/myeloid
.
Leukemia
.
2020
;
34
(
9
):
2509
-
2512
.
15.
Montefiori
LE
,
Bendig
SJ
,
Gu
Z
, et al
.
Enhancer hijacking drives oncogenic BCL11B expression in lineage-ambiguous stem cell leukemia
.
Cancer Discov
.
2021
;
11
(
11
):
2846
-
2867
.
16.
Takahashi
K
,
Wang
F
,
Morita
K
, et al
.
Integrative genomic analysis of adult mixed phenotype acute leukemia delineates lineage associated molecular subtypes
.
Nat Commun
.
2018
;
9
(
1
):
2670
.
17.
Hrusak
O
,
de Haas
V
,
Stancikova
J
, et al
.
International cooperative study identifies treatment strategy in childhood ambiguous lineage leukemia
.
Blood
.
2018
;
132
(
3
):
264
-
276
.
18.
Orgel
E
,
Alexander
TB
,
Wood
BL
, et al
.
Mixed-phenotype acute leukemia: a cohort and consensus research strategy from the Children's Oncology Group Acute Leukemia of Ambiguous Lineage Task Force
.
Cancer
.
2020
;
126
(
3
):
593
-
601
.
19.
Shimizu
H
,
Yokohama
A
,
Hatsumi
N
, et al
.
Philadelphia chromosome-positive mixed phenotype acute leukemia in the imatinib era
.
Eur J Haematol
.
2014
;
93
(
4
):
297
-
301
.
20.
Kurzer
JH
,
Weinberg
OK
.
To B- or not to B-: a review of lineage switched acute leukemia
.
Int J Lab Hematol
.
2022
;
44
(
suppl 1
):
64
-
70
.
21.
Novakova
M
,
Zaliova
M
,
Fiser
K
, et al
.
DUX4r, ZNF384r and PAX5-P80R mutated B-cell precursor acute lymphoblastic leukemia frequently undergo monocytic switch
.
Haematologica
.
2021
;
106
(
8
):
2066
-
2075
.
Sign in via your Institution