Journal Information
Vol. 43. Issue 4.
Pages 499-506 (October - December 2021)
Share
Share
Download PDF
More article options
Visits
4126
Vol. 43. Issue 4.
Pages 499-506 (October - December 2021)
Special article
Open Access
Updating recommendations of the Brazilian Group of Flow Cytometry (GBCFLUX) for diagnosis of acute leukemias using four-color flow cytometry panels
Visits
4126
Míriam P. Beltramea,
Corresponding author
mpbeltrame@yahoo.com.br

Corresponding author at: Hospital Erasto Gaertner, Rua Ovande do Amaral 201, 81520-060, Curitiba, PR, Brazil.
, Elizabeth Xisto Soutob,c, Mihoko Yamamotod, Felipe M Furtadoe,f, Elaine Sobral da Costag, Alex Freire Sandesd,h, Glicínia Pimentai, Geraldo Barroso Cavalcanti Júniorj, Maria Cláudia Santos-Silvak, Irene Lorand-Metzel, Maura R V Ikoma-Colturatoe,m
a Hospital Erasto Gaertner, Laboratório de Citometria de Fluxo, Curitiba, PR, Brazil
b Hospital do Câncer de Barretos, Barretos, SP, Brazil
c Hospital Brigadeiro, São Paulo, SP, Brazil
d Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
e Sabin Medicina Diagnóstica, Brasília, DF, Brazil
f Hospital da Criança de Brasília José Alencar, Brasilia, DF, Brazil
g Instituto de Puericultura e Pediatria Margatão Gesteira, Universidade Federal do Rio de Janeiro (IPPMG/UFRJ), Rio de Janeiro, RJ, Brazil
h Grupo Fleury - Divisão de Hematologia e Citometria de Fluxo, São Paulo, SP, Brazil
i Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
j Faculdade de Farmácia, Universidade Federal do Rio Grande do Norte (UFRN), Natal, RN, Brazil
k Universidade Federal de Santa Catarina (UFSC) - Florianópolis, SC, Brazil
l Faculdade de Ciências Médicas, Universidade Estadual de Campinas (FCM Unicamp), Campinas, SP, Brazil
m Hospital Amaral Carvalho, Jaú, SP, Brazil
Ver más
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Tables (6)
Table 1. AL Screening Antibody Panel for 4-Color Immunophenotyping.
Table 2. BCP-ALL Antibodies Panel for 4-Color Immunophenotyping – markers and fluorochromes.
Table 3. BCP-ALL molecular abnormalities and related immunophenotypic profiles.a
Table 4. T-ALL/LBL Antibodies Panel for 4-Color Immunophenotyping – markers and fluorochromes.
Table 5. AML molecular abnormalities and related immunophenotypic profiles.a
Table 6. Monoclonal antibodies combinations recommended for the diagnosis of AML
Show moreShow less
Additional material (2)
Abstract
Introduction

Flow cytometry has become an increasingly important tool in the clinical laboratory for the diagnosis and monitoring of many hematopoietic neoplasms. This method is ideal for immunophenotypic identification of cellular subpopulations in complex samples, such as bone marrow and peripheral blood. In general, 4-color panels appear to be adequate, depending on the assay. In acute leukemias (ALs), it is necessary identify and characterize the population of abnormal cells in order to recognize the compromised lineage and classify leukemia according to the WHO criteria. Although the use of eight- to ten-color immunophenotyping panels is wellestablished, many laboratories do not have access to this technology.

Objective and Method

In 2015, the Brazilian Group of Flow Cytometry (Grupo Brasileiro de Citometria de Fluxo, GBCFLUX) proposed antibody panels designed to allow the precise diagnosis and characterization of AL within available resources. As many Brazilian flow cytometry laboratories use four-color immunophenotyping, the GBCFLUX has updated that document, according to current leukemia knowledge and after a forum of discussion and validation of antibody panels.

Results

Recommendations for morphological analysis of bone marrow smears and performing screening panel for lineage (s) identification of AL were maintained from the previous publication. The lineage-oriented proposed panels for B and T cell acute lymphoblastic leukemia (ALL) and for acute myeloid leukemia (AML) were constructed for an appropriate leukemia classification.

Conclusion

Three levels of recommendations (i.e., mandatory, recommended, and optional) were established to enable an accurate diagnosis with some flexibility, considering local laboratory resources and patient-specific needs.

Keywords:
Flow cytometry
Acute leukemia panel
Monoclonal antibodies panel
ALL
AML
GBCFLUX
Full Text
Introduction

The Brazilian Group of Flow Cytometry (Grupo Brasileiro de Citometria de Fluxo, GBCFLUX) had previously proposed monoclonal antibodies (MoAb) panels to be subsequently validated in an interlaboratory study to assess their effectiveness in the diagnosis and classification of acute leukemia (AL).1 The new flow cytometers (CF) allow immunophenotyping using 8 or more colors,2-4 however, many Brazilian laboratories still work on 4-color multi-parameter flow cytometry (MCF) platforms. The present document aims to update the former reported in 2015.1 The panels previously proposed were revised, based on a literature review and the extensive experience of the professionals participating in this study. The former goals of the panels were maintained: 1) identification and quantification of abnormal leukemia cells, as well as lineage identification by the screening tubes; 2) disease classification according to the cell maturation stage; 3) identification of leukemiaassociated immunophenotypes (LAIPs) to be used for minimal residual disease (MRD) assessments, and; 4) identification of phenotypes associated with molecular alterations with well-recognized prognostic implications.1-5

All the panels were designed at different recommendation levels for diagnosis and classification, allowing flexibility compatible with the local laboratory resources. Three levels of recommendations were considered: mandatory, recommended and optional. Mandatory recommendations contain the minimum criteria for identification, quantification and classification of AL. Recommended level includes markers that are not essential for diagnosis, but are important for leukemia subclassification, prognosis and MRD detection. Optional recommendations include markers useful for MRD evaluation, detection of less frequent leukemia subtypes, associated with molecular or cytogenetic abnormalities, and prognosis, such as CD66c, CD123 and NG2.2 These panels were validated in the group's laboratories to assure their effectiveness (see supplementary files).

General recommendations

The recommended pre-analytical and analytical processes have been previously described.1,6

Detailed patient information is essential for initial evaluation and should contain age, gender, complete blood cell counts, disease phase (diagnosis and monitoring), previous treatment and whether it is a relapse or a secondary transformation. In addition, cytomorphology information is useful as a complementary method to flow cytometry for the appropriate diagnostic assessment of AL. It is therefore also desirable to have information about bone marrow (BM) cytology and a description of the blast cells.

However, the cell morphology analysis should not be used as a screening panel substitute, only as additional diagnostic information.

Although this document intends to standardize the approaches for the diagnosis of acute leukemia AL in 4-color MFC, the GBCFLUX Committee for Acute Leukemia recommends that Brazilian flow cytometry laboratories, when possible, migrate to platforms of 8 or more colors for a more accurate diagnosis and the use of more sensitive methods for MRD detection.

Acute leukemia screening tubes (ALST)

Two tubes are designed to lineage identification of the leukemic blast: B-cell lineage (CD19 and cyC79a), T cell lineage (cyCD3), myeloid (cyMPO) and ambiguous lineage leukemia (Table 1). In addition, some markers were added to improve blast cell identification, such as CD34 as an immaturity marker, CD7, which is very frequent in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL) and in some acute myeloid leukemia (AML), and CD45, that can be used as auxiliary marker for the gating strategy. Compared to the original publication, the 1st tube was maintained and the 2nd tube was slightly modified, with a switch of the CD7 and CD19 fluorochromes. The CD7 is a strong marker and has been combined with fluorescein isothiocyanate (FITC), according to the classic criterion that highlyexpressed markers must be conjugated with a weak fluorochrome.2,6,7 This is a general rule for choosing MoAb and fluorochromes for a panel. For the same reason, we recommended the CD19 conjugated with phycoerythrin (PE): low density markers should be conjugated with bright fluorochromes.6,7

Table 1.

AL Screening Antibody Panel for 4-Color Immunophenotyping.

Tubes  FITC  PE  PerCP-Cy5.5  APC 
cyMPO  cy CD79a  CD45  cy CD3 
CD7  CD19  CD45  CD 34 

FITC: fluorescein isothiocyanate; PE: phycoerythrin; PerCP-Cy5.5: peridinin chlorophyll protein/cyanin5; APC: allophycocyanin; Cy: cytoplasmic.

Based on the immunophenotypic information derived from the ALST tubes, lineage-directed panels [B-cell precursor acute lymphoblastic leukemia/lymphoma (BCP-ALL) or T-lymphoblastic leukemia (T-ALL) or acute myeloid leukemia /myelodysplastic syndrome (AML/MDS)] must be applied in order to provide the final diagnosis (see Tables 1, 2 and 3).

Table 2.

BCP-ALL Antibodies Panel for 4-Color Immunophenotyping – markers and fluorochromes.

Tubes  FITC  PE  PerCP-Cy5.5  APC 
Mandatory or Essential         
CD20  CD10  CD34  CD19 
CyIgM  CD22  CD19  CD123 
CD38  CD66c  CD19  CD81 
Recommended when cyIgM+         
SmKappa  SmLambda  CD19  SmIgM 
Recommended when CD10negative         
CD15+CD65  NG2 (71.)  CD34  CD19 
Optional         
CD58  CD9  CD45or CD34*  CD19 
NuTdT  CD13±CD33  CD45 or CD34*  CD19 

FITC: fluorescein isothiocyanate; PE: phycoerythrin; PerCP-Cy5.5: peridinin chlorophyll protein/cyanin5; APC: allophycocyanin; Cy: cytoplasmic; Nu: nuclear; Sm: surface membrane* according to expression in the screening panel.

Table 3.

BCP-ALL molecular abnormalities and related immunophenotypic profiles.a

Molecular Abnormality  Immunophenotypic expression. 
t(9;22)(q34.1;q11.2); BCR-ABL1  CD19+, CD10+, NuTdT+, CD34+, CD25+, dim expression of CD38. 
t(v;11q23.3); KMT2A-rearranged  Frequent expression of CD66c, CD13 and CD33, CD19+, CD10, CD24, NG2+, CD15/CD65+
t(12;21)(p13.2;q22.1); ETV-RUNX1  CD19+, CD10+, CD34+, CD13+, CD27+. Absence of CD9, CD20, CD66 and CD44. 
ALL with hyperdiploidy  CD19+, CD10+, CD66c+. Most of cases CD34+ and CD45 sometimes negative. 
LLA with hypodiploidy  CD19+ CD10+. Without specific immunophenotype. 
t(5;14)(q31.1;q32.1); IGH/IL3  CD19+, CD10+. Presence of eosinophilia. 
t(1;19)(q23;p13.3); TCF3-PBX1  CD10+, cyIgM+, CD9++ and CD34 or dim. 
a

WHO classification of tumors of hematopoietic and lymphoid tissues,4th ed., Lyon: International Agency for Research on Cancer.

The ALST should be used in all suspected cases of AL, even in those with typical cytomorphological findings, in order to identify the AL lineage and also the ambiguous lineage phenotype (Table 1). However, the screening tube is not enough to reach the final diagnosis. On the other hand, the lineage of some subtypes of leukemia, such as megakaryoblastic leukemia, AML with minimal differentiation, blastic plasmacytoid dendritic cell neoplasm, as well as acute undifferentiated leukemia, cannot be defined by the ALST tube. These AML subtypes often do not express intracytoplasmic myeloperoxidase (cyMPO) nor the lymphoid markers that define the B and T lineage. Thus, in any situation, an expanded assessment should be performed to accurately define the leukemia classification. The rationale for choosing ALST markers has already been described1.

Classification panel for B cell precursor acute lymphoblastic leukemia/lymphoma

BCP-ALL are immature B-cell malignancies involving bone marrow (BM), peripheral blood (PB) and even presenting primary involvement of nodal or extra nodal sites (BCP-LBL). The BCP-ALL immunophenotypically is diagnosed by the differences between the patterns of antigen expression in leukemic cells and their normal counterparts. As a result, the CD19 has been used as a backbone marker of B-cell lineage. Tubes 1 to 4 allow the assessment of B-cell maturation and can detect leukemia cells by "different from normal" cell patterns. The BCP ALL is diagnosed phenotypically by the differences in the constitution of the tubes, compared to the previous recommendations described below (Table 2).

In mandatory tubes, which include markers that allow the identification of normal maturation and asynchronous antigens expression of B cells (Table 2: tube 1), the CD34 FITC, present in the previous combination1 was changed to PerCP-Cy5.5, due to the better performance of this marker in this fluorochrome. In BCP-ALL, the CD38 and CD81 MoAb usually show expressions unlike normal B cell maturation, which is easily identifiable, as well as useful for detecting MRD6.

The rationale for the CD22 in the panel is its usefulness in evaluating the use of anti-CD22 therapy, according to its intensity of expression. It is also useful to replace the CD19 as a B cell marker for MRD detection after anti-CD19 immunotherapy.8 In addition, cross-lineage markers, such as CD66c and CD123, were included as mandatory markers instead of CD13 and CD33 from the previous panel.1 The CD66c and CD123 are more informative for molecular lesions (BCR-ABL fusion gene and hyperdiploidy)5,9,10 and both are more frequently expressed and stable after ALL treatment than the CD13 and CD33, being more useful for further MRD detection.6,11,12

Tube 4 is a complement to tubes 1, 2 and 3 for the classification of ALL, based on the stage of maturation, allowing the diagnosis of mature B-cell leukemia.13 This tube is recommended to be used in combination with the others, because the identification of mature B-cell ALL (CD20+) has therapeutic implications (for example: use of anti-CD20 associated with chemotherapy).14

Tube 5 is recommended for all infants and children with ALL CD10 negative. It is not optional, as in our previous guidelines, because CD10 negativity, added to the expression of NG2, CD15 and CD65 are generally associated with KMT2A rearrangements (MLL), which are more common in pediatric patients.15,16

In addition, the optional tube 6 includes the CD9 and CD58, which provide additional diagnosis information. The CD58 is an interesting LAIP marker since it is highly expressed in malignant B-cells, but found at low levels in normal/regenerating B-cell precursors and mature B-cells.17,18

Tubes 6 and 7 include CD45 or CD34 with CD19, according to expression in the screening panel. The choice between the two markers depends on the most informative antibodies in the previous tubes of the panel.

The NuTdT, CD58, CD13 and CD33 are markers that can provide some additional information, but the most important markers for these two tubes are the CD9 and CD25.19,20 We recommend the use of the CD9 instead of CD25 in the 2nd fluorescence if the phenotype suggests: 1) t (1; 19) or rearrangement TCF3-PBX1, that is, pre-B ALL phenotype with homogeneous CD10 and CD19 and partially positive CD20 with strong positive CD92 if the phenotype suggests t (12; 21) or fusion gene ETV6-RUNX1, that is, a common ALL phenotype, with partial or total loss of CD9.21 On the other hand, we recommend the use of the CD25 if the phenotype suggests Ph + ALL: common B-ALL phenotype with strong homogeneous CD10 and CD34, heterogeneous CD38 (positive to negative) and CD66c positive.2,19-20

Table 3 shows the BCP-ALL molecular abnormalities and related immunophenotypic profiles.22

The performance of the B-ALL diagnostic panel can be seen in figure 1S (supplementary files).

Classification panel for T lymphoblastic leukemia/lymphoma

T-lymphoblastic lymphoma (T-LBL), which is biologically similar to T-acute lymphoblastic leukemia (T-ALL), is derived from immature lymphoid cells of T-cell lineage.23 Bone marrow involvement < 25% BM blasts are classified as T-LBL, while patients with ≥ 25% BM blasts are diagnosed with T-ALL.22 Early T-cell precursor lymphoblastic leukemia (ETP-ALL) is considered a high risk subtype of T-cell ALL/LBL (T-ALL/LBL) by the 2017 WHO classification of hematopoietic neoplasms.22

The differences in the constitution of the tubes, compared to the previous recommendations,1 are described below (Table 4).

Table 4.

T-ALL/LBL Antibodies Panel for 4-Color Immunophenotyping – markers and fluorochromes.

Tubes  FITC  PE  PerCPCy5.5  APC 
Mandatory or Essential         
NuTdT  CD7  CyCD3  CD10 
CD8  CD7  SmCD3  CD4 
CD2  CD7  CD5  CD1a 
CD7  CD99  CyCD3  CD45RA 
CD7  CD13+CD33  CyCD3  CD117 
Recommended         
TCRαβa  TCRγδa  SmCD3  CD56* 
HLA-DR  CD7  CyCD3  CD123 
CD44  CD7  CyCD3 

FITC: fluorescein isothiocyanate; PE: phycoerythrin; PerCP-Cy5.5: peridinin chlorophyll protein/cyanin5; APC: allophycocyanin; cy: cytoplasmic; Nu: nuclear; Sm: surface membrane.

a

when SmCD3 positive or NK cell (*)

The immunophenotypic criteria that are useful in the diagnosis of T-cell neoplasms include the absence, under expression and overexpression of one or more of the pan-T antigens (CD2, SmCD3 and CD5), in addition to the expression of anomalous and cross-lineage antigens.22 The cytoplasm CD3 and/or CD7 has been used as a backbone marker for T- cell lineage.2 Tubes 1 to 3 (Table 3) allow the classification of T-ALL according to the maturation profile of leukemic cells. Tube 4 was designed for later detection of MRD: 1) the CD99 expression is very frequently expressed and recognizes more immature T-ALLs, in addition to being stable after treatment and useful for MRD detection,2,24 and; 2) the CD45RA is expressed only in more immature subtypes of T-ALL and can be useful for detecting MRD.25 Tube 5 is essential for the identification of ETP-ALL, whose characteristics are the co-expression of cyCD3+ and CD5− / low with stem cell markers (CD34 and CD117) or myeloid markers (CD13 and CD33), in the absence of CD1a and CD8 expressions.26,27

Tube 6 is useful for subclassifying T-ALL according to TCR expressions2,28, as well as for diagnosing the precursor NK-ALL / LBL, which is the CD2+, CD7+, CD56+, and even cyCD3+, without expressions of markers from other lineages.2,22 In tubes 7 and 8: the CD123 is a cross lineage marker2,29; the HLA-DR negativity is characteristic of T-ALL,2,22 and; the CD44 is an optional marker for MRD detection. The CD44 upregulation may be involved in T-ALL leukemogenesis30 and it has been reported to be highly expressed in pediatric T-ALL.31,32 Therefore, these markers may provide additional information for the diagnosis of T-ALL.

In comparison with the first guideline, the fluorochromes CD1a, CD2, CD3, CD4 and CD5 were changed, and CD3 and CD7 were maintained at the same fluorescence in the different tubes as backbone markers to standardize the selections of the gates and to minimize the cost of the panels because these markers are in accordance with the previous recommendations for the MRD panels.6 The performance of the T-ALL diagnostic panel can be seen in Figure 2S (supplementary files).

Classification panel for acute myeloid leukemia

Acute myeloid leukemia (AML) is classified by the WHO into different categories: AML with recurrent genetic abnormalities; AML with myelodysplasia-related changes, therapy-related myeloid neoplasms; not otherwise specified (NOS), and; myeloid sarcoma and myeloid proliferations associated with Down syndrome.22Most of them have immunophenotype-associated profiles, diagnosed by multiparametric flow cytometry.22,33

The development of AML from stem cells with specific founder mutations leads to an oligoclonal disease that progresses into a very heterogeneous leukemia at diagnosis.34,35Table 5 provides information on molecular abnormalities in AML and associated immunophenotypes.34 The immunophenotypic alterations observed in AML are asynchronous antigen expression, antigens under- or over-expression and aberrant /cross lineage marker expression.36,37

Table 5.

AML molecular abnormalities and related immunophenotypic profiles.a

Molecular abnormality  Immunophenotypic expression. 
AML with t(8;21)(q22;q22.1); RUNX1-RUNX1T1  CD34+, CD117+, CD13het, CD33brigh, cyMPO+, CD15+/-, CD65+/-, CD19+ and/or CD56+, CD11bneg 
APL with t(15;17)(q24.1;q21.2); PML-RARA  CD45+/-, SSC high (“flame-like”), CD34-, HLA-DR-, CD13+, CD33+ and MPOhigh, CD117+, CD15-, CD16-, CD11b-. 
AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22);CBFB-MYH11  CD4+ and CD36+ in myeloid blasts. Sometimes CD15+ and/or CD65+ and CD11b and CD2+ in the blasts. Presence of monocytic component. 
AML with t(9;11)(p21.3;q23.3); KMT2A-MLLT3  Monocytic markers, sometimes CD4+. CD33+ with CD13-, CD34-, NG2+. 
AML with t(6;9)Ip23;q34.1); DEK-NUP214  CD13+ CD33+ CD117+, CD34+, CD9+. Basophilia. 
AML with inv(3)(q21.3q26.2) or t(3;3)(q21.3;q26.2); GATA2, MECOM  Blasts: CD13+ CD117+, CD33 and MPO often negatives. CD11b+, CD11c+, CD34+. Aberrant: CD7 e CD123 frequently and CD56 in some cases.Megakaryocytic component when present is: CD41+, CD42+, CD61+ and when monocytic present is: CD14 e CD64. 
Acute megakaryoblastic leukemia with t(1;22)(p13.3;q13.1); RBM15-MKL1  CD41+, CD42+ and/or CD61+. 
AML with t(9;22)(q34.1;q11.2); BCR-ABL1  Blasts: expression of myeloid antigens. Frequently CD7+ and CD19+. 
AML with mutation NPM1  Blasts with myeloid expression. SSClow, CD34 and/or HLA-DR (especially in the “cuplike” type). 
AML with mutation biallelic CEBPA  CD34high, CD117 and HLA-DR with asynchronism (CD15+ CD65+). MPO high. Aberrant antigens: CD7 and CD56. CD64 in monocytes and neutrophils.Erythroblasts CD105+ and CD117+ 
AML with RUNX1mutation  Often MPO negative. CD34+ e HLA-DR-. CD33+, CD13+ e CD15+. CD56 + (in 13% of cases) 
a

Modified from Bain BJ Béné MC. Morphological and Immunophenotypic Clues to the WHO Categories of Acute Myeloid Leukaemia. Acta Haematol. 2019; 141:232–244 (58).

Compared to previous recommendations, a few changes have been made. The mandatory panel contains several markers that will contribute to the identification and quantification of blast cells, as well as allow classification according to the lineage differentiation or non-differentiation.

Tubes 1 and 2 (Table 6) allow the characterization of more immature myeloid compartments, including the pre-leukemic stem cells CD34+/CD38, which may be responsible for the leukemia recurrence. Therefore, these cells should also be evaluated in the MRD assays.38–40 The second tube also includes CD56 as a cross-lineage marker, which can be useful in detecting MRD. Other cross-lineage markers are CD7, CD19 and cyCD79a, which are contained in the screening panel, and CD2, contained in tube 10 (Table 6).36,37

Table 6.

Monoclonal antibodies combinations recommended for the diagnosis of AML

Tubes  FITC  PE  PerCPC5.5  APC 
Mandatory/Essential         
HLA-DR  CD117  CD45  CD34 
CD38  CD56  CD45  CD34 
CD16  CD13  CD45  CD11b 
CD15  CD33  CD45  CD34 
CD300e  CD64  CD45  CD14+CD34 
CD36  CD105  CD45  CD71 
HLA-DR  CD123  CD45  CD4 
Recommended         
CD42b+CD61  CD33  CD45  CD34+CD117 
CD45  CD203c  CD34  CD22 
10  CD2  CD25  CD45  CD117 
Optional         
11  CD44  NG2  CD45  CD34 
12  NuTdT  CD10  CD45  CD34 

FITC: fluorescein isothiocyanate; PE: phycoerythrin; PerCP-Cy5.5: peridinin chlorophyll protein/cyanin 5; APC: allophycocyanin.

Tubes 3 to 6 (Table 6) are designed to identify the leukemia committed lineage: neutrophil (3 and 4), monocytic (4 and 5) and erythroid (6)

Originally, tube 4 contained CD61 in the 2nd fluorochrome (FL), but this marker was replaced by CD33, which together with the other markers in tubes 3 and 4 (CD11b, CD13, CD15), allows for a better differentiation between leukemic blasts from the neutrophilic and monocytic lineage, including Acute Promyelocytic Leukemia (APL).

In tube 5, designed for assess monocytic maturation (CD64, CD36, CD14 and IREM2), CD36 was replaced by CD300e (IREM2), which can distinguish the more mature monocytic compartment (CD14+/CD300e+) from the promonocytes (CD14+/CD300e). It is a useful combination for subclassifying monoblastic and monocytic leukemias.41 In addition, CD34 was added to CD14 in the 4th FL to identify more immature monocytic precursors and asynchronisms of antigen expressions in this lineage.

In tube 6, designed for erythroid lineage evaluation (CD71, CD36 and CD105), the glycophorin marker present in the first guideline was removed because it did not offer additional information. The CD105 (endoglin) is expressed in the early stages of erythroid differentiation (CD117+/CD45++/CD34/CD13/HLA-DR cells), remains present after the levels of CD71 and CD36 increase and drops gradually after CD117 is lost, so that more mature red cell precursors no longer express CD105.1,2,42

The last mandatory tube (tube 7) is useful for the diagnosis of blastic plasmocytoid dendritic cell neoplasm (BPDCN).43,44Although this is a less frequent type of leukemia, its correct diagnosis is mandatory due to the severity of the disease and the therapeutic impact of this diagnosis. The CD4 included in this tube is useful for the diagnosis of BPDCN43 and is also expressed in monocytic leukemia.2 Furthermore, it is a cross-lineage marker that can be useful in the detection of the MRD.36

Tubes 8 to 10 are recommended in cases where the tubes described above are not able to subclassify the AML.

The recommended tube 8 corroborates the identification of blast cell involvement with the megakaryocytic lineage, as acute megakaryoblastic leukemia can be positive for CD61, CD41 and CD42.2,45 The CD41 recommended in the first guideline was replaced by CD42a plus CD61FITC due to the higher frequency of these markers in this leukemia subtype.2

Tubes 9 and 10 are intended for the diagnosis of acute basophilic leukemia and mast cell diseases respectively, including mast cell leukemia.2,46,47 The changes in the fluorochromes of CD22 and CD45 in tube 9 were justified by the better performance of CD22 in APC than in FITC and the good performance of CD45 in FITC. In addition, in tube 10 there are the CD2 and CD25 which can be useful markers for MRD purposes and prognosis in AML.48–50

Optional tube 11 contains markers to detect leukemic stem cells (CD44)38,39 and immunophenotype associated with KMT2A(MLL) rearrangements (NG2).51,52

Currently, the optional tube 12 included here is useful in the following circumstances: i) the TdT expression in myeloid and lymphoid precursors, ii) for the evaluation of lymphoid precursor cells (type I), and; iii) the CD10 expression in granulocytic cells in MDS. The CD10 and TdT unrelated to the lymphoid lineage are also good markers for detecting MRD.36,37

The AL screening and AML panel include several markers that detect the aberrant expression of lymphoid-associated antigens (CD19, cyCD79a, CD7, CD2, CD4 and CD56) and asynchronous antigen expression (CD14, CD15, CD16 and CD11b). Some of them can be useful in further MRD assessment.36,37,53 The performance of the AML diagnostic panel can be seen in Figure 3S (supplementary files).

Conclusion

In summary, this document presents updated guidelines for the use of validated 4-color panels considered relevant in the diagnosis of acute leukemia. The GBCFLUX Acute Leukemia Committee has revised the monoclonal antibody consensus panel to provide a well-defined diagnosis of acute leukemia for clinical flow cytometry laboratories, including those with limited resources. This technical standardization should improve the quality of the acute leukemia diagnostics among different laboratories. In addition, this work paves the way for multicenter cooperative studies, promoting scientific and technological advances in clinical flow cytometry in Brazil.

Finally, these diagnostic guidelines can be replicated in laboratories that still use 4-color cytometers until they migrate to the 8- to 12-color ones.

Acknowledgements

The group thanks Joana Espricigo Conte-Spilari, Camila Marques Bertolucci, Alef Rafael Severino, Diana AC Kluck, Raysa Rosseto Rodrigues, Vanessa Kukla and Regielly Cognialli for the technical support. This research was not funded by any specific grant from funding agencies in the public, commercial or not-for-profit sectors.

References
[1]
MR Ikoma, AF Sandes, TS Leand, GB Cavalcanti Júnior, IGH Lorand-Metze, ES Costa, et al.
First Proposed Panels on Acute Leukemia for Four-Color Immunophenotyping by Flow Cytometry from the Brazilian Group of Flow Cytometry-GBCFLUX.
Cytometry B, 88B (2015), pp. 194-203
[2]
JJ van Dongen, L Lhermitte, S Bottcher, J Almeida, VH van der Velden, J Flores-Montero, et al.
EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes.
Leukemia, 26 (2012), pp. 1908-1975
[3]
B. Wood.
9-color and 10-color flow cytometry in the clinical laboratory.
Arch Pathol Lab Med, 130 (2006), pp. 680-690
[4]
S Cherian, G Levin, WY Lo, M Mauck, D Kuhn, C Lee, BL. Wood.
Evaluation of an 8-color flow cytometric reference method for white blood cell differential enumeration.
Cytometry B Clin Cytometry, 78 (2010), pp. 319-328
[5]
T Kalina, M Vaskova, E Mejstrikova, J Madzo, J Trka, J Stary, O. Hrusak.
Myeloid antigens in childhood lymphoblastic leukemia: clinical data point to regulation of CD66c distinct from other myeloid antigens.
BMC Cancer, 5 (2005), pp. 38
[6]
Ikoma-Colturato MRV, Beltrame MP, Furtado FM, Pimenta Costa ES, Azambuja AP, M Malvezzi, Yamamoto M. Hematology, Transfus Cell Ther. 2020. 10.1016/j.htct.2020.09.148.
[7]
Flores-Montero J, Kalina T, Corral-Mateos A, Sanoja-Flores L, Perez-Andres M, Martin-Ayuso M, et al. Fluorochrome choices for multi-color flow cytometry J Immunol Methods 2019. 10.1016/j.jim.2019.06.009.
[8]
Cherian S, Miller V, McCullouch V, Dougherty K, Fromm JR, Wood BL. A novel flow cytometric assay for detection of residual disease in patients with B Lymphoblastic Leukemia/Lymphoma post anti-CD19 therapy. Cytometry B 2016 doi: 10.1002/cyto.b.21482.
[9]
MD Tabernero, AM Bortoluci, I Alaejos, MC Lopez-Berges, A Rasillo, R Garcia-Sanz, et al.
Adult precursor B-ALL with BCR/ABL gene rearrangements displays a unique immunophenotype based on the pattern of CD10, CD34, CD13 and CD38 expression.
Leukemia, 15 (2001), pp. 406-414
[10]
M Djokic, E Björklund, E Blennow, J Mazur, S Söderhäll, A. Porwit.
Overexpression of CD123 correlates with the hyperdiploid genotype in acute lymphoblastic leukemia.
Haematologica, 94 (2009), pp. 1016-1019
[11]
GS Tang, J Wu, M Liu, H Chen, SG Gong, JM Yang, et al.
BCR-ABL1 and CD66c exhibit high concordance in minimal residual disease detection of adult B-acute lymphoblastic leukemia.
Am J Transl Res, 7 (2015), pp. 632-639
[12]
P Theunissen, E Mejstrikova, L Sedek, AJ van der Sluijs-Gelling, G Gaipa, M Bartels, et al.
Standardized flow cytometry for highly sensitive MRD measurements in B-cell acute lymphoblastic leukemia.
[13]
MC Bene, T Nebe, P Bettelheim, B Buldini, H Bumbea, W Kern, et al.
Immunophenotyping of acute leukemia and lymphoproliferative disorders: a consensus proposal of the European Leukemia Net Work Package 10.
Leukemia, 25 (2011), pp. 567-574
[14]
L Levato, S. Molica.
Rituximab in the management of acute lymphoblastic leukemia.
Exp Op on Biol Therapy, 18 (2018), pp. 221-226
[15]
C Malouf, K. Ottersbach.
Molecular processes involved in B cell acute lymphoblastic Leukaemia Cell. Mol.
Life Sci, 75 (2018), pp. 417-446
[16]
C Bueno, R Montes, L Martin, I Prat, MC Hernandez, A Orfao.
Menendez P NG2 antigen is expressed in CD34+ HPCs and plasmacytoid dendritic cell precursors: is NG2 expression in leukemia dependent on the target cell where leukemogenesis is triggered?.
Leukemia, 22 (2008), pp. 1475-1478
[17]
M Veltroni, L De Zen, MC Sanzari, O Maglia, MN Dworzak, R Ratei, et al.
Expression of CD58 in normal, regenerating and leukemic bone marrow B cells: implications for the detection of minimal residual disease in acute lymphocytic leukemia.
Haematologica, 88 (2003), pp. 1245-1252
[18]
RV Lee, RC Braylan, LM. Rimsza.
CD58 expression decreases as nonmalignant B cells mature in bone marrow and is frequently overexpressed in adult and pediatric precursor B-cell acute lymphoblastic leukemia.
Am J Clin Pathol, 123 (2005), pp. 119-124
[19]
TM Owaidah, FI Rawas, MF Al Khayatt, NB Elkum.
Expression of CD66c and CD25 in acute lymphoblastic leukemia as a predictor of the presence of BCR/ABL rearrangement.
Hematol Oncol Stem Cell Ther, 1 (2008), pp. 34-37
[20]
K Blatt, I Menzl, G Eisenwort, S Cerny-Reiterer, H Herrmann, S Herndlhofer, et al.
Phenotyping and Target Expression Profiling of CD34 +/CD38 – and CD34 +/CD38 + Stem- and Progenitor cells in Acute Lymphoblastic Leukemia.
Neoplasia, 20 (2018), pp. 632-642
[21]
CB Blunck, E Terra-Granado, EP Noronha, G Wajnberg, F Passetti, MS Pombo-de-Oliveira, M. Emerenciano.
CD9 predicts ETV6-RUNX1 in childhood B-cell precursor acute lymphoblastic leukemia.
Hematol Transfus Cell Ther, 41 (2019), pp. 205-211
[22]
MJ Borowitz, MC Bene, NL Harris, A Porwit, E. Matutes.
Acute leukaemias of ambiguous lineage.
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, 4th edition, pp. 209-212
[23]
E Kroeze, JLC Loeffen, VM Poort, JPP. Meijerink.
T-cell lymphoblastic lymphoma and leukemia: different diseases from a common premalignant progenitor?.
Blood Adv, 4 (2020), pp. 3466-3473
[24]
MN Dworzak, G Fröschl, D Printz, L De Zen, G Gaipa, R Ratei, et al.
CD99 expression in T-lineage ALL: implications for flow cytometric detection of minimal residual disease.
Leukemia, 18 (2004), pp. 703-708
[25]
EM Schiavone, CL Pardo, R Di Noto, C Manzo, F Ferrara, C Vacca, L. Del Vecchio.
Expression of the leukocyte common antigen (LCA, CD45) isoforms RA and RO in acute haematological malignancies: possible relevance in the definition of new overlap points between normal and leukaemic haemopoiesis.
Br J Haematol, 91 (1995), pp. 899-906
[26]
E Coustan-Smith, CG Mullighan, M Onciu, FG Behm, SC Raimondi, D Pei, et al.
Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia.
Lancet Oncol, 10 (2009), pp. 147-156
[27]
J Zhang, L Ding, L Holmfeldt, G Wu, SL Heatley, D Payne-Turner, et al.
The genetic basis of early T-cell precursor acute lymphoblastic leukaemia.
Nature, 481 (2012), pp. 157-163
[28]
V Asnafi, K Beldjord, E Boulanger, B Comba, P Tutour, MH Estienne, et al.
Analysis of TCR, pT_, and RAG-1 in T-acute lymphoblastic leukemias improves understanding of early human T-lymphoid lineage commitment.
Blood, 101 (2003), pp. 2693-2703
[29]
AE Bras, V Haas, A Stigt, M Jongen-Lavrencic, HB Beverloo, JG Marvelde, et al.
CD123 Expression Levels in 846 Acute Leukemia Patients Based on Standardized Immunophenotyping.
Cytometry B, 96B (2019), pp. 134-142
[30]
M Garcia-Peydró, M Mosquera, MJ Garcia-Léon, J Alcain, A Rodriguez, PG Miguel, et al.
The NOTCH1/CD44 axis drives pathogenesis in a T cell acute lymphoblastic leukemia model.
J Clin Invest, 128 (2018), pp. 2802-2818
[31]
GB Cavalcante Júnior, W Savino, MS Pombo-de-Oliveira.
Braz J Med Biol Res, 27 (1994), pp. 2259-2266
[32]
LVC Marques, EP Noronha, FG Andrade, FV Santos-Bueno, MB Mansur, E Terra-Granado, MS. Pombo-de-Oliveira.
CD44 Expression Profile Varies According to Maturational Subtypes and Molecular Profiles of Pediatric T-Cell Lymphoblastic Leukemia.
Front Oncol, 8 (2018), pp. 1-10
[33]
O Hrusak, A. Porwit-MacDonald.
Antigen expression patterns reflecting genotype of acute leukemias.
Leukemia, 16 (2002), pp. 1233-1258
[34]
BJ Bain, MC. Béné.
Morphological and immunophenotypic clues to the WHO Categories of Acute Myeloid Leukaemia.
Acta Haematol, 141 (2019), pp. 232-244
[35]
JS Welch, TJ Ley, DC Link, CA Miller, DE Larsson, DC Koboldt, et al.
The Origin and Evolution of Mutations in Acute Myeloid Leukemia.
[36]
Wood BL Acute.
Myeloid Leukemia Minimal Residual Disease Detection: The Difference from Normal Approach.
Curr Protocols Cytometry, 93 (2020), pp. 1-16
[37]
G Rossi, V Giambra, MM Minervini, C Waure, S Mancinelli, M Ciavarella, et al.
Leukemia-associated immunophenotypes subdivided in “categories of specificity” improve de sensitivity of minimal residual disease in predicting relapse in acute myeloid leukemia.
Cytometry, 98 (2020), pp. 216-225
[38]
W Zeijlemaker, A Kelder, YJM Oussoren-Brockhoff, WJ Scholten, AN Snel, D Veldhuizen, et al.
A simple one-tube assay for immunophenotypical quantification of leukemic stem cells in acute myeloid leukemia.
Leukemia, 30 (2016), pp. 439-446
[39]
W Zeijlemaker, T Grob, R Meijer, D Hanekamp, A Kelder, JC Carbaat-Ham, et al.
CD34+CD38− leukemic stem cell frequency to predict outcome in acute myeloid leukemia.
Leukemia, 33 (2019), pp. 1102-1112
[40]
DM Bahia-Kerbauy, EY Kimura, ML Chauffaille, JO Bordin, J Kerbauy, M. Yamamoto.
The singular value of CD34 and CD117 expression for minimal residual disease detection in AML.
Leuk Res, 27 (2003), pp. 1069-1070
[41]
S Matarraz, J Almeida, J Flores-Montero, Q Lecrevisse, V Guerri, A Lopez, et al.
Introduction to the Diagnosis and Classification of Monocytic-Lineage Leukemias by Flow Cytometry.
Cytometry B, 92 (2017), pp. 218-227
[42]
Orfao A, Matarraz S, Pérez-Andrés M, Almeida J, Teodosio C, Berkowska MA, et al. Immunophenotypic dissection of normal hematopoiesis. J Immunol Methods 2018 10.1016/j.jim.2019.112684.
[43]
C Bueno, J Almeida, P Lucio, J Marco, R Garcia, JM de Pablos, et al.
Incidence and characteristics of CD4(+)/HLA DRhi dendritic cell malignancies.
Haematologica, 89 (2004), pp. 58-69
[44]
F Garnache-Ottou, J Feuillard, C Ferrand, S Biichle, F Trimoreau, E Seilles, et al.
Extended diagnostic criteria for plasmacytoid dendritic cell leukaemia.
Br J Haematol, 145 (2009), pp. 624-636
[45]
DA Arber, R.D. Brunning, A. Orazi, A. Porwit, L.C. Peterson, J. Thiele, et al.
Acute myeloid leukemia and related precursor neoplasms.
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, 4th edition, pp. 162-164
[46]
A Staal-Viliare, V Latger-Cannard, J Didion, MJ Gregoire, T Lecompte, P Jonveaux, Y. Rio.
CD203c /CD117-, an useful phenotype profile for acute basophilic leukaemia diagnosis in cases of undifferentiated blasts.
Leuk Lymphoma, 48 (2007), pp. 439-441
[47]
J Gotlib, A Pardanani, C Akin, A Reiter, T George, O Hermine, et al.
International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) & European Competence Network on Mastocytosis (ECNM) consensus response criteria in advanced systemic mastocytosis.
Blood, 121 (2013), pp. 2393-2401
[48]
E Coustan-Smith, G Song, S Shurtleff, AEJ Yeoh, WJ Chng, SP Chen, et al.
Universal monitoring of minimal residual disease in acute myeloid leukemia.
JCI Insight, 3 (2018), pp. e98561
[49]
TS Solinge, W Zeijlemaker, GJ Ossenkoppele, J Cloos, GJ. Schuurhuis.
The Interference of Genetic Associations in Establishing the Prognostic Value of the Immunophenotype in Acute Myeloid Leukemia.
Cytometry B, 94B (2018), pp. 151-158
[50]
Gönen M, Sun Z, Figueroa ME, Patel JP, Abdel-Wahab O, Racevskis J et al. CD25 Expression Status Improves Prognostic Risk Classification in AML Independent of Established Biomarkers: ECOG Phase III Trial, E1900. Blood 2012. doi:10.1182/blood-2012-02-414425.
[51]
C Wuchter, J Harbott, C Schoch, S Schnittger, A Borkhardt, L Karawajew, et al.
Detection of acute leukemia cells with mixed lineage leukemia (MLL) gene rearrangements by flow cytometry using monoclonal antibody 7.1.
Leukemia, 14 (2000), pp. 1232-1238
[52]
FO Smith, C Rauch, DE Williams, CJ March, D Arthur, J Hilden, et al.
The Human Homologue of Rat NG2, a Chondroitin Sulfate Proteoglycan, Is Not Expressed on the Cell Surface of Normal Hematopoietic Cells But Is Expressed by Acute Myeloid Leukemia Blasts From Poor-Prognosis Patients With Abnormalities of Chromosome Band llq23.
Blood, 87 (1996), pp. 1123-1133
[53]
DM Bahia, M Yamamoto, L Chauffaille Mde, EY Kimura, JO Bordin, MA Filgueiras, J Kerbauy.
Aberrant phenotypes in acute myeloid leukemia: a high frequency and its clinical significance.
Haematologica, 86 (2001), pp. 801-806
Idiomas
Hematology, Transfusion and Cell Therapy
Article options
Tools
Supplemental materials