High levels of CD34+CD38low/-CD123+ blasts are predictive of an adverse outcome in acute myeloid leukemia: a Groupe Ouest-Est des Leucemies Aigues et Maladies du Sang (GOELAMS) study

François Vergez, Alexa S Green, Jerome Tamburini, Jean-Emmanuel Sarry, Baptiste Gaillard, Pascale Cornillet-Lefebvre, Melanie Pannetier, Aymeric Neyret, Nicolas Chapuis, Norbert Ifrah, François Dreyfus, Stéphane Manenti, Cecile Demur, Eric Delabesse, Catherine Lacombe, Patrick Mayeux, Didier Bouscary, Christian Recher, Valerie Bardet, François Vergez, Alexa S Green, Jerome Tamburini, Jean-Emmanuel Sarry, Baptiste Gaillard, Pascale Cornillet-Lefebvre, Melanie Pannetier, Aymeric Neyret, Nicolas Chapuis, Norbert Ifrah, François Dreyfus, Stéphane Manenti, Cecile Demur, Eric Delabesse, Catherine Lacombe, Patrick Mayeux, Didier Bouscary, Christian Recher, Valerie Bardet

Abstract

Background: Acute myeloid leukemias arise from a rare population of leukemic cells, known as leukemic stem cells, which initiate the disease and contribute to frequent relapses. Although the phenotype of these cells remains unclear in most patients, these cells are enriched within the CD34(+)CD38(low/-) compartment expressing the interleukin-3 alpha chain receptor, CD123. The aim of this study was to determine the prognostic value of the percentage of blasts with the CD34(+)CD38(low/-)CD123(+) phenotype.

Design and methods: The percentage of CD34(+)CD38(low/-)CD123(+) cells in the blast population was determined at diagnosis using flow cytometry. One hundred and eleven patients under 65 years of age with de novo acute myeloid leukemia and treated with intensive chemotherapy were retrospectively included in the study. Correlations with complete response, disease-free survival and overall survival were evaluated with univariate and multivariate analyses.

Results: A proportion of CD34(+)CD38(low/-)CD123(+) cells greater than 15% at diagnosis and an unfavorable karyotype were significantly correlated with a lack of complete response. By logistic regression analysis, a percentage of CD34(+)CD38(low/-)CD123(+) higher than 15% retained significance with an odds ratio of 0.33 (0.1-0.97; P=0.044). A greater than 1% population of CD34(+)CD38(low/-)CD123(+) cells negatively affected disease-free survival (0.9 versus 4.7 years; P<0.0001) and overall survival (1.25 years versus median not reached; P<0.0001). A greater than 1% population of CD34(+)CD38(low/-)CD123(+) cells retained prognostic significance for both parameters after multivariate analysis.

Conclusions: The percentage of CD34(+)CD38(low/-)CD123(+) leukemic cells at diagnosis was significantly correlated with response to treatment and survival. This prognostic marker might be easily adopted in clinical practice to rapidly identify patients at risk of treatment failure.

Figures

Figure 1.
Figure 1.
The percentage of CD34+CD38low/−CD123+ leukemic cells is highly variable in AML patients. Blasts and mononuclear cells from 111 AML samples were analyzed by flow cytometry. First, bulk cells (blue and orange) and lymphocytes (purple) were gated according to CD45 intensity in a CD45/sidescatter dot plot: dot plot 1 in patient #24 (A) and patient #87 (B); CD45 intensity of AML blasts is low/dim whereas it is bright on lymphocytes. Blast expression of CD34 and CD38 was then assessed in a second dot plot (dot plot 2). Isotype controls were used as negative controls. Gate a focus on bulk cells with CD34+CD38low/− phenotype. Finally, CD123 expression on CD34+CD38low/−cells (in orange, gate a) was assessed in a third dot plot (dot plot 3 in patients #24 and #87) in order to exclude normal CD34+CD38low/− hematopoietic stem cells that do not express CD123. Percentages of CD34+CD38low/−CD123+ leukemic cells (gate b) for patients #24 and #87 are 0.78% and 10.81%, respectively. Isotype controls are shown in dot plots 4 and 5.
Figure 2.
Figure 2.
Comparison of disease-free and overall survival of AML patients according to CD34+CD38low/−CD123+ percentage. (A) All patients. (B) Intermediate cytogenetics group. (C) Favorable cytogenetics group.

References

    1. Steffen B, Muller-Tidow C, Schwable J, Berdel WE, Serve H. The molecular pathogenesis of acute myeloid leukemia. Crit Rev Oncol Hematol. 2005;56(2):195–221.
    1. Dick JE. Acute myeloid leukemia stem cells. Ann NY Acad Sci. 2005;1044:1–5.
    1. Fernandez HF, Sun Z, Yao X, Litzow MR, Luger SM, Paietta EM, et al. Anthracycline dose intensification in acute myeloid leukemia. N Engl J Med. 2009;361(13):1249–59.
    1. Deschler B, Lubbert M. Acute myeloid leukemia: epidemiology and etiology. Cancer. 2006;107(9):2099–107.
    1. Schlenk RF, Dohner K, Krauter J, Frohling S, Corbacioglu A, Bullinger L, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358(18):1909–18.
    1. Zhi L, Wang M, Rao Q, Yu F, Mi Y, Wang J. Enrichment of N-Cadherin and Tie2-bearing CD34+/CD38−/CD123+ leukemic stem cells by chemotherapy-resistance. Cancer Lett. 2010;296(1):65–73.
    1. Lane SW, Scadden DT, Gilliland DG. The leukemic stem cell niche: current concepts and therapeutic opportunities. Blood. 2009;114(6):1150–7.
    1. Guan Y, Gerhard B, Hogge DE. Detection, isolation, and stimulation of quiescent primitive leukemic progenitor cells from patients with acute myeloid leukemia (AML) Blood. 2003;101(8):3142–9.
    1. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367(6464):645–8.
    1. Taussig DC, Miraki-Moud F, Anjos-Afonso F, Pearce DJ, Allen K, Ridler C, et al. Anti-CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initiating cells. Blood. 2008;112(3):568–75.
    1. Taussig DC, Vargaftig J, Miraki-Moud F, Griessinger E, Sharrock K, Luke T, et al. Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34(−) fraction. Blood. 2010;115(10):1976–84.
    1. Martelli MP, Pettirossi V, Thiede C, Bonifacio E, Mezzasoma F, Cecchini D, et al. CD34+ cells from AML with mutated NPM1 harbor cytoplasmic mutated nucleophosmin and generate leukemia in immunocompromised mice. Blood. 2010;116(19):3907–22.
    1. Sarry JE, Murphy K, Perry R, Sanchez PV, Secreto A, Keefer C, et al. Human acute myelogenous leukemia stem cells are rare and heterogeneous when assayed in NOD/SCID/IL2Rgammac-deficient mice. J Clin Invest. 2011;121(1):384–95.
    1. Jordan CT, Upchurch D, Szilvassy SJ, Guzman ML, Howard DS, Pettigrew AL, et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia. 2000;14(10):1777–84.
    1. Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka S, et al. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol. 2007;25(11):1315–21.
    1. Ebinger M, Witte KE, Ahlers J, Schafer I, Andre M, Kerst G, et al. High frequency of immature cells at diagnosis predicts high minimal residual disease level in childhood acute lymphoblastic leukemia. Leuk Res. 2010;34(9):1139–42.
    1. van Rhenen A, Feller N, Kelder A, Westra AH, Rombouts E, Zweegman S, et al. High stem cell frequency in acute myeloid leukemia at diagnosis predicts high minimal residual disease and poor survival. Clin Cancer Res. 2005;11(18):6520–7.
    1. Witte KE, Ahlers J, Schafer I, Andre M, Kerst G, Scheel-Walter HG, et al. High proportion of leukemic stem cells at diagnosis is correlated with unfavorable prognosis in childhood acute myeloid leukemia. Pediatr Hematol Oncol. 2011;28(2):91–9.
    1. Chevallier P, Prebet T, Pigneux A, Hunault M, Delaunay J, Perry F, et al. Influence of NPM1 and FLT3-ITD status on outcome in relapsed/refractory AML patients receiving salvage therapy including gemtuzumab ozogamicin. Leukemia. 2010;24(2):467–9.
    1. van der Pol MA, Feller N, Roseboom M, Moshaver B, Westra G, Broxterman HJ, et al. Assessment of the normal or leukemic nature of CD34+ cells in acute myeloid leukemia with low percentages of CD34 cells. Haematologica. 2003;88(9):983–93.
    1. Cheson BD, Bennett JM, Kopecky KJ, Buchner T, Willman CL, Estey EH, et al. Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia. J Clin Oncol. 2003;21(24):4642–9.
    1. Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, Theil KS, Mohamed A, et al. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study. Blood. 2000;96(13):4075–83.
    1. Stone RM. Prognostic factors in AML in relation to abnormal karyotype. Best Pract Res Clin Haematol. 2009;22(4):523–8.
    1. Guzman ML, Neering SJ, Upchurch D, Grimes B, Howard DS, Rizzieri DA, et al. Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood. 2001;98(8):2301–7.
    1. Kern W, Bacher U, Haferlach C, Schnittger S, Haferlach T. The role of multiparameter flow cytometry for disease monitoring in AML. Best Pract Res Clin Haematol. 2010;23(3):379–90.
    1. Jin L, Lee EM, Ramshaw HS, Busfield SJ, Peoppl AG, Wilkinson L, et al. Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Cell stem cell. 2009;5(1):31–42.
    1. Kugler M, Stein C, Kellner C, Mentz K, Saul D, Schwenkert M, et al. A recombinant trispecific single-chain Fv derivative directed against CD123 and CD33 mediates effective elimination of acute myeloid leukaemia cells by dual targeting. Br J Haematol. 2010;150(5):574–86.
    1. Stein C, Kellner C, Kugler M, Reiff N, Mentz K, Schwenkert M, et al. Novel conjugates of single-chain Fv antibody fragments specific for stem cell antigen CD123 mediate potent death of acute myeloid leukaemia cells. Br J Haematol. 2010;148(6):879–89.

Source: PubMed

3
Se inscrever