Impact of FLT3 Mutation on Outcomes after Venetoclax and Azacitidine for Patients with Treatment-Naïve Acute Myeloid Leukemia

Marina Konopleva, Michael J Thirman, Keith W Pratz, Jacqueline S Garcia, Christian Recher, Vinod Pullarkat, Hagop M Kantarjian, Courtney D DiNardo, Monique Dail, Yinghui Duan, Brenda Chyla, Jalaja Potluri, Catherine L Miller, Andrew H Wei, Marina Konopleva, Michael J Thirman, Keith W Pratz, Jacqueline S Garcia, Christian Recher, Vinod Pullarkat, Hagop M Kantarjian, Courtney D DiNardo, Monique Dail, Yinghui Duan, Brenda Chyla, Jalaja Potluri, Catherine L Miller, Andrew H Wei

Abstract

Purpose: To evaluate efficacy and safety of venetoclax + azacitidine among treatment-naïve patients with FLT3-mutant acute myeloid leukemia.

Patients and methods: Data were pooled from patients enrolled in a phase III study (NCT02993523) that compared patients treated with venetoclax + azacitidine or placebo + azacitidine and a prior phase Ib study (NCT02203773) where patients were treated with venetoclax + azacitidine. Enrolled patients were ineligible for intensive therapy due to age ≥75 years and/or comorbidities. Patients on venetoclax + azacitidine received venetoclax 400 mg orally (days 1-28) and azacitidine (75 mg/m2; days 1-7/28-day cycle). FLT3 mutation was analyzed centrally on pretreatment bone marrow aspirates.

Results: In the biomarker evaluable population, FLT3 mutation was detected in 42 (15%) and 22 (19%) patients in the venetoclax + azacitidine and azacitidine groups. Composite complete remission [CRc; complete remission (CR) + CR with incomplete hematologic recovery (CRi)] rates (venetoclax + azacitidine/azacitidine) for FLT3-mutant patients were 67%/36%, median duration of remission (DoR) was 17.3/5.0 months, and median OS was 12.5/8.6 months. The CRc rates among FLT3 wild-type patients were 67%/25%, median DoR 18.4/13.4 months, and median OS 14.7/10.1 months. In patients treated with venetoclax + azacitidine, CRc in patients with FLT3-ITD and FLT3-TKD was 63% and 77% and median OS was 9.9 and 19.2 months, and in comutated FLT3-ITD + NPM1 patients, CRc was 70%, median DoR was not reached, and median OS was 9.1 months. There were no unexpected toxicities in the venetoclax + azacitidine group.

Conclusions: When treated with venetoclax + azacitidine, patients with FLT3 mutations and FLT3 wild-type had similar outcomes. Future analyses in larger patient populations may further define the impact of venetoclax + azacitidine in patients harboring FLT3-ITD. See related commentary by Perl and Vyas, p. 2719.

©2022 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
Venetoclax and azacitidine work synergistically to overcome antiapoptotic signals downstream of an activated FLT3 pathway.
Figure 2.
Figure 2.
Study design and molecular classification. *One patient in the venetoclax and azacitidine group and one patient in the azacitidine group had both FLT3-ITD and TKD mutation; †One patient in the venetoclax and azacitidine group was indeterminate for NPM1 comutation status.
Figure 3.
Figure 3.
A, Remission rates in patients with FLT3 mutations and FLT3 wild type by treatment groups. B, Remission rates in patients with FLT3-ITD and FLT3-TKD in the venetoclax and azacitidine group. C, Remission rates in patients with FLT3-ITD+NPM1-mutated versus FLT3-ITD+NPM1 wild type in the venetoclax and azacitidine group. D, Duration of remission among responders with FLT3-ITD+NPM1 mutation versus FLT3-ITD+ NPM1 wild type.
Figure 4.
Figure 4.
Kaplan–Meier curves for OS. A, Patients with FLT3 mutation by treatment groups. B, Patients with FLT3 mutation or FLT3 wild type in the venetoclax and azacitidine group. C, Patients with FLT3-ITD and FLT3-TKD in the venetoclax and azacitidine group. D, Patients with FLT3-ITD+NPM1 mutation versus FLT3-ITD+ NPM1 wild type in the venetoclax and azacitidine group.

References

    1. Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, et al. . Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med 2016;374:2209–21.
    1. Schneider F, Hoster E, Schneider S, Dufour A, Benthaus T, Kakadia PM, et al. . Age-dependent frequencies of NPM1 mutations and FLT3-ITD in patients with normal karyotype AML (NK-AML). Ann Hematol 2012;91:9–18.
    1. Grafone T, Palmisano M, Nicci C, Storti S. An overview on the role of FLT3-tyrosine kinase receptor in acute myeloid leukemia: biology and treatment. Oncol Rev 2012;6:e8.
    1. Bacher U, Haferlach C, Kern W, Haferlach T, Schnittger S. Prognostic relevance of FLT3-TKD mutations in AML: the combination matters—an analysis of 3082 patients. Blood 2008;111:2527–37.
    1. Gale RE, Green C, Allen C, Mead AJ, Burnett AK, Hills RK, et al. . The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood 2008;111:2776–84.
    1. Kayser S, Schlenk RF, Londono MC, Breitenbuecher F, Wittke K, Du J, et al. . Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome. Blood 2009;114:2386–92.
    1. Schlenk RF, Kayser S, Bullinger L, Kobbe G, Casper J, Ringhoffer M, et al. . Differential impact of allelic ratio and insertion site in FLT3-ITD–positive AML with respect to allogeneic transplantation. Blood 2014;124:3441–9.
    1. Huang Y, Hu J, Lu T, Luo Y, Shi J, Wu W, et al. . Acute myeloid leukemia patient with FLT3-ITD and NPM1 double mutation should undergo allogeneic hematopoietic stem cell transplantation in CR1 for better prognosis. Cancer Manag Res 2019;11:4129–42.
    1. Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, et al. . Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017;129:424–47.
    1. National Comprehensive Cancer Network. NCCN Guidelines for patients with Acute Myeloid Leukemia, Version 3.2021. 2021.
    1. Stone RM, Mandrekar SJ, Sanford BL, Laumann K, Geyer S, Bloomfield CD, et al. . Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 Mutation. N Engl J Med 2017;377:454–64.
    1. Perl AE, Martinelli G, Cortes JE, Neubauer A, Berman E, Paolini S, et al. . Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N Engl J Med 2019;381:1728–40.
    1. Cortes J, Perl AE, Döhner H, Kantarjian H, Martinelli G, Kovacsovics T, et al. . Quizartinib, an FLT3 inhibitor, as monotherapy in patients with relapsed or refractory acute myeloid leukaemia: an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol 2018;19:889–903.
    1. Maiti A, DiNardo CD, Daver NG, Rausch CR, Ravandi F, Kadia TM, et al. . Triplet therapy with venetoclax, FLT3 inhibitor and decitabine for FLT3-mutated acute myeloid leukemia. Blood Cancer J 2021;11:25.
    1. Astellas. Astellas reports XOSPATA® (gilteritinib) in combination with azacitidine did not meet endpoint of overall survival in newly diagnosed FLT3 mutation-positive acute myeloid leukemia patients ineligible for intensive induction chemotherapy Tokyo 2020.
    1. Adams JM, Cory S. The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene 2007;26:1324–37.
    1. Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood 2002;100:1532–42.
    1. Choudhary C, Olsen JV, Brandts C, Cox J, Reddy PN, Böhmer FD, et al. . Mislocalized activation of oncogenic RTKs switches downstream signaling outcomes. Mol Cell 2009;36:326–39.
    1. Choudhary C, Schwäble J, Brandts C, Tickenbrock L, Sargin B, Kindler T, et al. . AML-associated Flt3 kinase domain mutations show signal transduction differences compared with Flt3 ITD mutations. Blood 2005;106:265–73.
    1. Mizuki M, Fenski R, Halfter H, Matsumura I, Schmidt R, Müller C, et al. . Flt3 mutations from patients with acute myeloid leukemia induce transformation of 32D cells mediated by the Ras and STAT5 pathways. Blood 2000;96:3907–14.
    1. Takahashi S. Downstream molecular pathways of FLT3 in the pathogenesis of acute myeloid leukemia: biology and therapeutic implications. J Hematol Oncol 2011;4:13.
    1. DiNardo CD, Tiong IS, Quaglieri A, MacRaild S, Loghavi S, Brown FC, et al. . Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood 2020;135:791–803.
    1. Jin S, Cojocari D, Purkal JJ, Popovic R, Talaty NN, Xiao Y, et al. . 5-Azacitidine induces NOXA to prime AML cells for venetoclax-mediated apoptosis. Clin Cancer Res 2020;26:3371.
    1. Singh Mali R, Zhang Q, DeFilippis RA, Cavazos A, Kuruvilla VM, Raman J, et al. . Venetoclax combines synergistically with FLT3 inhibition to effectively target leukemic cells in FLT3-ITD+ acute myeloid leukemia models. Haematologica 2021;106:1034–46.
    1. Raghuveer Singh M, Qi Z, RosaAnna D, Antonio C, Vinitha Mary K, Jayant R, et al. . Venetoclax combines synergistically with FLT3 inhibition to effectively target leukemic cells in FLT3-ITD+ acute myeloid leukemia models. Haematologica 2020;106:1034–46.
    1. DiNardo CD, BA J, Pullarkat V, Thirman MJ, Garcia JS, Wei AH, et al. . Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med 2020;383:617–29.
    1. Konopleva M, Thirman M, Pratz KW, Letai AG, Recher C, Pullarkat VA, et al. . Results of venetoclax and azacitidine combination in chemotherapy ineligible untreated patients with acute myeloid leukemia with FLT3 mutations. Blood 2020;136:8–10.
    1. DiNardo CD, Pratz K, Pullarkat V, Jonas BA, Arellano M, Becker PS, et al. . Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood 2019;133:7–17.
    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;21:4642–9.
    1. Pratz KW, Jonas BA, Pullarkat V, Recher C, Schuh AC, Thirman MJ, et al. . Measurable residual disease response and prognosis in treatment-naïve acute myeloid leukemia with venetoclax and azacitidine. J Clin Oncol 2021.
    1. U.S. Department of Health and Human Services. 2018 August 14. National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE) v4.03. Available from: .
    1. Chyla B, Daver N, Doyle K, McKeegan E, Huang X, Ruvolo V, et al. . Genetic biomarkers of sensitivity and resistance to venetoclax monotherapy in patients with relapsed acute myeloid leukemia. Am J Hematol 2018;93:E202–5.
    1. Mali RS, Lasater EA, Doyle K, Malla R, Boghaert E, Souers A, et al. . FLT3-ITD activation mediates resistance to the BCL-2 selective antagonist, venetoclax, in FLT3-ITD mutant AML models. Blood 2017;130:1348.
    1. Bose P, Grant S. Mcl-1 as a therapeutic target in acute myelogenous leukemia (AML). Leukemia Research Reports 2013;2:12–4.
    1. Ma J, Zhao S, Qiao X, Knight T, Edwards H, Polin L, et al. . Inhibition of Bcl-2 synergistically enhances the antileukemic activity of midostaurin and gilteritinib in preclinical models of FLT3-mutated acute myeloid leukemia. Clin Cancer Res 2019;25:6815–26.

Source: PubMed

3
購読する