Molecular profile of FLT3-mutated relapsed/refractory patients with AML in the phase 3 ADMIRAL study of gilteritinib

Catherine C Smith, Mark J Levis, Alexander E Perl, Jason E Hill, Matt Rosales, Erkut Bahceci, Catherine C Smith, Mark J Levis, Alexander E Perl, Jason E Hill, Matt Rosales, Erkut Bahceci

Abstract

The phase 3 Study of ASP2215 Versus Salvage Chemotherapy in Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML) With FMS-like Tyrosine Kinase (FLT3) Mutation (ADMIRAL) trial demonstrated the superiority of the FLT3 inhibitor, gilteritinib, to salvage chemotherapy (SC) in patients with FLT3-mutated relapsed or refractory (R/R) AML. Baseline comutations, FLT3-internal tandem duplication (ITD) allelic ratio and length, and treatment-emergent mutations were analyzed in patients in the ADMIRAL trial. Baseline comutations were grouped according to gene subgroups (DNA methylation/hydroxymethylation, transcription, chromatin-spliceosome, receptor tyrosine kinase-Ras signaling, TP53-aneuploidy, NPM1, DNMT3A, DNMT3A/NPM1, WT-1, and IDH1/IDH2). Across all but 1 gene subgroup (TP53-aneuploidy), higher pretransplant response rates and a trend toward longer overall survival were observed with gilteritinib vs SC. Patients with DNMT3A/NPM1 comutations who received gilteritinib had the most favorable outcomes of any molecular subgroup analyzed. Survival outcomes with gilteritinib were not adversely affected by FLT3-ITD allelic ratio, FLT3-ITD length, or multiple FLT3-ITD mutations. Among patients who relapsed on gilteritinib, Ras/mitogen-activated protein kinase (MAPK) pathway and FLT3 F691L gene mutations were the most common mutational events associated with treatment resistance. However, the occurrence of Ras/MAPK pathway gene mutations at baseline did not preclude a clinical benefit from gilteritinib. Acquisition of multiple Ras/MAPK pathway gene mutations at relapse suggests a high level of pathway reactivation is needed to overcome the gilteritinib treatment effect. These findings provide insight into the R/R AML molecular profile and the impact of FLT3 inhibitors on mutational evolution associated with treatment resistance and benefit of gilteritinib across a wide spectrum of molecular and genetic subgroups in FLT3-mutated R/R AML. This trial was registered at www.clinicaltrials.gov as #NCT02421939.

© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Response rates across gene subgroups. (A) Rates of CR/CRh. (B) Rates of CR/CRh in patients eligible for high-intensity chemotherapy. ITT, intention-to-treat.
Figure 2.
Figure 2.
Overall survival by molecular risk and comutated gene categories. (A) Intention-to-treat population. (B) Patients eligible for high-intensity chemotherapy. (C) Dual comutated DNMT3A and NPM1: patients eligible for high-intensity chemotherapy regimens. NE, not estimable.
Figure 3.
Figure 3.
Overall survival by FLT3-ITD length at baseline. (A) Impact of FLT3-ITD length relative to median value: ≤51 bp vs >51 bp. (B) Relative impact of FLT3-ITD length and multiple FLT3-ITD mutations in the gilteritinib arm. (C) Relative impact of FLT3-ITD length and multiple FLT3-ITD mutations in the salvage chemotherapy arm.
Figure 4.
Figure 4.
Mutations detected at baseline and at relapse in patients who relapsed on gilteritinib.
Figure 5.
Figure 5.
Mutation profile of patients who relapsed on gilteritinib therapy. (A) New mutations at relapse. (B) New Ras/MAPK gene mutations at relapse. (C) New FLT3 mutations at relapse. Analyses were restricted to patients who had blood or bone marrow samples available at both baseline and at relapse. †Some patients had mutations in more than 1 Ras/MAPK pathway gene. ‡One patient acquired a FLT3 F691L gatekeeper mutation as well as a JMD point mutation at relapse.

References

    1. Ley TJ, Miller C, Ding L, et al. ; Cancer Genome Atlas Research Network . Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059-2074.
    1. Papaemmanuil E, Gerstung M, Bullinger L, et al. . Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209-2221.
    1. Thiede C, Steudel C, Mohr B, et al. . Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood. 2002;99(12):4326-4335.
    1. Ravandi F, Kantarjian H, Faderl S, et al. . Outcome of patients with FLT3-mutated acute myeloid leukemia in first relapse. Leuk Res. 2010;34(6): 752-756.
    1. Kottaridis PD, Gale RE, Frew ME, et al. . The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98(6):1752-1759.
    1. Wattad M, Weber D, Döhner K, et al. . Impact of salvage regimens on response and overall survival in acute myeloid leukemia with induction failure. Leukemia. 2017;31(6):1306-1313.
    1. Schlenk RF, Kayser S, Bullinger L, et al. ; German-Austrian AML Study Group . Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood. 2014;124(23):3441-3449.
    1. DiNardo CD, Stein EM, de Botton S, et al. . Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med. 2018;378(25):2386-2398.
    1. Perl AE, Martinelli G, Cortes JE, et al. . Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N Engl J Med. 2019;381(18):1728-1740.
    1. Cortes JE, Khaled S, Martinelli G, et al. . Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): a multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2019;20(7):984-997.
    1. Stein EM, DiNardo CD, Pollyea DA, et al. . Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood. 2017;130(6):722-731.
    1. Roboz GJ, Rosenblat T, Arellano M, et al. . International randomized phase III study of elacytarabine versus investigator choice in patients with relapsed/refractory acute myeloid leukemia. J Clin Oncol. 2014;32(18):1919-1926.
    1. Lee LY, Hernandez D, Rajkhowa T, et al. . Preclinical studies of gilteritinib, a next-generation FLT3 inhibitor. Blood. 2017;129(2):257-260.
    1. Mori M, Kaneko N, Ueno Y, et al. . Gilteritinib, a FLT3/AXL inhibitor, shows antileukemic activity in mouse models of FLT3 mutated acute myeloid leukemia. Invest New Drugs. 2017;35(5):556-565.
    1. Perl AE, Altman JK, Cortes J, et al. . Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid leukaemia: a multicentre, first-in-human, open-label, phase 1-2 study. Lancet Oncol. 2017;18(8):1061-1075.
    1. Dhillon S. Gilteritinib: first global approval. Drugs. 2019;79(3):331-339.
    1. Smith CC, Wang Q, Chin CS, et al. . Validation of ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia. Nature. 2012;485(7397):260-263.
    1. Smith CC, Lin K, Stecula A, Sali A, Shah NP. FLT3 D835 mutations confer differential resistance to type II FLT3 inhibitors. Leukemia. 2015;29(12):2390-2392.
    1. Smith CC, Zhang C, Lin KC, et al. . Characterizing and overriding the structural mechanism of the quizartinib-resistant FLT3 “gatekeeper” F691L mutation with PLX3397. Cancer Discov. 2015;5(6):668-679.
    1. Ma HS, Nguyen B, Duffield AS, et al. . FLT3 kinase inhibitor TTT-3002 overcomes both activating and drug resistance mutations in FLT3 in acute myeloid leukemia. Cancer Res. 2014;74(18):5206-5217.
    1. McMahon CM, Ferng T, Canaani J, et al. . Clonal selection with RAS pathway activation mediates secondary clinical resistance to selective FLT3 inhibition in acute myeloid leukemia. Cancer Discov. 2019;9(8):1050-1063.
    1. Zhang H, Savage S, Schultz AR, et al. . Clinical resistance to crenolanib in acute myeloid leukemia due to diverse molecular mechanisms. Nat Commun. 2019;10(1):244.
    1. Kohlmann A, Nadarajah N, Alpermann T, et al. . Monitoring of residual disease by next-generation deep-sequencing of RUNX1 mutations can identify acute myeloid leukemia patients with resistant disease. Leukemia. 2014;28(1):129-137.
    1. Döhner H, Estey E, Grimwade D, et al. . Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424-447.
    1. Mendez LM, Posey RR, Pandolfi PP. The interplay between the genetic and immune landscapes of AML: mechanisms and implications for risk stratification and therapy. Front Oncol. 2019;9:1162.
    1. Loghavi S, Zuo Z, Ravandi F, et al. . Clinical features of de novo acute myeloid leukemia with concurrent DNMT3A, FLT3 and NPM1 mutations. J Hematol Oncol. 2014;7(1):74.
    1. Ley TJ, Ding L, Walter MJ, et al. . DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424-2433.
    1. Döhner K, Schlenk RF, Habdank M, et al. . Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations. Blood. 2005;106(12):3740-3746.
    1. Schnittger S, Schoch C, Kern W, et al. . Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype. Blood. 2005;106(12):3733-3739.
    1. Sakaguchi M, Yamaguchi H, Najima Y, et al. . Prognostic impact of low allelic ratio FLT3-ITD and NPM1 mutation in acute myeloid leukemia. Blood Adv. 2018;2(20):2744-2754.
    1. Döhner K, Thiede C, Jahn N, et al. . Impact of NPM1/FLT3-ITD genotypes defined by the 2017 European LeukemiaNet in patients with acute myeloid leukemia. Blood. 2020;135(5):371-380.
    1. Piloto O, Wright M, Brown P, Kim KT, Levis M, Small D. Prolonged exposure to FLT3 inhibitors leads to resistance via activation of parallel signaling pathways. Blood. 2007;109(4):1643-1652.
    1. Traer E, Martinez J, Javidi-Sharifi N, et al. . FGF2 from marrow microenvironment promotes resistance to FLT3 inhibitors in acute myeloid leukemia. Cancer Res. 2016;76(22):6471-6482.
    1. Smith CC, Paguirigan A, Jeschke GR, et al. . Heterogeneous resistance to quizartinib in acute myeloid leukemia revealed by single-cell analysis. Blood. 2017;130(1):48-58.
    1. Schmalbrock LK, Dolnik A, Cocciardi S, et al. . Clonal evolution of acute myeloid leukemia with FLT3-ITD mutation under treatment with midostaurin. Blood. 2021;137(22):3093-3104.
    1. Croce L, Coperchini F, Magri F, Chiovato L, Rotondi M. The multifaceted anti-cancer effects of BRAF-inhibitors. Oncotarget. 2019;10(61): 6623-6640.
    1. O’Bryan JP. Pharmacological targeting of RAS: recent success with direct inhibitors. Pharmacol Res. 2019;139:503-511.
    1. Levis MJ, Perl AE, Altman JK, et al. . Evaluation of the impact of minimal residual disease, FLT3 allelic ratio, and FLT3 mutation status on overall survival in FLT3 mutation-positive patients with relapsed/refractory (R/R) acute myeloid leukemia (AML) in the Chrysalis phase 1/2 study. Blood. 2017;130(suppl 1):2705.

Source: PubMed

3
Se inscrever