Midostaurin plus intensive chemotherapy for younger and older patients with AML and FLT3 internal tandem duplications

Hartmut Döhner, Daniela Weber, Julia Krzykalla, Walter Fiedler, Gerald Wulf, Helmut Salih, Michael Lübbert, Michael W M Kühn, Thomas Schroeder, Hans Salwender, Katharina Götze, Jörg Westermann, Lars Fransecky, Karin Mayer, Bernd Hertenstein, Mark Ringhoffer, Hans-Joachim Tischler, Sigrid Machherndl-Spandl, Anika Schrade, Peter Paschka, Verena I Gaidzik, Frauke Theis, Felicitas Thol, Michael Heuser, Richard F Schlenk, Lars Bullinger, Maral Saadati, Axel Benner, Richard Larson, Richard Stone, Konstanze Döhner, Arnold Ganser, Hartmut Döhner, Daniela Weber, Julia Krzykalla, Walter Fiedler, Gerald Wulf, Helmut Salih, Michael Lübbert, Michael W M Kühn, Thomas Schroeder, Hans Salwender, Katharina Götze, Jörg Westermann, Lars Fransecky, Karin Mayer, Bernd Hertenstein, Mark Ringhoffer, Hans-Joachim Tischler, Sigrid Machherndl-Spandl, Anika Schrade, Peter Paschka, Verena I Gaidzik, Frauke Theis, Felicitas Thol, Michael Heuser, Richard F Schlenk, Lars Bullinger, Maral Saadati, Axel Benner, Richard Larson, Richard Stone, Konstanze Döhner, Arnold Ganser

Abstract

We conducted a single-arm, phase 2 trial (German-Austrian Acute Myeloid Leukemia Study Group [AMLSG] 16-10) to evaluate midostaurin with intensive chemotherapy followed by allogeneic hematopoietic-cell transplantation (HCT) and a 1-year midosta urin maintenance therapy in adult patients with acute myeloid leukemia (AML) and fms-related tyrosine kinase 3 (FLT3) internal tandem duplication (ITD). Patients 18 to 70 years of age with newly diagnosed FLT3-ITD-positive AML were eligible. Primary and key secondary endpoints were event-free survival (EFS) and overall survival (OS). Results were compared with a historical cohort of 415 patients treated on 5 prior AMLSG trials; statistical analysis was performed using a double-robust adjustment with propensity score weighting and covariate adjustment. Results were also compared with patients (18-59 years) treated on the placebo arm of the Cancer and Leukemia Group B (CALGB) 10603/RATIFY trial. The trial accrued 440 patients (18-60 years, n = 312; 61-70 years, n = 128). In multivariate analysis, EFS was significantly in favor of patients treated within the AMLSG 16-10 trial compared with the AMLSG control (hazard ratio [HR], 0.55; P < .001); both in younger (HR, 0.59; P < .001) and older patients (HR, 0.42; P < .001). Multivariate analysis also showed a significant beneficial effect on OS compared with the AMLSG control (HR, 0.57; P < .001) as well as to the CALGB 10603/RATIFY trial (HR, 0.71; P = .005). The treatment effect of midostaurin remained significant in sensitivity analysis including allogeneic HCT as a time-dependent covariate. Addition of midostaurin to chemotherapy was safe in younger and older patients. In comparison with historical controls, the addition of midostaurin to intensive therapy led to a significant improvement in outcome in younger and older patients with AML and FLT3-ITD. This trial is registered at clinicaltrialsregistry.eu as Eudra-CT number 2011-003168-63 and at clinicaltrials.gov as NCT01477606.

© 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.
CONSORT diagram.
Figure 2.
Figure 2.
Survival distribution for the primary endpoint event-free survival (EFS) and key secondary endpoint overall survival (OS) according to study population and age group. (A) Median EFS times of the 440 patients from the AMLSG 16-10 trial and the 415 patients from the historical control group were 13.6 months and 5.3 months, respectively; and the 2- and 4-year EFS rates 0.41/0.34 and 0.21/0.18, respectively. (B) EFS by cohort age group ≤60 vs >60 years (for median EFS times and EFS rates, see supplemental Table 3). (C) Median OS times of the 440 patients from the AMLSG 16-10 trial and the 415 patients from the historical control group were 36.2 months and 13.2 months, respectively; and the 2- and 4-year OS rates 0.55/0.47 and 0.38/0.31, respectively. (D) OS by age group ≤60 vs >60 years (for median OS times and OS rates, see supplemental Table 4).

References

    1. Döhner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015;373(12):1136-1152.
    1. Bullinger L, Döhner K, Döhner H. Genomics of acute myeloid leukemia diagnosis and pathways. J Clin Oncol. 2017;35(9):934-946.
    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. Nagel G, Weber D, Fromm E, et al. ; German-Austrian AML Study Group (AMLSG) . Epidemiological, genetic, and clinical characterization by age of newly diagnosed acute myeloid leukemia based on an academic population-based registry study (AMLSG BiO). Ann Hematol. 2017;96(12): 1993-2003.
    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. Fröhling S, Schlenk RF, Breitruck J, et al. ; AML Study Group Ulm. Acute myeloid leukemia . Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: a study of the AML Study Group Ulm. Blood. 2002; 100(13):4372-4380.
    1. Gale RE, Green C, Allen C, et al. ; Medical Research Council Adult Leukaemia Working Party . 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(5):2776-2784.
    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. Schlenk RF, Döhner K, Krauter J, et al. ; German-Austrian Acute Myeloid Leukemia Study Group . Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358(18):1909-1918.
    1. Pratcorona M, Brunet S, Nomdedéu J, et al. ; Grupo Cooperativo Para el Estudio y Tratamiento de las Leucemias Agudas Mieloblásticas . Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: relevance to post-remission therapy. Blood. 2013;121(14):2734-2738.
    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. Stone RM, Mandrekar SJ, Sanford BL, et al. . Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377(5):454-464.
    1. Döhner H, Wei AH, Löwenberg B. Towards precision medicine for AML. Nat Rev Clin Oncol. 2021;18(9):577-590.
    1. Schlenk RF, Weber D, Fiedler W, et al. ; German-Austrian AML Study Group . Midostaurin added to chemotherapy and continued single-agent maintenance therapy in acute myeloid leukemia with FLT3-ITD. Blood. 2019;133(8):840-851.
    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. Döhner H, Estey EH, Amadori S, et al. ; European LeukemiaNet . Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453-474.
    1. van Buuren S, Groothuis-Oudshoorn K. Mice: multivariate imputation by chained equations in R. J Stat Softw. 2011;45:1-67.
    1. Bretz F, Posch M, Glimm E, Klinglmueller F, Maurer W, Rohmeyer K. Graphical approaches for multiple comparison procedures using weighted Bonferroni, Simes, or parametric tests. Biom J. 2011;53(6):894-913.
    1. Ho AD, Schetelig J, Bochtler T, et al. ; Study Alliance Leukemia . Allogeneic stem cell transplantation improves survival in patients with acute myeloid leukemia characterized by a high allelic ratio of mutant FLT3-ITD. Biol Blood Marrow Transplant. 2016;22(3):462-469.
    1. Larson RA, Mandrekar SJ, Huebner LJ, et al. . Midostaurin reduces relapse in FLT3-mutant acute myeloid leukemia: the Alliance CALGB 10603/RATIFY trial. Leukemia. 2021;35(9):2539-2551.
    1. Burchert A, Bug G, Fritz LV, et al. . Sorafenib maintenance after allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia with FLT3-internal tandem duplication mutation (SORMAIN). J Clin Oncol. 2020;38(26):2993-3002.
    1. Xuan L, Wang Y, Huang F, et al. . Sorafenib maintenance in patients with FLT3-ITD acute myeloid leukaemia undergoing allogeneic haematopoietic stem-cell transplantation: an open-label, multicentre, randomised phase 3 trial. Lancet Oncol. 2020;21(9):1201-1212.
    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. 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. Thol F, Gabdoulline R, Liebich A, et al. . Measurable residual disease monitoring by NGS before allogeneic hematopoietic cell transplantation in AML. Blood. 2018;132(16):1703-1713.
    1. Blätte TJ, Schmalbrock LK, Skambraks S, et al. . getITD for FLT3-ITD-based MRD monitoring in AML. Leukemia. 2019;33(10):2535-2539.

Source: PubMed

3
Se inscrever