Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid leukaemia: a multicentre, first-in-human, open-label, phase 1-2 study

Alexander E Perl, Jessica K Altman, Jorge Cortes, Catherine Smith, Mark Litzow, Maria R Baer, David Claxton, Harry P Erba, Stan Gill, Stuart Goldberg, Joseph G Jurcic, Richard A Larson, Chaofeng Liu, Ellen Ritchie, Gary Schiller, Alexander I Spira, Stephen A Strickland, Raoul Tibes, Celalettin Ustun, Eunice S Wang, Robert Stuart, Christoph Röllig, Andreas Neubauer, Giovanni Martinelli, Erkut Bahceci, Mark Levis, Alexander E Perl, Jessica K Altman, Jorge Cortes, Catherine Smith, Mark Litzow, Maria R Baer, David Claxton, Harry P Erba, Stan Gill, Stuart Goldberg, Joseph G Jurcic, Richard A Larson, Chaofeng Liu, Ellen Ritchie, Gary Schiller, Alexander I Spira, Stephen A Strickland, Raoul Tibes, Celalettin Ustun, Eunice S Wang, Robert Stuart, Christoph Röllig, Andreas Neubauer, Giovanni Martinelli, Erkut Bahceci, Mark Levis

Abstract

Background: Internal tandem duplication mutations in FLT3 are common in acute myeloid leukaemia and are associated with rapid relapse and short overall survival. The clinical benefit of FLT3 inhibitors in patients with acute myeloid leukaemia has been limited by rapid generation of resistance mutations, particularly in codon Asp835 (D835). We aimed to assess the highly selective oral FLT3 inhibitor gilteritinib in patients with relapsed or refractory acute myeloid leukaemia.

Methods: In this phase 1-2 trial, we enrolled patients aged 18 years or older with acute myeloid leukaemia who either were refractory to induction therapy or had relapsed after achieving remission with previous treatment. Patients were enrolled into one of seven dose-escalation or dose-expansion cohorts assigned to receive once-daily doses of oral gilteritinib (20 mg, 40 mg, 80 mg, 120 mg, 200 mg, 300 mg, or 450 mg). Cohort expansion was based on safety and tolerability, FLT3 inhibition in correlative assays, and antileukaemic activity. Although the presence of an FLT3 mutation was not an inclusion criterion, we required ten or more patients with locally confirmed FLT3 mutations (FLT3mut+) to be enrolled in expansion cohorts at each dose level. On the basis of emerging findings, we further expanded the 120 mg and 200 mg dose cohorts to include FLT3mut+ patients only. The primary endpoints were the safety, tolerability, and pharmacokinetics of gilteritinib. Safety and tolerability were assessed in the safety analysis set (all patients who received at least one dose of gilteritinib). Responses were assessed in the full analysis set (all patients who received at least one dose of study drug and who had at least one datapoint post-treatment). Pharmacokinetics were assessed in a subset of the safety analysis set for which sufficient data for concentrations of gilteritinib in plasma were available to enable derivation of one or more pharmacokinetic variables. This study is registered with ClinicalTrials.gov, number NCT02014558, and is ongoing.

Findings: Between Oct 15, 2013, and Aug 27, 2015, 252 adults with relapsed or refractory acute myeloid leukaemia received oral gilteritinib once daily in one of seven dose-escalation (n=23) or dose-expansion (n=229) cohorts. Gilteritinib was well tolerated; the maximum tolerated dose was established as 300 mg/day when two of three patients enrolled in the 450 mg dose-escalation cohort had two dose-limiting toxicities (grade 3 diarrhoea and grade 3 elevated aspartate aminotransferase). The most common grade 3-4 adverse events irrespective of relation to treatment were febrile neutropenia (97 [39%] of 252), anaemia (61 [24%]), thrombocytopenia (33 [13%]), sepsis (28 [11%]), and pneumonia (27 [11%]). Commonly reported treatment-related adverse events were diarrhoea (92 [37%] of 252]), anaemia (86 [34%]), fatigue (83 [33%]), elevated aspartate aminotransferase (65 [26%]), and increased alanine aminotransferase (47 [19%]). Serious adverse events occurring in 5% or more of patients were febrile neutropenia (98 [39%] of 252; five related to treatment), progressive disease (43 [17%]), sepsis (36 [14%]; two related to treatment), pneumonia (27 [11%]), acute renal failure (25 [10%]; five related to treatment), pyrexia (21 [8%]; three related to treatment), bacteraemia (14 [6%]; one related to treatment), and respiratory failure (14 [6%]). 95 people died in the safety analysis set, of which seven deaths were judged possibly or probably related to treatment (pulmonary embolism [200 mg/day], respiratory failure [120 mg/day], haemoptysis [80 mg/day], intracranial haemorrhage [20 mg/day], ventricular fibrillation [120 mg/day], septic shock [80 mg/day], and neutropenia [120 mg/day]). An exposure-related increase in inhibition of FLT3 phosphorylation was noted with increasing concentrations in plasma of gilteritinib. In-vivo inhibition of FLT3 phosphorylation occurred at all dose levels. At least 90% of FLT3 phosphorylation inhibition was seen by day 8 in most patients receiving a daily dose of 80 mg or higher. 100 (40%) of 249 patients in the full analysis set achieved a response, with 19 (8%) achieving complete remission, ten (4%) complete remission with incomplete platelet recovery, 46 (18%) complete remission with incomplete haematological recovery, and 25 (10%) partial remission INTERPRETATION: Gilteritinib had a favourable safety profile and showed consistent FLT3 inhibition in patients with relapsed or refractory acute myeloid leukaemia. These findings confirm that FLT3 is a high-value target for treatment of relapsed or refractory acute myeloid leukaemia; based on activity data, gilteritinib at 120 mg/day is being tested in phase 3 trials.

Funding: Astellas Pharma, National Cancer Institute (Leukemia Specialized Program of Research Excellence grant), Associazione Italiana Ricerca sul Cancro.

Copyright © 2017 Elsevier Ltd. All rights reserved.

Figures

Figure 1. Study Design and Accrual
Figure 1. Study Design and Accrual
* Three evaluable subjects ** Enrollment stopped early for low response rate CR, complete remission; CRi, complete remission with incomplete hematologic recovery; CRp, complete remission with incomplete platelet recovery; DLT, dose-limiting toxicity; FLT3, Fms-like tyrosine kinase 3
Figure 2. Gilteritinib Pharmacokinetic Profile and Pharmacodynamic…
Figure 2. Gilteritinib Pharmacokinetic Profile and Pharmacodynamic Effects
A) Gilteritinib plasma concentrations over the 24-hour dosing period following a single dose (Cycle 0 Day −2). B) Gilteritinib plasma concentrations over the 24-hour dosing period following multiple doses (Cycle 1 Day 15). C) Plasma inhibitory activity assay results. Graphs depicting gilteritinib plasma concentrations over the 24-hour dosing period are representative of the following numbers of patients in each dose cohort: 20mg/d, n=4; 40mg/d, n=3; 80mg/d, n=3; 120mg/d, n=3; 200mg/d, n=2; 300mg/d, n=3; 400mg/d, n=1. Plasma samples from patients treated at different dose levels were assayed for inhibitory activity against FLT3-ITD receptors in Molm14 cells using immunoblotting as described in the Methods section. (Left) Each filled circle represents a sample from a single patient at the indicated dose level, collected immediately prior to dosing on Day 15 Cycle 1. For each dose level, the mean plasma inhibitory activity result is indicated by a red line. For each point, the reference sample was collected immediately prior to the first dose on Day 1 Cycle 1. (Right) Representative immunoblots of PIA assays from pre- and post-dose on Days 1, 8, and 15 of Cycle 1 are shown.
Figure 2. Gilteritinib Pharmacokinetic Profile and Pharmacodynamic…
Figure 2. Gilteritinib Pharmacokinetic Profile and Pharmacodynamic Effects
A) Gilteritinib plasma concentrations over the 24-hour dosing period following a single dose (Cycle 0 Day −2). B) Gilteritinib plasma concentrations over the 24-hour dosing period following multiple doses (Cycle 1 Day 15). C) Plasma inhibitory activity assay results. Graphs depicting gilteritinib plasma concentrations over the 24-hour dosing period are representative of the following numbers of patients in each dose cohort: 20mg/d, n=4; 40mg/d, n=3; 80mg/d, n=3; 120mg/d, n=3; 200mg/d, n=2; 300mg/d, n=3; 400mg/d, n=1. Plasma samples from patients treated at different dose levels were assayed for inhibitory activity against FLT3-ITD receptors in Molm14 cells using immunoblotting as described in the Methods section. (Left) Each filled circle represents a sample from a single patient at the indicated dose level, collected immediately prior to dosing on Day 15 Cycle 1. For each dose level, the mean plasma inhibitory activity result is indicated by a red line. For each point, the reference sample was collected immediately prior to the first dose on Day 1 Cycle 1. (Right) Representative immunoblots of PIA assays from pre- and post-dose on Days 1, 8, and 15 of Cycle 1 are shown.
Figure 2. Gilteritinib Pharmacokinetic Profile and Pharmacodynamic…
Figure 2. Gilteritinib Pharmacokinetic Profile and Pharmacodynamic Effects
A) Gilteritinib plasma concentrations over the 24-hour dosing period following a single dose (Cycle 0 Day −2). B) Gilteritinib plasma concentrations over the 24-hour dosing period following multiple doses (Cycle 1 Day 15). C) Plasma inhibitory activity assay results. Graphs depicting gilteritinib plasma concentrations over the 24-hour dosing period are representative of the following numbers of patients in each dose cohort: 20mg/d, n=4; 40mg/d, n=3; 80mg/d, n=3; 120mg/d, n=3; 200mg/d, n=2; 300mg/d, n=3; 400mg/d, n=1. Plasma samples from patients treated at different dose levels were assayed for inhibitory activity against FLT3-ITD receptors in Molm14 cells using immunoblotting as described in the Methods section. (Left) Each filled circle represents a sample from a single patient at the indicated dose level, collected immediately prior to dosing on Day 15 Cycle 1. For each dose level, the mean plasma inhibitory activity result is indicated by a red line. For each point, the reference sample was collected immediately prior to the first dose on Day 1 Cycle 1. (Right) Representative immunoblots of PIA assays from pre- and post-dose on Days 1, 8, and 15 of Cycle 1 are shown.
Figure 3. Clinical Response to Gilteritinib (≥80mg)…
Figure 3. Clinical Response to Gilteritinib (≥80mg) in FLT3-Mutation-Positive Relapsed/Refractory AML Patients
A) Overall clinical response, by dose, in patients with FLT3-mutation-positive R/R AML. CR, complete remission; CRi, complete remission with incomplete hematologic recovery; CRp, complete remission with incomplete platelet recovery; PR, partial remission. Composite response (CRc=CR+CRi+CRp) and overall response (ORR=CRc+PR) are noted in bold type. B) Kaplan–Meier curve showing the overall survival of patients receiving ≤40mg gilteritinib versus ≥80mg gilteritinib.
Figure 3. Clinical Response to Gilteritinib (≥80mg)…
Figure 3. Clinical Response to Gilteritinib (≥80mg) in FLT3-Mutation-Positive Relapsed/Refractory AML Patients
A) Overall clinical response, by dose, in patients with FLT3-mutation-positive R/R AML. CR, complete remission; CRi, complete remission with incomplete hematologic recovery; CRp, complete remission with incomplete platelet recovery; PR, partial remission. Composite response (CRc=CR+CRi+CRp) and overall response (ORR=CRc+PR) are noted in bold type. B) Kaplan–Meier curve showing the overall survival of patients receiving ≤40mg gilteritinib versus ≥80mg gilteritinib.

References

    1. Cancer Genome Atlas Research N. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–74.
    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):4326–35.
    1. Levis M. FLT3 mutations in acute myeloid leukemia: what is the best approach in 2013? Hematology Am Soc Hematol Educ Program. 2013;2013:220–6.
    1. Nakao M, Yokota S, Iwai T, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia. 1996;10(12):1911–8.
    1. Yamamoto Y, Kiyoi H, Nakano Y, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood. 2001;97(8):2434–9.
    1. Smith BD, Levis M, Beran M, et al. Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia. Blood. 2004;103(10):3669–76.
    1. Fischer T, Stone RM, Deangelo DJ, et al. Phase IIB trial of oral Midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and high-risk myelodysplastic syndrome with either wild-type or mutated FLT3. J Clin Oncol. 2010;28(28):4339–45.
    1. Fiedler W, Serve H, Dohner H, et al. A phase 1 study of SU11248 in the treatment of patients with refractory or resistant acute myeloid leukemia (AML) or not amenable to conventional therapy for the disease. Blood. 2005;105(3):986–93.
    1. O’Farrell AM, Yuen HA, Smolich B, et al. Effects of SU5416, a small molecule tyrosine kinase receptor inhibitor, on FLT3 expression and phosphorylation in patients with refractory acute myeloid leukemia. Leuk Res. 2004;28(7):679–89.
    1. Borthakur G, Kantarjian H, Ravandi F, et al. Phase I study of sorafenib in patients with refractory or relapsed acute leukemias. Haematologica. 2011;96(1):62–8.
    1. Zhang W, Konopleva M, Shi YX, et al. Mutant FLT3: a direct target of sorafenib in acute myelogenous leukemia. J Natl Cancer Inst. 2008;100(3):184–98.
    1. Cortes JE, Kantarjian H, Foran JM, et al. Phase I study of quizartinib administered daily to patients with relapsed or refractory acute myeloid leukemia irrespective of FMS-like tyrosine kinase 3-internal tandem duplication status. J Clin Oncol. 2013;31(29):3681–7.
    1. Metzelder S, Wang Y, Wollmer E, et al. Compassionate use of sorafenib in FLT3-ITD-positive acute myeloid leukemia: sustained regression before and after allogeneic stem cell transplantation. Blood. 2009;113(26):6567–71.
    1. Ravandi F, Alattar ML, Grunwald MR, et al. Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation. Blood. 2013;121(23):4655–62.
    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):260–3.
    1. Man CH, Fung TK, Ho C, et al. Sorafenib treatment of FLT3-ITD(+) acute myeloid leukemia: favorable initial outcome and mechanisms of subsequent nonresponsiveness associated with the emergence of a D835 mutation. Blood. 2012;119(22):5133–43.
    1. Alvarado Y, Kantarjian HM, Luthra R, et al. Treatment with FLT3 inhibitor in patients with FLT3-mutated acute myeloid leukemia is associated with development of secondary FLT3-tyrosine kinase domain mutations. Cancer. 2014;120(14):2142–9.
    1. Lee LY, Hernandez D, Rajkhowa T, et al. Pre-clinical studies of gilteritinib, a next-generation FLT3 inhibitor. Blood. 2017;129(2):257–60.
    1. Neubauer A, Fiebeler A, Graham DK, et al. Expression of axl, a transforming receptor tyrosine kinase, in normal and malignant hematopoiesis. Blood. 1994;84(6):1931–41.
    1. Park IK, Mishra A, Chandler J, Whitman SP, Marcucci G, Caligiuri MA. Inhibition of the receptor tyrosine kinase Axl impedes activation of the FLT3 internal tandem duplication in human acute myeloid leukemia: implications for Axl as a potential therapeutic target. Blood. 2013;121(11):2064–73.
    1. Park IK, Mundy-Bosse B, Whitman SP, et al. Receptor tyrosine kinase Axl is required for resistance of leukemic cells to FLT3-targeted therapy in acute myeloid leukemia. Leukemia. 2015;29(12):2382–9.
    1. Mori M, Kaneko N, Ueno Y, et al. ASP2215, a novel FLT3/AXL inhibitor: preclinical evaluation in acute myeloid leukemia (AML) J Clin Oncol. 2014;32(5s suppl) abstract 7070.
    1. Levis M, Brown P, Smith BD, et al. Plasma inhibitory activity (PIA): a pharmacodynamic assay reveals insights into the basis for cytotoxic response to FLT3 inhibitors. Blood. 2006;108(10):3477–83.
    1. Cheson BD, Bennett JM, Kopecky KJ, 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. Yin G, Li Y, Ji Y. Bayesian dose-finding in phase I/II clinical trials using toxicity and efficacy odds ratios. Biometrics. 2006;62(3):777–84.
    1. Levis M, Ravandi F, Wang ES, et al. Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse. Blood. 2011;117(12):3294–301.
    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):1919–26.
    1. Cortes JE, Tallman MS, Schiller G, et al. Results of a phase 2 randomized, open-label, study of lower doses of quizartinib (AC220; ASP2689) in subjects with FLT3-ITD positive relapsed or refractory acute myeloid leukemia (AML) Blood. 2013;122(494)
    1. Reindl C, Bagrintseva K, Vempati S, et al. Point mutations in the juxtamembrane domain of FLT3 define a new class of activating mutations in AML. Blood. 2006;107(9):3700–7.
    1. Zheng R, Levis M, Piloto O, et al. FLT3 ligand causes autocrine signaling in acute myeloid leukemia cells. Blood. 2004;103(1):267–74.
    1. Gallogly MM, Lazarus HM. Midostaurin: an emerging treatment for acute myeloid leukemia patients. J Blood Med. 2016;7:73–83.
    1. Strati P, Kantarjian H, Ravandi F, et al. Phase I/II trial of the combination of midostaurin (PKC412) and 5-azacytidine for patients with acute myeloid leukemia and myelodysplastic syndrome. Am J Hematol. 2015;90(4):276–81.
    1. Knapper S, Russell N, Gilkes A, et al. A randomized assessment of adding the kinase inhibitor lestaurtinib to first-line chemotherapy for FLT3-mutated AML. Blood. 2017;129(9):1143–54.
    1. Röllig C, Serve H, Huttmann A, et al. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial. Lancet Oncol. 2015;16(16):1691–9.
    1. Serve H, Krug U, Wagner R, et al. Sorafenib in combination with intensive chemotherapy in elderly patients with acute myeloid leukemia: results from a randomized, placebo-controlled trial. J Clin Oncol. 2013;31(25):3110–8.
    1. Metzelder SK, Schroeder T, Finck A, et al. High activity of sorafenib in FLT3-ITD-positive acute myeloid leukemia synergizes with allo-immune effects to induce sustained responses. Leukemia. 2012;26(11):2353–9.
    1. Levis MJ, Martinelli G, Perl AE, et al. The benefit of treatment with quizartinib and subsequent bridging to HSCT for FLT3-ITD(+) patients with AML. J Clin Oncol. 2014;32(5s)
    1. Wander SA, Levis MJ, Fathi AT. The evolving role of FLT3 inhibitors in acute myeloid leukemia: quizartinib and beyond. Ther Adv Hematol. 2014;5(3):65–77.
    1. Randhawa J, Kantarjian HM, Borthakur G, et al. Results of a Phase II Study of Crenolanib in Relapsed/Refractory Acute Myeloid Leukemia Patients (Pts) with Activating FLT3 Mutations. Blood. 2014;124(21) abstract 389.
    1. Cortes JE, Kantarjian HM, Kadia TM, et al. Crenolanib besylate, a type I pan-FLT3 inhibitor, to demonstrate clinical activity in multiply relapsed FLT3-ITD and D835 AML. J Clin Oncol. 2016;34(Suppl) Abstract 7008.
    1. Sexauer A, Perl A, Yang X, et al. Terminal myeloid differentiation in vivo is induced by FLT3 inhibition in FLT3/ITD AML. Blood. 2012;120(20):4205–14.
    1. Leischner H, Albers C, Grundler R, et al. SRC is a signaling mediator in FLT3-ITD- but not in FLT3-TKD-positive AML. Blood. 2012;119(17):4026–33.

Source: PubMed

3
Prenumerera