Clinical profile of gilteritinib in Japanese patients with relapsed/refractory acute myeloid leukemia: An open‐label phase 1 study

Gilteritinib, a novel, highly specific, potent fms‐like tyrosine kinase 3/AXL inhibitor, demonstrated antileukemic activity in patients with relapsed/refractory (R/R) acute myeloid leukemia (AML). In this open‐label phase 1 study (NCT02181660), Japanese patients (aged ≥18 years) with R/R AML received once‐daily gilteritinib, escalating from 20 to 300 mg/d. Primary endpoints were safety/tolerability, including the maximum tolerated dose (MTD) and the recommended dose (RD); secondary endpoints were antileukemic activity and pharmacokinetics (PK). Twenty‐four Japanese patients with R/R AML received once‐daily oral gilteritinib in 1 of 6 dose‐escalation cohorts (20, 40, 80, 120, 200, and 300 mg/d). Gilteritinib was well tolerated. The MTD was 200 mg/d; dose‐limiting toxicities were grade 3 tumor lysis syndrome (120 mg/d; n = 1); and grade 3 elevated blood lactate dehydrogenase, amylase, blood creatine phosphokinase levels, and syncope (all n = 2; 300 mg/d). The RD was 120 mg/d. The most common drug‐related grade ≥3 adverse events were thrombocytopenia (n = 4 [16.7%]) and increased blood creatine phosphokinase (n = 3 [12.5%]). Gilteritinib had a dose‐proportional PK profile. Among patients with mutated fms‐like tyrosine kinase 3, the overall response rate (ORR) was 80% (n = 4 of 5; complete remission [CR] with incomplete platelet recovery, 1 [20%]; CR with incomplete hematologic recovery, 2 [40%]; partial remission (PR), 1 [20%]). Among patients with wild‐type fms‐like tyrosine kinase 3, ORR was 36.4%; (n = 4 of 11; CR, 1 [9.1%]; CR with incomplete platelet recovery, 2 [18.2%]; PR, 1 [9.1%]). In conclusion, gilteritinib was well tolerated and demonstrated antileukemic activity in a Japanese R/R AML population.

[1]  Yu‐quan Wei,et al.  AXL receptor tyrosine kinase as a promising anti-cancer approach: functions, molecular mechanisms and clinical applications , 2019, Molecular Cancer.

[2]  Yongping Song,et al.  Gilteritinib: a novel FLT3 inhibitor for acute myeloid leukemia , 2019, Biomarker Research.

[3]  Sohita Dhillon Gilteritinib: First Global Approval , 2019, Drugs.

[4]  R. Larson,et al.  Selective Inhibition of FLT3 by Gilteritinib in Relapsed/Refractory Acute Myeloid Leukemia: a Multicenter, First-in-human, Open-label, Phase 1/2 Study , 2017, The Lancet. Oncology.

[5]  K. Mori,et al.  Gilteritinib, a FLT3/AXL inhibitor, shows antileukemic activity in mouse models of FLT3 mutated acute myeloid leukemia , 2017, Investigational New Drugs.

[6]  M. Levis,et al.  Preclinical studies of gilteritinib, a next-generation FLT3 inhibitor. , 2017, Blood.

[7]  R. Collins,et al.  Crenolanib besylate, a type I pan-FLT3 inhibitor, to demonstrate clinical activity in multiply relapsed FLT3-ITD and D835 AML. , 2016 .

[8]  H. Einsele,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. , 2015, The Lancet. Oncology.

[9]  M. Caligiuri,et al.  Receptor tyrosine kinase Axl is required for resistance of leukemic cells to FLT3-targeted therapy in acute myeloid leukemia , 2015, Leukemia.

[10]  M. Levis,et al.  Inhibition of c-Kit by tyrosine kinase inhibitors , 2015, Haematologica.

[11]  Z. Estrov,et al.  Results of a Phase II Study of Crenolanib in Relapsed/Refractory Acute Myeloid Leukemia Patients (Pts) with Activating FLT3 Mutations , 2014 .

[12]  M. Konopleva,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 , 2014, Cancer.

[13]  E. Estey,et al.  The Benefit of Treatment with Quizartinib and Subsequent Bridging to HSCT for FLT3-ITD(+) Patients with AML , 2014 .

[14]  M. Levis,et al.  The evolving role of FLT3 inhibitors in acute myeloid leukemia: quizartinib and beyond , 2014, Therapeutic advances in hematology.

[15]  S. Miyano,et al.  Comprehensive analysis of genetic alterations and their prognostic impacts in adult acute myeloid leukemia patients , 2014, Leukemia.

[16]  M. Levis FLT3 mutations in acute myeloid leukemia: what is the best approach in 2013? , 2013, Hematology. American Society of Hematology. Education Program.

[17]  H. Kantarjian,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. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[18]  P. Carmeliet,et al.  Axl, a prognostic and therapeutic target in acute myeloid leukemia mediates paracrine crosstalk of leukemia cells with bone marrow stroma. , 2013, Blood.

[19]  A. Krämer,et al.  Sorafenib in combination with intensive chemotherapy in elderly patients with acute myeloid leukemia: results from a randomized, placebo-controlled trial. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[20]  Benjamin J. Raphael,et al.  Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. , 2013, The New England journal of medicine.

[21]  M. Caligiuri,et al.  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. , 2013, Blood.

[22]  E. Estey,et al.  Final Results of a Phase 2 Open-Label, Monotherapy Efficacy and Safety Study of Quizartinib (AC220) in Patients with FLT3-ITD Positive or Negative Relapsed/Refractory Acute Myeloid Leukemia After Second-Line Chemotherapy or Hematopoietic Stem Cell Transplantation , 2012 .

[23]  K. Götze,et al.  High activity of sorafenib in FLT3-ITD-positive acute myeloid leukemia synergizes with allo-immune effects to induce sustained responses , 2012, Leukemia.

[24]  S. Lok,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. , 2012, Blood.

[25]  Andrew Kasarskis,et al.  Validation of ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia , 2012, Nature.

[26]  E. Estey,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. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[27]  C. Bloomfield,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. , 2003, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[28]  J. Griffin,et al.  The roles of FLT3 in hematopoiesis and leukemia. , 2002, Blood.

[29]  G. Ehninger,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 , 2022 .

[30]  T. Naoe,et al.  Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. , 2001, Blood.

[31]  H. Kaneko,et al.  Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. , 1996, Leukemia.

[32]  J O'Quigley,et al.  Continual reassessment method: a practical design for phase 1 clinical trials in cancer. , 1990, Biometrics.