Mutant Isocitrate Dehydrogenase 1 Inhibitor Ivosidenib in Combination With Azacitidine for Newly Diagnosed Acute Myeloid Leukemia

Courtney D DiNardo, Anthony S Stein, Eytan M Stein, Amir T Fathi, Olga Frankfurt, Andre C Schuh, Hartmut Döhner, Giovanni Martinelli, Prapti A Patel, Emmanuel Raffoux, Peter Tan, Amer M Zeidan, Stéphane de Botton, Hagop M Kantarjian, Richard M Stone, Mark G Frattini, Frederik Lersch, Jing Gong, Diego A Gianolio, Vickie Zhang, Aleksandra Franovic, Bin Fan, Meredith Goldwasser, Scott Daigle, Sung Choe, Bin Wu, Thomas Winkler, Paresh Vyas, Courtney D DiNardo, Anthony S Stein, Eytan M Stein, Amir T Fathi, Olga Frankfurt, Andre C Schuh, Hartmut Döhner, Giovanni Martinelli, Prapti A Patel, Emmanuel Raffoux, Peter Tan, Amer M Zeidan, Stéphane de Botton, Hagop M Kantarjian, Richard M Stone, Mark G Frattini, Frederik Lersch, Jing Gong, Diego A Gianolio, Vickie Zhang, Aleksandra Franovic, Bin Fan, Meredith Goldwasser, Scott Daigle, Sung Choe, Bin Wu, Thomas Winkler, Paresh Vyas

Abstract

Purpose: Ivosidenib is an oral inhibitor of the mutant isocitrate dehydrogenase 1 (IDH1) enzyme, approved for treatment of IDH1-mutant (mIDH1) acute myeloid leukemia (AML). Preclinical work suggested that addition of azacitidine to ivosidenib enhances mIDH1 inhibition-related differentiation and apoptosis.

Patients and methods: This was an open-label, multicenter, phase Ib trial comprising dose-finding and expansion stages to evaluate safety and efficacy of combining oral ivosidenib 500 mg once daily continuously with subcutaneous azacitidine 75 mg/m2 on days 1-7 in 28-day cycles in patients with newly diagnosed mIDH1 AML ineligible for intensive induction chemotherapy (ClinicalTrials.gov identifier: NCT02677922).

Results: Twenty-three patients received ivosidenib plus azacitidine (median age, 76 years; range, 61-88 years). Treatment-related grade ≥ 3 adverse events occurring in > 10% of patients were neutropenia (22%), anemia (13%), thrombocytopenia (13%), and electrocardiogram QT prolongation (13%). Adverse events of special interest included all-grade IDH differentiation syndrome (17%), all-grade electrocardiogram QT prolongation (26%), and grade ≥ 3 leukocytosis (9%). Median treatment duration was 15.1 months (range, 0.3-32.2 months); 10 patients remained on treatment as of February 19, 2019. The overall response rate was 78.3% (18/23 patients; 95% CI, 56.3% to 92.5%), and the complete remission rate was 60.9% (14/23 patients; 95% CI, 38.5% to 80.3%). With median follow-up of 16 months, median duration of response in responders had not been reached. The 12-month survival estimate was 82.0% (95% CI, 58.8% to 92.8%). mIDH1 clearance in bone marrow mononuclear cells by BEAMing (beads, emulsion, amplification, magnetics) digital polymerase chain reaction was seen in 10/14 patients (71.4%) achieving complete remission.

Conclusion: Ivosidenib plus azacitidine was well tolerated, with an expected safety profile consistent with monotherapy with each agent. Responses were deep and durable, with most complete responders achieving mIDH1 mutation clearance.

Figures

FIG 1.
FIG 1.
Treatment duration, response over time, and IDH1 mutation status. Responses were assessed by the investigator. Complete remission (CR) with partial hematologic recovery (CRh) was derived by the sponsor and defined as meeting all CR criteria except absolute neutrophil count > 0.5 × 109/L (500/μL) and platelet count > 50 × 109/L (50,000/μL). The last dose date (for either treatment, whichever was later) was applied for the treatment duration calculation for any patient who was still on treatment as of the data cutoff date. (*) Patient continued on commercially available ivosidenib. (†) Patient had mutant IDH1 (mIDH1) clearance in peripheral blood mononuclear cells (PBMCs) only (bone marrow mononuclear cells [BMMCs] not available); all other patients had mIDH1 clearance in both BMMCs and PBMCs. (‡) Patient received ivosidenib + azacitidine for 23 days before withdrawing from study treatment and died 14 months after end of treatment (unknown cause); no additional therapy after ivosidenib + azacitidine was reported. CRi, CR with incomplete neutrophil recovery; CRp, CR with incomplete platelet recovery; HSCT, hematopoietic stem cell transplantation; MLFS, morphologic leukemia-free state; NA, not assessed; PR, partial remission; SD, stable disease.
FIG 2.
FIG 2.
Baseline co-occurring mutation analysis and association with clinical response. (A) Baseline IDH1 variant allele frequency (VAF) levels as detected by next-generation sequencing (NGS) in either bone marrow mononuclear cells (BMMCs; n = 17) or peripheral blood mononuclear cells (PBMCs; n = 20). VAF in patients achieving complete remission (CR)/CR with partial hematologic recovery (CRh) was compared with that of patients with non-CR/CRh responses (including stable disease [SD]) using Student’s t test (two-sided). VAF levels in neither BMMCs (P = .89) nor PBMCs (P = .17) were associated with clinical response. (B) Mutations co-occurring in ≥ 5% of patients at baseline in order of frequency in BMMC and/or PBMC samples, depending on sample availability. (C) Heat map showing baseline co-occurring mutations identified by variant type and patient characteristics (mutant IDH1 clearance by BEAMing (beads, emulsion, amplification, magnetics) digital polymerase chain reaction, cytogenetic risk, de novo or secondary acute myeloid leukemia) and grouped by best overall response and altered pathway. NGS data were derived from BMMCs (n = 17) or from PBMCs (n = 6) if no screening bone marrow sample was available. IDH1-MC, IDH1 mutation clearance; MLFS, morphologic leukemia-free state; NE, not evaluable; RTK, receptor tyrosine kinase.

References

    1. Dombret H Seymour JF Butrym A, et al. : International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood 126:291-299, 2015
    1. Kantarjian HM Thomas XG Dmoszynska A, et al. : Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol 30:2670-2677, 2012
    1. Ward PS Patel J Wise DR, et al. : The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 17:225-234, 2010
    1. Paschka P Schlenk RF Gaidzik VI, et al. : IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication. J Clin Oncol 28:3636-3643, 2010
    1. DiNardo CD Ravandi F Agresta S, et al. : Characteristics, clinical outcome, and prognostic significance of IDH mutations in AML. Am J Hematol 90:732-736, 2015
    1. Medeiros BC Fathi AT DiNardo CD, et al. : Isocitrate dehydrogenase mutations in myeloid malignancies. Leukemia 31:272-281, 2017
    1. Dang L White DW Gross S, et al. : Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 462:739-744, 2009
    1. Figueroa ME Abdel-Wahab O Lu C, et al. : Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 18:553-567, 2010
    1. Turcan S Rohle D Goenka A, et al. : IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483:479-483, 2012
    1. Chowdhury R Yeoh KK Tian YM, et al. : The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep 12:463-469, 2011
    1. Lu C Ward PS Kapoor GS, et al. : IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 483:474-478, 2012
    1. Xu W Yang H Liu Y, et al. : Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 19:17-30, 2011
    1. Wang F Travins J DeLaBarre B, et al. : Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science 340:622-626, 2013
    1. Popovici-Muller J Lemieux RM Artin E, et al. : Discovery of AG-120 (ivosidenib): A first-in-class mutant IDH1 inhibitor for the treatment of IDH1 mutant cancers. ACS Med Chem Lett 9:300-305, 2018
    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 378:2386-2398, 2018
    1. Roboz GJ DiNardo CD Stein EM, et al. : Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia. Blood 135:463-471, 2020
    1. Yen K, Chopra VS, Tobin E, et al: Functional characterization of the ivosidenib (AG-120) and azacitidine combination in a mutant IDH1 AML cell model. Cancer Res 78, 2018 (suppl; abstr 4956)
    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 21:4642-4649, 2003
    1. Fan B Dai D DiNardo CD, et al. : Clinical pharmacokinetics and pharmacodynamics of ivosidenib in patients with advanced hematologic malignancies with an IDH1 mutation. Cancer Chemother Pharmacol 85:959-968, 2020
    1. Fan B Mellinghoff IK Wen PY, et al. : Clinical pharmacokinetics and pharmacodynamics of ivosidenib, an oral, targeted inhibitor of mutant IDH1, in patients with advanced solid tumors. Invest New Drugs 38:433-444, 2020
    1. Papaemmanuil E Gerstung M Bullinger L, et al. : Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med 374:2209-2221, 2016
    1. Craddock CF Houlton AE Quek LS, et al. : Outcome of azacitidine therapy in acute myeloid leukemia is not improved by concurrent vorinostat therapy but is predicted by a diagnostic molecular signature. Clin Cancer Res 23:6430-6440, 2017
    1. Heidel F Solem FK Breitenbuecher F, et al. : Clinical resistance to the kinase inhibitor PKC412 in acute myeloid leukemia by mutation of Asn-676 in the FLT3 tyrosine kinase domain. Blood 107:293-300, 2006
    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 9:1050-1063, 2019
    1. Smith CC Paguirigan A Jeschke GR, et al. : Heterogeneous resistance to quizartinib in acute myeloid leukemia revealed by single-cell analysis. Blood 130:48-58, 2017
    1. DiNardo CD Tiong IS Quaglieri A, et al. : Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood 135:791-803, 2020
    1. DiNardo CD Pratz K Pullarkat V, et al. : Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood 133:7-17, 2019
    1. AbbVie: VENCLEXTA (venetoclax tablets) for oral use (prescribing information), 2019. .

Source: PubMed

3
S'abonner