7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      IDH-Mutated AML: Beyond Enasidenib and Ivosidenib Monotherapy: Highlights From SOHO 2021

      meeting-report
      Journal of the Advanced Practitioner in Oncology
      Harborside Press LLC

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Alexis C. Geppner, MLS, CTTS, PA-C, of The University of Texas MD Anderson Cancer Center, distills information reviewed in the session on IDH-mutated acute myeloid leukemia presented by Courtney D. DiNardo, MD, MSCE, also of MD Anderson Cancer Center, at the 2021 SOHO Annual Meeting. Isocitrate dehydrogenase (IDH) driver mutations are detected in 20% of patients with acute myeloid leukemia (AML). Abnormal function of IDH1 and IDH2 genes involves metabolic and epigenetic cellular changes. IDH1 and IDH2 mutations generate an accumulation of 2-hydroxyglutarate (2-HG) leading to DNA hypermethylation, aberrant gene expression, and cell proliferation with impaired cellular differentiation (Montalban-Bravo & DiNardo, 2018). Both IDH1 and IDH2 mutations induce BCL-2 dependence through inhibition of cytochrome c oxidase leading to a lower apoptotic threshold of BCL inhibition (Chan et al., 2015). IDH mutations often (85%) occur in de novo AML, diploid, or trisomy 8 and are more prevalent with increasing age. The frequency of mutations increases from lower to higher-risk disease, suggesting a role in disease progression (Molenaar et al., 2015). Roughly 25% of patients with a myeloproliferative neoplasm (MPN) will acquire an IDH mutation at time of progression to AML. The primary therapeutic approach is intensive cytotoxic chemotherapy and consolidative chemotherapy. Elderly patients and those with relapsed/refractory (R/R) AML are often unable to tolerate intensive chemotherapy and do not have the option to proceed with allogeneic stem cell transplantation. U.S. Food and Drug Administration approvals of enasidenib (Idhifa; Agios Pharmaceuticals, formerly AG-221) and ivosidenib (Tibsovo, formerly AG-120) have allowed for a safe and well-tolerated treatment option that has also been shown to induce hematologic responses in this patient population. ENASIDENIB Enasidenib is an oral selective inhibitor of mutant IDH2 enzymes. A phase I/II study evaluated the response, tolerability, and safety of AG-221 in R/R AML. The overall response rate (ORR) was 40.3% with a median overall survival (OS) of 9.3 months, and a 1-year OS of 39%. In newly diagnosed AML, the median OS was reported as 11.3 months. Even with monotherapy, patients were found to have improved responses and survival when enasidenib was used in earlier stages of treatment. Enasidenib, when used to treat unfit patients with newly diagnosed mutant IDH2 AML, showed 2-HG reductions of 97.8% in the enasidenib plus azacitidine arm compared with 54.3% in the azacitidine alone arm. This combination therapy improved the ORR from 42% to 71%, complete remission (CR) from 12% to 53%, and median event-free survival from 10.4 months to 17.4 months. Enasidenib monotherapy, when given in combination with 5-azacitidine, improves both response and survival. Key Points Notable mechanisms of relapse and patterns of clonal selection (associated with increased 2-HG) when patients begin to lose response to an IDH inhibitor include: Presence of second site mutations of IDH1 and IDH2 leading to restoration of 2-HG, indicating that 2-HG reduction alone is not sufficient (Choe et al., 2020). Primary resistance with co-occurring mutations at baseline. One such is receptor tyrosine kinase (RTK) mutations, which are more associated with mutant IDH1 R/R AML. Secondary resistance to enasidenib after clinical response was associated with emergence of AML-related mutations (RUNX1, GATA2, FLT3). Reconstitution of a differentiation block. Isoform switching: acquired mutant IDH2 in IDH1-mutant clone with elevation of 2-HG at relapse. Enasidenib-induced indirect hyperbilirubinemia occurred in 35% of patients, possibly due to off-target inhibition of the UGT1A1 enzyme responsible for bilirubin metabolism (Stein et al., 2017). Non–dose-dependent, noninfectious leukocytosis occurred in 17% of patients within the first 2 cycles. IVOSIDENIB Ivosidenib is an oral small-molecule inhibitor of mutant IDH1. A phase I study evaluating the safety and tolerance of AG-120 in R/R AML yielded an ORR of 39.1% with a median OS of 8.8 months and 18-month OS of 50.1%. Ivosidenib induced myeloid differentiation without a period of bone marrow aplasia (DiNardo et al., 2018). Common adverse effects included diarrhea (30.7%), leukocytosis (29.6%), febrile neutropenia (28.5%), nausea (27.9%), dyspnea (24.6%), QT prolongation (24.6%), peripheral edema (21.8%), and cough (20.7%; DiNardo et al., 2018). The AGILE double-blind study of azacitidine plus ivosidenib for newly diagnosed AML is ongoing and currently has a CR rate of 36.7% vs. 17.9% in the azacitidine-only arm. VENETOCLAX IDH2 mutations induce BCL-2 dependence by mediating inhibition of cytochrome c oxidase leading to a lower threshold triggering apoptosis with BCL-2 inhibition. Preclinical data confirms synergistic activity of IDH inhibitors plus venetoclax (Venclexta). COMBINATION THERAPY AND FUTURE DIRECTIONS Given the effectiveness of IDH1/2 inhibitors in the relapsed/refractory AML setting, a phase I study was created to evaluate the safety and efficacy of enasidenib and ivosidenib combined with intensive chemotherapy in patients with newly diagnosed AML with mutated IDH1/2 (Stein et al., 2021). A phase 1b trial of ivosidenib plus venetoclax plus azacitidine is currently enrolling patients. Although early, out of six patients initially progressing on hypomethylating agent–based therapy +/- venetoclax who relapsed with an IDH1 mutation, five were able to achieve another remission. This demonstrates the synergy of IDH inhibitors and venetoclax. Newly emerging data reveal continuous response. The Advanced Practitioner Perspective Emerging data from clinical trials evaluating novel agents require advanced practitioners to stay current to improve the quality of care of patients with AML. Elderly and R/R AML patients remain a population with minimal options who can benefit from low-intensity combination chemotherapy. Despite the availability of oral therapy, IDH inhibitors (whether given as monotherapy or in combination) still have the potential for adverse effects. It is imperative that advanced practitioners understand the mechanism of action and educate patients on both expectations and toxicities of therapy. Disclosure: Ms. Geppner has served as a consultant and advisor for AbbVie.

          Related collections

          Most cited references7

          • Record: found
          • Abstract: not found
          • Article: not found

          Durable Remissions with Ivosidenib in IDH1-Mutated Relapsed or Refractory AML

            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia

            Recurrent mutations in isocitrate dehydrogenase 2 (IDH2) occur in ∼12% of patients with acute myeloid leukemia (AML). Mutated IDH2 proteins neomorphically synthesize 2-hydroxyglutarate resulting in DNA and histone hypermethylation, which leads to blocked cellular differentiation. Enasidenib (AG-221/CC-90007) is a first-in-class, oral, selective inhibitor of mutant-IDH2 enzymes. This first-in-human phase 1/2 study assessed the maximum tolerated dose (MTD), pharmacokinetic and pharmacodynamic profiles, safety, and clinical activity of enasidenib in patients with mutant-IDH2 advanced myeloid malignancies. We assessed safety outcomes for all patients and clinical efficacy in the largest patient subgroup, those with relapsed or refractory AML, from the phase 1 dose-escalation and expansion phases of the study. In the dose-escalation phase, an MTD was not reached at doses ranging from 50 to 650 mg per day. Enasidenib 100 mg once daily was selected for the expansion phase on the basis of pharmacokinetic and pharmacodynamic profiles and demonstrated efficacy. Grade 3 to 4 enasidenib-related adverse events included indirect hyperbilirubinemia (12%) and IDH-inhibitor-associated differentiation syndrome (7%). Among patients with relapsed or refractory AML, overall response rate was 40.3%, with a median response duration of 5.8 months. Responses were associated with cellular differentiation and maturation, typically without evidence of aplasia. Median overall survival among relapsed/refractory patients was 9.3 months, and for the 34 patients (19.3%) who attained complete remission, overall survival was 19.7 months. Continuous daily enasidenib treatment was generally well tolerated and induced hematologic responses in patients for whom prior AML therapy had failed. Inducing differentiation of myeloblasts, not cytotoxicity, seems to drive the clinical efficacy of enasidenib. This trial was registered at www.clinicaltrials.gov as #NCT01915498.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in acute myeloid leukemia.

              Mutant isocitrate dehydrogenase (IDH) 1 and 2 proteins alter the epigenetic landscape in acute myeloid leukemia (AML) cells through production of the oncometabolite (R)-2-hydroxyglutarate (2-HG). Here we performed a large-scale RNA interference (RNAi) screen to identify genes that are synthetic lethal to the IDH1(R132H) mutation in AML and identified the anti-apoptotic gene BCL-2. IDH1- and IDH2-mutant primary human AML cells were more sensitive than IDH1/2 wild-type cells to ABT-199, a highly specific BCL-2 inhibitor that is currently in clinical trials for hematologic malignancies, both ex vivo and in xenotransplant models. This sensitization effect was induced by (R)-2-HG-mediated inhibition of the activity of cytochrome c oxidase (COX) in the mitochondrial electron transport chain (ETC); suppression of COX activity lowered the mitochondrial threshold to trigger apoptosis upon BCL-2 inhibition. Our findings indicate that IDH1/2 mutation status may identify patients that are likely to respond to pharmacologic BCL-2 inhibition and form the rational basis for combining agents that disrupt ETC activity with ABT-199 in future clinical studies.
                Bookmark

                Author and article information

                Journal
                J Adv Pract Oncol
                J Adv Pract Oncol
                J Adv Pract Oncol
                JADPRO
                Journal of the Advanced Practitioner in Oncology
                Harborside Press LLC
                2150-0878
                2150-0886
                January 2022
                2 February 2022
                : 13
                : Suppl 1
                : 12-14
                Article
                2022.13.1.11
                10.6004/jadpro.2022.13.1.11
                8824283
                aee48158-511d-4364-86e1-99574f0538a3
                © 2022 Harborside™

                This article is distributed under the terms of the Creative Commons Attribution Non-Commercial Non-Derivative License, which permits unrestricted non-commercial and non-derivative use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                Categories
                Meeting Reports

                Comments

                Comment on this article