Vorasidenib, a Dual Inhibitor of Mutant IDH1/2, in Recurrent or Progressive Glioma; Results of a First-in-Human Phase I Trial

Ingo K Mellinghoff, Marta Penas-Prado, Katherine B Peters, Howard A Burris 3rd, Elizabeth A Maher, Filip Janku, Gregory M Cote, Macarena I de la Fuente, Jennifer L Clarke, Benjamin M Ellingson, Saewon Chun, Robert J Young, Hua Liu, Sung Choe, Min Lu, Kha Le, Islam Hassan, Lori Steelman, Shuchi S Pandya, Timothy F Cloughesy, Patrick Y Wen, Ingo K Mellinghoff, Marta Penas-Prado, Katherine B Peters, Howard A Burris 3rd, Elizabeth A Maher, Filip Janku, Gregory M Cote, Macarena I de la Fuente, Jennifer L Clarke, Benjamin M Ellingson, Saewon Chun, Robert J Young, Hua Liu, Sung Choe, Min Lu, Kha Le, Islam Hassan, Lori Steelman, Shuchi S Pandya, Timothy F Cloughesy, Patrick Y Wen

Abstract

Purpose: Lower grade gliomas (LGGs) are malignant brain tumors. Current therapy is associated with short- and long-term toxicity. Progression to higher tumor grade is associated with contrast enhancement on MRI. The majority of LGGs harbor mutations in the genes encoding isocitrate dehydrogenase 1 or 2 (IDH1/IDH2). Vorasidenib (AG-881) is a first-in-class, brain-penetrant, dual inhibitor of the mutant IDH1 and mutant IDH2 enzymes.

Patients and methods: We conducted a multicenter, open-label, phase I, dose-escalation study of vorasidenib in 93 patients with mutant IDH1/2 (mIDH1/2) solid tumors, including 52 patients with glioma that had recurred or progressed following standard therapy. Vorasidenib was administered orally, once daily, in 28-day cycles until progression or unacceptable toxicity. Enrollment is complete; this trial is registered with ClinicalTrials.gov, NCT02481154.

Results: Vorasidenib showed a favorable safety profile in the glioma cohort. Dose-limiting toxicities of elevated transaminases occurred at doses ≥100 mg and were reversible. The protocol-defined objective response rate per Response Assessment in Neuro-Oncology criteria for LGG in patients with nonenhancing glioma was 18% (one partial response, three minor responses). The median progression-free survival was 36.8 months [95% confidence interval (CI), 11.2-40.8] for patients with nonenhancing glioma and 3.6 months (95% CI, 1.8-6.5) for patients with enhancing glioma. Exploratory evaluation of tumor volumes in patients with nonenhancing glioma showed sustained tumor shrinkage in multiple patients.

Conclusions: Vorasidenib was well tolerated and showed preliminary antitumor activity in patients with recurrent or progressive nonenhancing mIDH LGG.

©2021 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
Clinical activity and efficacy of vorasidenib in patients with glioma. A, Best response in evaluable patients with measurable disease (25 enhancing and 22 nonenhancing) expressed as the percentage change in SPD of target lesions from the start of treatment. Among the 52 patients, 4 patients with enhancing disease had evaluable but nonmeasurable disease, and 1 withdrew from the study before tumor response evaluations. B, left, Treatment duration and best response for patients with nonenhancing glioma; 8 patients remained on treatment. B, right, Treatment duration and best response for patients with enhancing glioma; 1 patient remained on treatment. In A and B, shaded patient case ID numbers (#) written in bold brown font indicate patients with nonenhancing glioma for whom brain MRI images and volumetric growth curves are shown in Fig. 2. C, One patient with nonenhancing disease had a >50% reduction from baseline that was not confirmed with subsequent scan and is therefore categorized as mR. aLesion growth >100%. bAn mR is defined as a ≥25% but ≤50% decrease in tumor measurements relative to baseline. cA >50% decrease in tumor measurements relative to baseline corresponds to a PR. A >50% reduction from baseline was not confirmed with subsequent scan in 1 patient with nonenhancing disease and was therefore categorized as mR. One patient with enhancing disease had a >50% reduction that was not confirmed and was categorized as SD. PD, progressive disease; SD, stable disease; SPD, sum of products of the diameters.
Figure 2.
Figure 2.
Brain MRI and volume growth curves in 3 patients with nonenhancing glioma treated with vorasidenib. Visual inspection of the images, as well as tumor size and volume measurements, showed tumor shrinkage after vorasidenib treatment. A, Patient #15 is a 47-year-old male with an anaplastic astrocytoma that was initially treated with surgery, radiotherapy, and procarbazine/CCNU/vincristine. Best response as of data cutoff: stable disease. B, Patient #19 is a 40-year-old female with an oligoastrocytoma that was initially treated with surgery and temozolomide. Best response as of data cutoff: mR. C, Patient #22 is a 49-year-old female with an oligodendroglioma that was initially treated with surgery and no other treatment. Best response as of data cutoff: PR. Scan collection time points relative to first dose and corresponding on-treatment cycle numbers are shown.

References

    1. GBD 2016 Brain and Other CNS Cancer Collaborators. Global, regional, and national burden of brain and other CNS cancer, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol 2019;18:376–93.
    1. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. . The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol 2016;131:803–20.
    1. Cancer Genome Atlas Research Network, Brat DJ, Verhaak RGW, Aldape KD, Yung WKA, Salama SR, Cooper LAD, et al.. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med 2015;372:2481–98.
    1. Pallud J, Capelle L, Taillandier L, Fontaine D, Mandonnet E, Guillevin R, et al. . Prognostic significance of imaging contrast enhancement for WHO grade II gliomas. Neuro Oncol 2009;11:176–82.
    1. van den Bent MJ, Smits M, Kros JM, Chang SM. Diffuse infiltrating oligodendroglioma and astrocytoma. J Clin Oncol 2017;35:2394–401.
    1. Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, et al. . EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol 2021;18:170–86.
    1. Claus EB, Walsh KM, Wiencke JK, Molinaro AM, Wiemels JL, Schildkraut JM, et al. . Survival and low-grade glioma: the emergence of genetic information. Neurosurg Focus 2015;38:E6.
    1. Hardee ME, Zagzag D. Mechanisms of glioma-associated neovascularization. Am J Pathol 2012;181:1126–41.
    1. Klein M, Heimans JJ, Aaronson NK, van der Ploeg HM, Grit J, Muller M, et al. . Effect of radiotherapy and other treatment-related factors on mid-term to long-term cognitive sequelae in low-grade gliomas: a comparative study. Lancet 2002;360:1361–8.
    1. McAleer MF, Brown PD. Neurocognitive function following therapy for low-grade gliomas. Semin Radiat Oncol 2015;25:210–8.
    1. Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, et al. . Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 2010;465:966.
    1. Ward PS, Patel J, Wise DR, Abdel-Wahab O, Bennett BD, Coller HA, 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 2010;17:225–34.
    1. Lu C, Ward PS, Kapoor GS, Rohle D, Turcan S, Abdel-Wahab O, et al. . IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 2012;483:474–8.
    1. Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, et al. . Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 2011;19:17–30.
    1. Rohle D, Popovici-Muller J, Palaskas N, Turcan S, Grommes C, Campos C, et al. . An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science 2013;340:626–30.
    1. Wang F, Travins J, DeLaBarre B, Penard-Lacronique V, Schalm S, Hansen E, et al. . Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science 2013;340:622–6.
    1. Saha SK, Parachoniak CA, Ghanta KS, Fitamant J, Ross KN, Najem MS, et al. . Mutant IDH inhibits HNF-4α to block hepatocyte differentiation and promote biliary cancer. Nature 2014;513:110–4.
    1. DiNardo CD, Stein EM, de Botton S, Roboz GJ, Altman JK, Mims AS, et al. . Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med 2018;378:2386–98.
    1. Stein EM, DiNardo CD, Pollyea DA, Fathi AT, Roboz GJ, Altman JK, et al. . Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017;130:722–31.
    1. Mellinghoff IK, Ellingson BM, Touat M, Maher E, De La Fuente MI, Holdhoff M, et al. . Ivosidenib in isocitrate dehydrogenase 1-mutated advanced glioma. J Clin Oncol 2020;38:3398–406.
    1. Konteatis Z, Artin E, Nicolay B, Straley K, Padyana AK, Jin L, et al. . Vorasidenib (AG-881): a first-in-class, brain-penetrant dual inhibitor of mutant IDH1 and 2 for treatment of glioma. ACS Med Chem Lett 2020;11:101–7.
    1. Nicolay B, Narayanaswamy R, Amatangelo MD, Aguado E, Nagaraja R, Murtie J, et al. . EXTH-34. Combined use of the pan-IDH mutant inhibitor AG-881 with radiation therapy shows added benefit in an orthotopic IDH1 mutant glioma model in vivo. Neuro Oncol 2017;19:vi79.
    1. Harding JJ, Lowery MA, Shih AH, Schvartzman JM, Hou S, Famulare C, et al. . Isoform switching as a mechanism of acquired resistance to mutant isocitrate dehydrogenase inhibition. Cancer Discov 2018;8:1540–7.
    1. Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, Galanis E, et al. . Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 2010;28:1963–72.
    1. van den Bent MJ, Wefel JS, Schiff D, Taphoorn MJ, Jaeckle K, Junck L, et al. . Response assessment in neuro-oncology (a report of the RANO group): assessment of outcome in trials of diffuse low-grade gliomas. Lancet Oncol 2011;12:583–93.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. . New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228–47.
    1. Okada H, Weller M, Huang R, Finocchiaro G, Gilbert MR, Wick W, et al. . Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol 2015;16:e534–42.
    1. Neuenschwander B, Branson M, Gsponer T. Critical aspects of the Bayesian approach to phase I cancer trials. Stat Med 2008;27:2420–39.
    1. Mellinghoff I, Cloughesy T, Wen P, Taylor J, Maher E, Arrillaga-Romany I, et al. . ACTR-66. A phase 1, open-label, perioperative study of ivosidenib (AG-120) and vorasidenib (AG-881) in recurrent IDH1 mutant, low-grade glioma: updated results. Neuro Oncol 2019;21:vi28–9.
    1. Baumert BG, Hegi ME, van den Bent MJ, von Deimling A, Gorlia T, Hoang-Xuan K, et al. . Temozolomide chemotherapy versus radiotherapy in high-risk low-grade glioma (EORTC 22033–26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol 2016;17:1521–32.
    1. Houillier C, Wang X, Kaloshi G, Mokhtari K, Guillevin R, Laffaire J, et al. . IDH1 or IDH2 mutations predict longer survival and response to temozolomide in low-grade gliomas. Neurology 2010;75:1560–6.
    1. Jonsson P, Lin AL, Young RJ, DiStefano NM, Hyman DM, Li BT, et al. . Genomic correlates of disease progression and treatment response in prospectively characterized gliomas. Clin Cancer Res 2019;25:5537–47.
    1. Johnson BE, Mazor T, Hong C, Barnes M, Aihara K, McLean CY, et al. . Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 2014;343:189–93.

Source: PubMed

3
Subskrybuj