Ivosidenib in Isocitrate Dehydrogenase 1 - Mutated Advanced Glioma

Ingo K Mellinghoff, Benjamin M Ellingson, Mehdi Touat, Elizabeth Maher, Macarena I De La Fuente, Matthias Holdhoff, Gregory M Cote, Howard Burris, Filip Janku, Robert J Young, Raymond Huang, Liewen Jiang, Sung Choe, Bin Fan, Katharine Yen, Min Lu, Chris Bowden, Lori Steelman, Shuchi S Pandya, Timothy F Cloughesy, Patrick Y Wen, Ingo K Mellinghoff, Benjamin M Ellingson, Mehdi Touat, Elizabeth Maher, Macarena I De La Fuente, Matthias Holdhoff, Gregory M Cote, Howard Burris, Filip Janku, Robert J Young, Raymond Huang, Liewen Jiang, Sung Choe, Bin Fan, Katharine Yen, Min Lu, Chris Bowden, Lori Steelman, Shuchi S Pandya, Timothy F Cloughesy, Patrick Y Wen

Abstract

Purpose: Diffuse gliomas are malignant brain tumors that include lower-grade gliomas (LGGs) and glioblastomas. Transformation of low-grade glioma into a higher tumor grade is typically associated with contrast enhancement on magnetic resonance imaging. Mutations in the isocitrate dehydrogenase 1 (IDH1) gene occur in most LGGs (> 70%). Ivosidenib is an inhibitor of mutant IDH1 (mIDH1) under evaluation in patients with solid tumors.

Methods: We conducted a multicenter, open-label, phase I, dose escalation and expansion study of ivosidenib in patients with mIDH1 solid tumors. Ivosidenib was administered orally daily in 28-day cycles.

Results: In 66 patients with advanced gliomas, ivosidenib was well tolerated, with no dose-limiting toxicities reported. The maximum tolerated dose was not reached; 500 mg once per day was selected for the expansion cohort. The grade ≥ 3 adverse event rate was 19.7%; 3% (n = 2) were considered treatment related. In patients with nonenhancing glioma (n = 35), the objective response rate was 2.9%, with 1 partial response. Thirty of 35 patients (85.7%) with nonenhancing glioma achieved stable disease compared with 14 of 31 (45.2%) with enhancing glioma. Median progression-free survival was 13.6 months (95% CI, 9.2 to 33.2 months) and 1.4 months (95% CI, 1.0 to 1.9 months) for the nonenhancing and enhancing glioma cohorts, respectively. In an exploratory analysis, ivosidenib reduced the volume and growth rates of nonenhancing tumors.

Conclusion: In patients with mIDH1 advanced glioma, ivosidenib 500 mg once per day was associated with a favorable safety profile, prolonged disease control, and reduced growth of nonenhancing tumors.

Trial registration: ClinicalTrials.gov NCT02073994.

Figures

FIG 1.
FIG 1.
Clinical activity and efficacy of ivosidenib in patients with glioma. (A) Time receiving ivosidenib for the 35 patients with nonenhancing glioma. Twelve patients remain on treatment as of the data cutoff. (B) Time receiving ivosidenib for the 31 patients with enhancing glioma. Three patients remain on treatment as of the data cutoff. (C) Best response in evaluable patients with measurable disease (27 enhancing and 33 nonenhancing), expressed as the percent change in sum of products of the diameters from the target lesions at start of treatment. (D) Investigator-assessed progression-free survival according to glioma type for all evaluable patients with glioma (n = 66). Tick marks indicate censored data. PD, progressive disease; PR, partial response; SD, stable disease. (*) Lesion growth > 100%. (†) Two patients with enhancing disease had decreases of > 50% that were not confirmed and are indicated as SD.
FIG 2.
FIG 2.
(A-D) Examples of brain magnetic resonance images and manually segmented tumor volume growth curves in 4 patients with nonenhancing glioma treated with ivosidenib. IVO, ivosidenib; Tx, treatment with ivosidenib.

References

    1. Mandonnet E, Delattre JY, Tanguy ML, et al. Continuous growth of mean tumor diameter in a subset of grade II gliomas. Ann Neurol. 2003;53:524–528.
    1. van den Bent MJ, Smits M, Kros JM, et al. Diffuse infiltrating oligodendroglioma and astrocytoma. J Clin Oncol. 2017;35:2394–2401.
    1. Weller M, van den Bent M, Tonn JC, et al. European Association for Neuro-Oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas. Lancet Oncol. 2017;18:e315–e329.
    1. Buckner JC, Shaw EG, Pugh SL, et al. Radiation plus procarbazine, CCNU, and vincristine in low-grade glioma. N Engl J Med. 2016;374:1344–1355.
    1. Jonsson P, Lin AL, Young RJ, et al. Genomic correlates of disease progression and treatment response in prospectively characterized gliomas. Clin Cancer Res. 2019;25:5537–5547.
    1. Cairncross JG, Pexman JH, Rathbone MP, et al. Postoperative contrast enhancement in patients with brain tumor. Ann Neurol. 1985;17:570–572.
    1. Pallud J, Taillandier L, Capelle L, et al. Quantitative morphological magnetic resonance imaging follow-up of low-grade glioma: A plea for systematic measurement of growth rates. Neurosurgery. 2012;71:729–739, discussion 739-740.
    1. Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009;360:765–773.
    1. Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321:1807–1812.
    1. Watanabe T, Nobusawa S, Kleihues P, et al. IDH1 mutations are early events in the development of astrocytomas and oligodendrogliomas. Am J Pathol. 2009;174:1149–1153.
    1. van den Bent MJ, Dubbink HJ, Marie Y, et al. IDH1 and IDH2 mutations are prognostic but not predictive for outcome in anaplastic oligodendroglial tumors: A report of the European Organization for Research and Treatment of Cancer Brain Tumor Group. Clin Cancer Res. 2010;16:1597–1604.
    1. Turcan S, Rohle D, Goenka A, et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature. 2012;483:479–483.
    1. Sanson M, Marie Y, Paris S, et al. Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol. 2009;27:4150–4154.
    1. Noushmehr H, Weisenberger DJ, Diefes K, et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell. 2010;17:510–522.
    1. Brat DJ, Verhaak RG, Aldape KD, et al. Comprehensive, integrative genomic analysis of diffuse lower grade gliomas. N Engl J Med. 2015;372:2481–2498.
    1. Dang L, White DW, Gross S, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462:739–744.
    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. 2010;17:225–234.
    1. Chowdhury R, Yeoh KK, Tian YM, et al. The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep. 2011;12:463–469.
    1. Xu W, Yang H, Liu Y, et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell. 2011;19:17–30.
    1. Losman JA, Looper RE, Koivunen P, et al. (R)-2-hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible. Science. 2013;339:1621–1625.
    1. Wang F, Travins J, DeLaBarre B, et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science. 2013;340:622–626.
    1. Rohle D, Popovici-Muller J, Palaskas N, et al. An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science. 2013;340:626–630.
    1. Kosmider O, Gelsi-Boyer V, Slama L, et al. Mutations of IDH1 and IDH2 genes in early and accelerated phases of myelodysplastic syndromes and MDS/myeloproliferative neoplasms. Leukemia. 2010;24:1094–1096.
    1. Mardis ER, Ding L, Dooling DJ, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med. 2009;361:1058–1066.
    1. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood. 2017;130:722–731.
    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. 2018;378:2386–2398.
    1. Lowery MA, Burris HA, III, Janku F, et al. Safety and activity of ivosidenib in patients with IDH1-mutant advanced cholangiocarcinoma: A phase 1 study. Lancet Gastroenterol Hepatol. 2019;4:711–720.
    1. Tap WD, Villalobos VM, Cote GM, et al: Phase 1 study of the mutant IDH1 inhibitor ivosidenib: Safety and clinical activity in patients with advanced chondrosarcoma. J Clin Oncol 38:1693-1701, 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. 2020;38:433–444.
    1. Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 2016;131:803–820.
    1. Louis DN, Ohgaki H, Wiestler OD, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114:97–109.
    1. Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: Response assessment in neuro-oncology working group. J Clin Oncol. 2010;28:1963–1972.
    1. van den Bent MJ, Wefel JS, Schiff D, 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;12:583–593.
    1. Ellingson BM, Bendszus M, Boxerman J, et al. Consensus recommendations for a standardized brain tumor imaging protocol in clinical trials. Neuro-oncol. 2015;17:1188–1198.
    1. Fitzmaurice GM, Laird NM, Ware JH. Applied longitudinal analysis. ed 2. Hoboken, NJ: John Wiley & Sons; 2011.
    1. Frampton GM, Fichtenholtz A, Otto GA, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31:1023–1031.
    1. Reuter M, Gerstner ER, Rapalino O, et al. Impact of MRI head placement on glioma response assessment. J Neurooncol. 2014;118:123–129.
    1. Dubbink HJ, Taal W, van Marion R, et al. IDH1 mutations in low-grade astrocytomas predict survival but not response to temozolomide. Neurology. 2009;73:1792–1795.
    1. Nahed BV, Redjal N, Brat DJ, et al. Management of patients with recurrence of diffuse low grade glioma: A systematic review and evidence-based clinical practice guideline. J Neurooncol. 2015;125:609–630.
    1. Rees J, Watt H, Jäger HR, et al. Volumes and growth rates of untreated adult low-grade gliomas indicate risk of early malignant transformation. Eur J Radiol. 2009;72:54–64.
    1. Freyschlag CF, Krieg SM, Kerschbaumer J, et al. Imaging practice in low-grade gliomas among European specialized centers and proposal for a minimum core of imaging. J Neurooncol. 2018;139:699–711.
    1. Mellinghoff IK, Cloughesy TF, Wen PY, et al: A phase 1, open-label, perioperative study of AG-120 and AG-881 in recurrent IDH1 mutant, low-grade glioma: Results from cohort 1. J Clin Oncol 37(15_suppl abstr):2003, 2019.
    1. Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001;344:1031–1037.
    1. Druker BJ, Sawyers CL, Kantarjian H, et al. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N Engl J Med. 2001;344:1038–1042.
    1. Shirahata M, Ono T, Stichel D, et al. Novel, improved grading system(s) for IDH-mutant astrocytic gliomas. Acta Neuropathol. 2018;136:153–166.

Source: PubMed

3
Sottoscrivi