touchONCOLOGY touchONCOLOGY
Neuro-oncology
Read Time: 10 mins

Targeting Mutant IDH to Treat Low-grade Glioma

Copy Link
Published Online: Nov 21st 2023 touchREVIEWS in Oncology & Haematology. 2023;19(2):7-11 DOI: https://doi.org/10.17925/OHR.2023.19.2.3
Authors: Katherine B Peters
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

Adult-type diffuse low-grade gliomas (LGGs) develop in young adults and represent 5–10% of all primary brain tumours. While patients with LGG can survive longer than those with higher-grade tumours, progression, transformation and, ultimately, mortality occur. Median overall survival for patients with LGGs with multimodal treatment is roughly 13 years from time of diagnosis. Treatment regimens include surgery, radiation therapy and chemotherapy and are based not only on older clinical trials specific to LGG, but also on observations from larger trials in more prevalent high-grade gliomas, such as glioblastoma. LGGs are genetically distinct from glioblastoma, as they harbour mutations in isocitrate dehydrogenase, a key enzyme in the Krebs cycle, and this mutation leads to the production of 2-hydroxyglutarate, the critical driver of gliomagenesis. This article summarizes LGG treatment and how the recent results from the pivotal INDIGO (ClinicalTrials.gov identifier: NCT04164901) study involving vorasidenib, an oral brain-penetrant IDH mutant inhibitor, will change future treatment regimens and outcomes for patients with IDH-mutant LGG.

Keywords

Gliomaisocitrate dehydrogenaselow-grademutanttargeted therapyvorasidenib

Article:

Adult-type diffuse low-glade gliomas (LGGs) (World Health Organization [WHO] grade 2) represent 5–10% of all primary brain tumours.1 The median age of onset is in the third and fourth decades of life. Common clinical presentations include seizures, focal neurological deficits, and new-onset headaches.2 Magnetic resonance imaging (MRI) demonstrates T1 signal hypointensity and T2/fluid attenuated inversion recovery (FLAIR) hyperintensity with occasional enhancement. They tend to develop in the frontal or temporal lobes; however, all lobes of the brain can harbor these tumours.2 Traditionally, LGG pathology was described in terms of histological appearance. With the updates to WHO 2021 classification for central nervous system tumours, LGG pathology is defined not only by the histology, but also by the molecular/genetic makeup of the tumour.3 The most important genetic aberration that defines adult-type diffuse LGG is mutations in the isocitrate dehydrogenase (IDH) gene.4 IDH has two isoforms, IDH1 in the cytoplasm and IDH2 in the mitochondria, with the most common mutation being the IDH1 mutation in the 132 codon from arginine to histidine (Figure 1).5,6 The normal IDH enzyme is part of the Krebs cycle and converts isocitrate to α-ketoglutarate. When the IDH1/2 mutant enzyme is present, α-ketoglutarate is converted to 2-hydroxyglutarate, the oncometabolite responsible for gliomagenesis.7 The observation is that all glial tumours with IDH mutations have an improved prognosis compared with those with IDH wildtype genotype, and this was the key motivation to make these specific categories in WHO 2021 classification system. Adult-type LGGs WHO grade 2 are currently described as oligodendroglioma, IDH-mutant, and 1p/19q-deleted and astrocytoma, IDH-mutant. 1p/19q co-deletion involves complete deletion of the short arm of chromosome 1 (1p) and the long arm of chromosome 19 (19q) and is specific to oligodendrogliomas (Table 1). Moreover, it is a predictive and prognostic biomarker in patients with oligodendroglioma as patients respond better to combinatorial chemotherapy and have a better prognosis.8,9

Figure 1: Mutant IDH lead to conversion of α-ketoglutarate to production of 2-HG, a oncometabolite, causing gliomagenesis

CoA = coenzyme A; IDH1 = isocitrate dehydrogenase 1; IDH2 = isocitrate dehydrogenase 2; 2-HG = 2-hydroxyglutarate. Created with BioRender.com.

Table 1: Criteria for low-grade glioma diagnosis in adults (per World Health Organization 2021 guidelines3)

IDH1 mutation or IDH2 mutation

IDH1 mutation or IDH2 mutation

Associated mutations

ATRX retained, 1 p/19q co-deletion

ATRX mutant, TP53 mutation, 1 p/19q intact

Integrated WHO 2021 diagnosis

Oligodendriglioma, IDH-mutant, 1 p/19q co-deleted, WHO grade 2

Astrocytoma, IDH-mutant, WHO grade 2

ATRX = alpha-thalassemia/mental retardation syndrome X-linked; IDH1 = isocitrate dehydrogenase 1; IDH2 = isocitrate dehydrogenase 2; IDH = isocitrate dehydrogenase; WHO = World Health Organization .

Table 2: Active and future clinical trials utilizing IDH-mutant inhibitors

Trial name (ClinicalTrial.gov identifier)

Compound (route)

Mechanism

Study population

Phase

Estimated completion date

A Study of HMPL-306 in Advanced Solid Tumors With IDH Mutations (NCT04762602)27

HMPL-306 (oral)

Dual IDH1/2-mutant inhibitor

Advanced solid tumours

Phase I

30 September 2023

A Study of DS-1001b in Patients With Chemotherapy- and Radiotherapy-Naive IDH1 Mutated WHO Grade II Glioma (NCT04458272)28,29

DS-1001b (oral)

IDH1-mutant inhibitor

Grade 2 glioma

Phase II

30 June 2026

ViCToRy: Vorasidenib in Combination With Tumor Specific Peptide Vaccine for Recurrent IDH1 Mutant Lower Grade Gliomas (ViCToRy) (NCT05609994)30

Vorasidenib (oral) and peptide vaccine (SC)

IDH1/2-mutant inhibitor and peptide vaccine to R132H IDH1 mutation

Lower grade glioma

Phase I

August 2027

Study of Vorasidenib and Pembrolizumab Combination in Recurrent or Progressive Enhancing IDH-1 Mutant Astrocytomas (NCT05484622)31

Vorasidenib (oral) and pembrolizumab (IV)

IDH1/2-mutant inhibitor and PD-1 inhibitor

Recurrent mutant astrocytoma

Phase I

30 August 2027

Ivosidenib (AG-120) With Nivolumab in IDH1 Mutant Tumors (NCT04056910)32

Ivosidenib (oral) and nivolumab (IV)

IDH1-mutant inhibitor and PD-1 inhibitor

Gliomas and advanced solid tumours

Phase II

31 May 2026

Safusidenib Phase 2 Study in IDH1 Mutant Glioma (NCT05303519)33

Safusidenib (oral)

IDH1-mutant inhibitor

Recurrent glioma

Phase II

31 July 2027

IDH1 = isocitrate dehydrogenase 1; IDH2 = isocitrate dehydrogenase 2; IV = intravenous; PD = programmed cell death protein 1; SC = subcutaneous; WHO = World Health Organization .

Treatment of adult-type diffuse LGG before IDH mutation inhibition

Treatment of IDH-mutant adult-type diffuse LGG requires a multimodal approach with contributions from neurosurgery, neuropathology, radiation oncology, neuro-oncology, neurology and medical oncology. The first step in treatment is to determine the proper diagnosis. Classically, this determination was made by histology obtained by biopsy or craniotomy, but now providers are using molecular/genetic testing to determine the correct diagnosis. Providers can obtain this testing with in-house efforts from neuropathology or next-generation sequencing platforms. While variability exists in histopathology laboratories based on proprietary practices, in general, when a tumour is determined to be glial in origin, commercial antibody testing is performed for mutant IDH1 specific for the R132H mutation.10 If this test is negative, then it prompts more in-depth testing using next-generation sequencing. It is key to note that providers need to be aware that non-canonical mutations in IDH1 occur, along with IDH2 mutations. In special cases, DNA tumour methylation analysis is utilized, but it is not widely available.11

Greater extent of surgical resection is associated with improved prognosis in a systemic meta-analysis and is more attainable given the improved technology using functional mapping and awake craniotomy techniques.12,13 Moreover, uncontrolled studies point out that early initial resection is associated with improved prognosis over initial biopsy only.14

After the best possible safe surgery, recommendations for IDH-mutant adult-type diffuse LGG can include observation, radiation therapy with concurrent and adjuvant chemotherapy and consideration of clinical trials. This strategy is highlighted in the guidelines from the National Comprehensive Cancer Network (NCCN), European Association of Neuro-Oncology (EANO) and the Korean Society of Neuro-Oncology (KSNO).15 Consideration of observation after surgery with delay of radiation therapy is supported by the critical EORTC 22845 randomized clinical trial (Early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults) that randomized patients with LGG to either early radiation therapy or delayed radiation therapy.16 In this study of 314 patients with LGG randomized 1:1 to either early radiation therapy or delayed radiation therapy, results demonstrated that early radiation therapy improved progression-free survival, but there was a similar median overall survival for both groups (7.4 years for early radiation therapy versus 7.2 years for delayed radiation therapy, hazard ratio [HR] 0.97).

At the time of disease progression, patients are generally recommended to have radiation therapy with concurrent and adjuvant chemotherapy. In both the NCCN and KSNO guidelines, they stratify patients by high- and low-risk, in terms of recommendations for initial treatment after surgery.15 Factors associated with a higher risk of tumor progression include age >40 years and surgery less than gross total resection. The RTOG 9802 trial (Observation or radiation therapy with or without combination chemotherapy in treating patients with low-grade glioma; ClinicalTrials.gov identifier: NCT00003375) delineated the role of chemotherapy for patients with high-risk LGG, showing that the addition of combinatorial chemotherapy with procarbazine, lomustine and vincristine (PCV) to radiation therapy improved progression-free and overall survival compared with radiation therapy alone.17 In RTOG 9802, 251 patients with high-risk LGG were randomized to receive either radiation therapy alone or radiation therapy with PCV. Median progression-free survival was improved significantly in the combination group (RT + PCV) at 10.4 years when compared with the RT alone group at 4.0 years (HR=0.50, p<0.001). Median overall survival was superior in the combination group (RT + PCV) at 13.3 years compared with the RT alone group at 7.8 years (HR=0.59, p=0.003). Tolerability of PCV is challenging, as it can lead to cytopenias, fatigue and other toxicities, so temozolomide (TMZ), an oral alkylating agent US Food and Drug Administration (FDA)-approved for newly diagnosed glioblastoma IDH wildtype WHO grade 4, is commonly prescribed instead by providers.18

Many assumptions are made about how to approach the treatment of LGG, and these arise from clinical trial outcomes from newly diagnosed high-grade glioma studies. If avoiding toxicity is a goal in treating LGG, then delaying or eliminating the need for radiation therapy should be considered. Radiation therapy can cause significant cognitive dysfunction and lead to short- and long-term deficits.19 Douw and colleagues evaluated long-term LGG survivors who received radiation therapy and demonstrated a progressive decline in cognitive abilities.20 Additionally, these cognitive deficits were correlated with radiographic findings of white matter damage and cortical atrophy20. Better therapeutics for IDH-mutant adult-type diffuse LGG are needed to avoid the toxicities seen with conventional chemotherapy and radiation therapy. All in all, paradigms to treat IDH-mutant adult-type diffuse LGG deserve to be tailored to the specifics of the disease itself and should not extrapolated from treatment for high-grade gliomas such as glioblastoma IDH wildtype.

INDIGO trial: Vorasidenib in IDH-mutant LGG

An approach to treat LGG by targeting the tumour-specific mutation in the IDH1 and IDH2 genes has shown promise, based on recent results from a study by Mellinghoff et al.21 Vorasidenib is an oral, highly brain-penetrant inhibitor of mutant IDH1 and IDH2. In a phase I study of vorasidenib in recurrent and progressive glioma, vorasidenib was well-tolerated and safe, and median progression-free survival in patients with non-enhancing glioma was 36.8 months (95% confidence interval [CI], 11.2–40.8 months). Based on this study and a perioperative clinical trial that included vorasidenib, the INDIGO study (Study of vorasidenib [AG-881] in participants with residual or recurrent grade 2 glioma with an IDH1 or IDH2 mutation; ClinicalTrials.gov identifier: NCT04164901) was developed, and the dose of 40 mg orally once a day was determined to be the recommended phase III dose.22,23 In this pivotal phase III study, 331 patients with IDH-mutant LGG were enrolled and randomized in a blinded fashion to receive either placebo or vorasidenib. Key inclusion criteria included that prior treatment was limited to surgery only, and surgery should have occurred 1–5 years ago, and there had to be measurable non-enhancing disease of >1 cm and <1 cm enhancing disease. The primary endpoint was progression-free survival, and the key secondary endpoint was time-to-next intervention. The desired goal was to extend both of these endpoints particularly to delay the need for radiation therapy, further surgery, or initiation of chemotherapy. Patients receiving vorasidenib exhibited significantly longer progression-free survival than patients receiving placebo (27.7 months versus 11.1 months, HR 0.39, p<0.001). Time-to-next intervention was extended in the vorasidenib arm compared with the placebo arm in a highly significant manner (not reached at the time of analysis versus 17.7 months, HR 0.26, p<0.001). Given the success of these results, the study was concluded after an interim analysis, and patients receiving placebo were allowed to cross over to the vorasidenib arm. Vorasidenib was highly tolerated in this patient population, with only 3% of patients discontinued during the study. There was a higher incidence of elevated liver function tests in the vorasidenib arm, so care should be taken to monitor liver function tests appropriately and regularly. During the INDIGO clinical trial, liver function tests were monitored on day 1 and day 15 of the first two cycles and then on day 1 for all subsequent cycles (monthly) as long as the liver function tests remained within normal limits. With these results, vorasidenib is primed to become a critical therapeutic in treating newly diagnosed IDH-mutant LGG.

The INDIGO study enrolled patients from North America, Europe and Israel. A majority of patients were from North America (n=193). When comparing patients from North America, Europe, and Israel, there appears to be some suggestion of increased benefit in patients with IDH-mutant LGG in North America (HR=0.34) compared with patients in Europe (HR=0.54) and Israel (HR=0.45). Given the smaller number of patients enrolled in Europe and Israel, it is challenging to draw more significant conclusions. Still, these observations provide a rationale for exploring outcomes of vorasidenib in other populations on different continents, including South America, Asia, and Africa.

Before vorasidenib, ivosidenib (AG-120, Tibsovo®, Servier Pharmaceuticals, Paris, France), which is FDA-approved for both newly diagnosed and recurrent/refractory acute myelogenous leukemia, was the first-in-class oral small-molecule inhibitor of mutant IDH enzyme that blocks the conversion of α-ketoglutarate to 2-hydroxyglutarate.24,25 In a phase I dose-escalation study, ivosidenib had clinical activity in patients with recurrent IDH1-mutant non-enhancing glioma with a median progression-free survival of 13.6 months (95% CI, 9.2–22.2 months), and they were able to achieve stable disease in 35 patients (85.7%).26 When and how ivosidenib will be used in patients with brain tumours is limited by its poorer brain penetrance compared with vorasidenib, which can more easily cross the blood–brain barrier. One cannot discount the apparent usefulness of ivosidenib given that it appears to have similar efficacy in tumour response and reduction of 2-hydroxyglutarate as determined in the perioperative study comparing vorasidenib and ivosidenib in patients with recurrent IDH1-mutant LGG.23 Where ivosidenib fits into the future treatment regimens for IDH1-mutant glioma remains to be determined.

Next steps for inhibitors of IDH mutations

The results from the INDIGO study in newly diagnosed IDH-mutant LGG will undoubtedly change how patients with these tumours are treated. Vorasidenib is on track for FDA approval for this patient population. Moreover, this study highlights the recommendations from the WHO 2021 classification system for central nervous system tumours; obtaining molecular/genetic information for glial tumours is critical, and genotype status for IDH1 and IDH2 is paramount for prognostic and driving treatment decisions.

Three questions stand out from this randomized clinical trial. First, what is the optimal timing of these agents? For this study, patients were naïve to prior chemotherapy and radiation therapy, and only had prior surgery. Additionally, patients had to have measurable non-enhancing disease, but <1 cm of enhancing disease. Extrapolations from prior clinical trials, such as EORTC 22845 and RTOG 9802, that did not have the technology available for IDH genotype testing, are currently challenging to make for IDH-mutant adult-type diffuse glioma. One implication is that the extent and timing of surgical resection might not hold the same prognostic value in patients with IDH-mutant adult-type diffuse glioma. Time will tell how we can best use IDH-mutant inhibitors. One can posit that future clinical studies in newly diagnosed IDH-mutant LGG could include exploring the role of IDH-mutant inhibition at the time of diagnosis, perhaps even before definitive surgical resection.

Regarding the optimal timing of when to start IDH-mutant inhibitors, one can imagine that initiation of therapy could be different for patients depending on histological type (oligodendroglioma versus astrocytoma), location (non-eloquent versus eloquent), and extent of surgical resection (gross total resection versus subtotal resection versus biopsy). One can posit that healthcare providers of patients with more favorable factors, such as oligodendroglioma histology, location in non-eloquent regions and greater extent of surgery, might consider waiting before initiating therapy. On the other hand, providers with patients with less favorable factors, such as astrocytoma histology, location in eloquent regions, and suboptimal surgical resection, might favor the initiation of IDH-mutant inhibitors. Other factors to consider in initiating IDH-mutant inhibitors might include the need to preserve fertility and mitigate side effects, despite the favourable safety profile of vorasidenib.

Next, can we translate these results from INDIGO to our newly diagnosed patients with high-grade glioma with IDH mutations? Similar to how researchers and providers took the observations from WHO grade 3–4 glioma studies to guide therapeutic regimens in IDH-mutant LGG, vorasidenib will likely be utilized in the research and treatment of IDH-mutant high-grade gliomas. Upcoming and active studies are looking at low-grade and high-grade glioma populations (Table 2). Future trials will need to explore the role of IDH-mutant inhibitors during radiation therapy or during adjuvant therapy for higher-grade IDH-mutant tumours. Future endeavours will only add to the evidence and provide more options for all patients with IDH-mutant glioma.

Finally, how does vorasidenib impact the quality-of-life outcomes in patients with IDH-mutant glioma? INDIGO explored the quality of life, cognition and seizure outcomes in these patients, and the results will be published in the future. Patients with IDH-mutant adult-type diffuse LGG experience significant symptoms and impairments in their quality of life.34 Common issues these patients face include seizures, fatigue, cognitive dysfunction, headaches, sleep disturbances and mood disorders.35 Interprofessional, multidisciplinary teams from neuro-oncology, neurology, neurosurgery, neuropsychology, psychiatry, radiation oncology, physical therapy, occupational therapy and other subspecialties are vital to supporting these patients through their disease trajectory. In particular, seizures are more common in patients with IDH-mutant glioma. Work from Chen and colleagues showed that preoperative seizures were more common in patients with IDH-mutant glioma (59–74%) compared with patients with IDH-wildtype glioma (18–34%).36 While this association could be secondary to the location in epileptically prone areas (frontal and temporal lobes), they found that exogenous 2-hydroxyglutarate promoted excitatory activities in cultured neurons. The presence of 2-hydroxyglutarate in IDH-mutant tumours stimulates local neurons towards stimulatory activity that leads to seizures. Researchers will have an opportunity with the data from INDIGO to explore these particular issues in this patient population. Meaningful design of the relevant patient-reported outcomes and integration into future clinical trials are warranted.

This randomized clinical trial represents a practice-changing step in treating IDH-mutant diffuse adult-type LGG by utilizing a targeted approach to IDH mutations. Active and upcoming clinical studies utilizing IDH-mutant inhibitors are ongoing and are likely to establish when and how providers will use IDH-mutant inhibitors in treating all grades of glioma that have an IDH mutation (Table 2).27–33

Article Information:
Disclosure

Katherine B Peters has served on advisory boards for ONO Pharmaceuticals, Servier, Sapience and Vivacitas Oncology and received research funding from Biomimetix, Servier, Sapience and Varian.

Compliance With Ethics

This article involves a review of the literature and does not report new clinical data, and does not involve any studies with human or animal subjects performed by any of the authors.

Review Process

Double-blind peer review.

Authorship

The named author meets the criteria of the International Committee of Medical Journal Editors for authorship for this manuscript, takes responsibility for the integrity of the work as a whole and has given final approval for the version to be published.

Correspondence

Katherine B PetersDepartments of Neurosurgery and Neurology, Preston Tisch Brain Tumour Center,Duke University School of Medicine, DUMC 3624DurhamNCUSAkatherine.peters@duke.edu

Support

No funding was received in the publication of this article.

Access

This article is freely accessible at touchONCOLOGY.com © Touch Medical Media 2023.

Data Availability

Data sharing is not applicable to this article as no datasets were generated or analyzed during the writing of this article.

Received

2023-07-24

References

1. Ostrom QTPrice MNeff Cet alCBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2015-2019Neuro Oncol2022;24(Suppl.5):v195. DOI: 10.1093/neuonc/noac202.

2. Schiff DLow-grade gliomasContinuum (Minneap Minn). 2017;23:156479. DOI: 10.1212/CON.0000000000000537.

3. Louis DNPerry AWesseling Pet alThe 2021 WHO classification of tumors of the central nervous system: A summaryNeuro Oncol. 2021;23:123151. DOI: 10.1093/neuonc/noab106.

4. Yan HParsons DWJin Get alIDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009;360:76573. DOI: 10.1056/NEJMoa0808710.

5. Capper DWeissert SBalss Jet alCharacterization of R132H mutation-specific IDH1 antibody binding in brain tumorsBrain Pathol2010;20:24554DOI10.1111/j.1750-3639.2009.00352.x.

6. Reitman ZJYan HIsocitrate dehydrogenase 1 and 2 mutations in cancer: Alterations at a crossroads of cellular metabolismJ Natl Cancer Inst. 2010;102:93241. DOI10.1093/jnci/djq187.

7. Chou FJLiu YLang FYang C. D-2-hydroxyglutarate in glioma biology. Cells. 2021;10:2345. DOI: 10.3390/cells10092345.

8. Jenkins RBBlair HBallman KVet alA t(1;19)(q10;p10) mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Cancer Res. 2006;66:985261. DOI: 10.1158/0008-5472.CAN-06-1796.

9. Zhao JMa WZhao HLoss of heterozygosity 1p/19q and survival in glioma: A meta-analysis. Neuro Oncol. 2014;16:10312. DOI: 10.1093/neuonc/not145.

10. Capper DReuss DSchittenhelm Jet alMutation-specific IDH1 antibody differentiates oligodendrogliomas and oligoastrocytomas from other brain tumors with oligodendroglioma-like morphology. Acta Neuropathol. 2011;121:24152. DOI: 10.1007/s00401-010-0770-2.

11. Capper DJones DTWSill Met alDNA methylation-based classification of central nervous system tumours. Nature. 2018;555:46974. DOI10.1038/nature26000.

12. Brown TJBrennan MCLi Met alAssociation of the extent of resection with survival in glioblastoma: A systematic review and meta-analysis. JAMA Oncol. 2016;2:14609. DOI: 10.1001/jamaoncol.2016.1373.

13. Hervey-Jumper SLBerger MSMaximizing safe resection of low- and high-grade glioma. J Neurooncol. 2016;130:26982. DOI: 10.1007/s11060-016-2110-4.

14. Jakola ASUnsgård GMyrmel KSet alSurgical strategies in low-grade gliomas and implications for long-term quality of lifeJ Clin Neurosci2014;21:13049DOI10.1016/j.jocn.2013.11.027.

15. Kim YZKim C-YLim DHThe overview of practical guidelines for gliomas by KSNO, NCCN, and EANO. Brain Tumor Res Treat. 2022;10:8393. DOI: 10.14791/btrt.2022.0001.

16. van den Bent MJAfra Dde Witte Oet alLong-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: The EORTC 22845 randomised trial. Lancet. 2005;366:98590. DOI10.1016/S0140-6736(05)67070-5.

17. Buckner JCShaw EGPugh SLet alRadiation plus procarbazine, CCNU, and vincristine in low-grade gliomaN Engl J Med2016;374:134455DOI10.1056/NEJMoa1500925.

18. van den Bent MJChemotherapy for low-grade glioma: When, for whom, which regimen? Curr Opin Neurol. 2015;28:633938. DOI10.1097/WCO.0000000000000257.

19. Klein MHeimans JJAaronson NKet alEffect of radiotherapy and other treatment-related factors on mid-term to long-term cognitive sequelae in low-grade gliomas: A comparative study. Lancet. 2002;360:13618. DOI: 10.1016/s0140-6736(02)11398-5.

20. Douw LKlein MFagel SSet alCognitive and radiological effects of radiotherapy in patients with low-grade glioma: Long-term follow-up. Lancet Neurol. 2009;8:8108. DOI: 10.1016/S1474-4422(09)70204-2.

21. Mellinghoff IKvan den Bent MJBlumenthal DTet alVorasidenib in IDH1- or IDH2-mutant low-grade glioma. N Engl J Med. 2023;389:589601. DOI: 10.1056/NEJMoa2304194.

22. Mellinghoff IKPenas-Prado MPeters KBet alVorasidenib, a dual inhibitor of mutant IDH1/2, in recurrent or progressive glioma; results of a first-in-human phase I trial. Clin Cancer Res. 2021;27:44919. DOI: 10.1158/1078-0432.CCR-21-0611.

23. Mellinghoff IKLu MWen PYet alVorasidenib and Ivosidenib in IDH1-mutant low-grade glioma: A randomized, perioperative phase 1 trialNat Med2023;29:61522DOI10.1038/s41591-023-02473-7.

24. Abou-Alfa GKMeyer TCheng A-Let alCabozantinib (C) versus placebo (P) in patients (Pts) with advanced hepatocellular carcinoma (HCC) who have received prior sorafenib: results from the randomized phase III CELESTIAL trial. J Clin Oncol. 2018;36:Abstr.4019. DOI: 10.1200/JCO.2018.36.15_suppl.4019.

25. Dhillon SIvosidenib: First global approval. Drugs. 2018;78:150916. DOI: 10.1007/s40265-018-0978-3.

26. Mellinghoff IKEllingson BMTouat Met alIvosidenib in isocitrate dehydrogenase 1-mutated advanced glioma. J Clin Oncol. 2020;38:3398406. DOI: 10.1200/JCO.19.03327.

27. ClinicalTrials.govA Study of HMPL-306 in Advanced Solid Tumors With IDH Mutations. ClinicalTrials.gov identifier: NCT04762602. Available at: https://clinicaltrials.gov/study/NCT04762602 (Date last accessed: 18 October 2023).

28. ClinicalTrials.govA Study of DS-1001b in Patients With Chemotherapy- and Radiotherapy-Naive IDH1 Mutated WHO Grade II Glioma. ClinicalTrials.gov identifier: NCT04458272. Available athttps://clinicaltrials.gov/study/NCT04458272 (Date last accessed: 18 October 2023).

29. Natsume AArakawa YNarita Yet alThe first-in-human phase I study of a brain-Penetrant mutant Idh1 inhibitor DS-1001 in patients with recurrent or progressive Idh1-mutant gliomas. Neuro Oncol. 2023;25:32636. DOI10.1093/neuonc/noac155.

30. ClinicalTrials.govViCToRy: Vorasidenib in Combination With Tumor Specific Peptide Vaccine for Recurrent IDH1 Mutant Lower Grade Gliomas (ViCToRy). ClinicalTrials.gov identifier: NCT05609994. Available at: https://clinicaltrials.gov/study/NCT05609994 (Date last accessed: 18 October 2023).

31. ClinicalTrials.govStudy of Vorasidenib and Pembrolizumab Combination in Recurrent or Progressive Enhancing IDH-1 Mutant Astrocytomas. ClinicalTrials.gov identifier: NCT05484622 Available athttps://clinicaltrials.gov/study/NCT05484622 (Date last accessed: 18 October 2023).

32. ClinicalTrials.govIvosidenib (AG-120) With Nivolumab in IDH1 Mutant Tumors. ClinicalTrials.gov identifier: NCT04056910 Available atwww.clinicaltrials.gov/study/NCT04056910 (Date last accessed: 18 October 2023).

33. ClinicalTrials.govSafusidenib Phase 2 Study in IDH1 Mutant Glioma. ClinicalTrials.gov identifier: NCT05303519 Available atwww.clinicaltrials.gov/study/NCT05303519 (Date last accessed: 18 October 2023).

34. Aaronson NKTaphoorn MJBHeimans JJet alCompromised health-related quality of life in patients with low-grade gliomaJ Clin Oncol2011;29:44305DOI10.1200/JCO.2011.35.5750.

35. Affronti MLRandazzo DLipp ESet alPilot study to describe the trajectory of symptoms and adaptive strategies of adults living with low-grade glioma. Semin Oncol Nurs. 2018;34:47285. DOI: 10.1016/j.soncn.2018.10.006.

36. Chen HJudkins JThomas Cet alMutant IDH1 and seizures in patients with glioma. Neurology. 2017;88:180513. DOI: 10.1212/WNL.0000000000003911.

Further Resources

Share this Article
Related Content In Neuro-oncology
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72