Abstract

The 2016 and 2021 World Health Organization 2021 Classification of central nervous system tumors have resulted in a major improvement in the classification of isocitrate dehydrogenase (IDH)-mutant gliomas. With more effective treatments many patients experience prolonged survival. However, treatment guidelines are often still based on information from historical series comprising both patients with IDH wild-type and IDH-mutant tumors. They provide recommendations for radiotherapy and chemotherapy for so-called high-risk patients, usually based on residual tumor after surgery and age over 40. More up-to-date studies give a better insight into clinical, radiological, and molecular factors associated with the outcome of patients with IDH-mutant glioma. These insights should be used today for risk stratification and for treatment decisions. In many patients with IDH-mutant grades 2 and 3 glioma, if carefully monitored postponing radiotherapy and chemotherapy is safe, and will not jeopardize the overall outcome of patients. With the INDIGO trial showing patient benefit from the IDH inhibitor vorasidenib, there is a sizable population in which it seems reasonable to try this class of agents before recommending radio-chemotherapy with its delayed adverse event profile affecting quality of survival. Ongoing trials should help to further identify the patients that are benefiting from this treatment.

Key Points
  • The volume of the residual tumor of IDH-mutant tumors after surgery is of major prognostic significance for outcome, but not the age of the patient.

  • IDHmt tumors that are recurring after radiotherapy and/or chemotherapy usually have novel genetic alterations which are associated with poor outcomes.

  • An objective grading system of IDH-mutant gliomas based on molecular findings is needed.

The 2016 World Health Organization (WHO) classification of central nervous system (CNS) tumors that integrated isocitrate dehydrogenase (IDH) mutational status with histology in classifying gliomas, and the introduction of mutant IDH inhibitors (from here on: IDH inhibitors) into clinical practice has led to a reassessment of the biology and optimal therapies for grades 2 and 3 IDH-mutant gliomas. Under current guidelines, a “watch and wait” approach (ie, active monitoring without immediate adjuvant treatment after surgery) is typically restricted to patients with “low risk” IDH-mutant glioma. These are defined as younger patients (< 40 years) and after gross total resection or with limited residual disease (≤ 2 cm diameter after surgery), no functional deficits due to the tumor, and grade 2 histology.1 These recommendations are based on analyses from trials conducted prior to the discovery of the IDH mutation in 2008 and its introduction in the WHO classification of CNS tumors.2,3 Early trials on mutant IDH inhibitors suggested activity mainly in patients with non-enhancing IDHmt glioma tumors, as opposed to patients with enhancing tumors.4–6 Based on these observations, the recent phase 3 placebo-controlled INDIGO trial evaluated vorasidenib in a trial on IDH-mutant grade 2 glioma patients with measurable disease, who had undergone surgery as their only previous treatment, without an enhancing lesion on the MR scan and who were considered to be appropriate candidates for a watch and- wait approach.7 Benefit of vorasidenib was convincingly demonstrated: Median progression-free survival (PFS) improved from 11.1 months for patients in the placebo arm to 27.7 months for patients in the vorasidenib arm (HR 0.39, P < .001).7

A major question emerging from the INDIGO trial is whether these results are applicable only to patients fulfilling the narrow inclusion criteria of the trial, or whether current data on IDH-mutant glioma allow a broader biological perspective and allow generalizability beyond the INDIGO population (ie, beyond grade 2 and non-enhancing IDHmt tumors). To answer the question, a critical review is needed to evaluate if traditionally used risk factors for tumor progression and adverse outcomes truly capture the biology of the disease and correlate with outcomes of patients using the 2021 IDH-status-based glioma WHO CNS classification. This question is not only important in identifying patient groups that are most likely to benefit from treatment with IDH-mutant inhibitors; it has also major implications for patient referral for radiotherapy and chemotherapy after surgery.

Clinical Risk Factors of IDH-Mutant Glioma

In the pre-WHO 2016 era, several prognostic factors were well established for outcome in low-grade glioma patients: Age, performance status, presentation with seizures versus presence of neurological deficits, size of the tumor, radiological characteristics including the presence of enhancement, tumor growth rate (TGR), tumor grade and treatment including extent of resection.3,8,9 The historical studies that identified these factors included both IDHmt and IDH wild-type (IDHwt) tumors, and it cannot be assumed that they remain valid for determining prognosis in IDH-mutant glioma patients following the introduction of the WHO 2016 classification.10 In that same period, (post hoc) analyses of the trials in grades 2 and 3 gliomas receiving adjuvant chemotherapy after radiotherapy showed clinical benefit mainly in patients with IDH-mutant gliomas, with much longer survival in patients with grade 3 IDHmt tumors compared to historical studies (Table 1).2,11–15 With the increasing use of early maximal safe surgery and radiotherapy followed by alkylating chemotherapy, the natural history of these tumors can no longer be studied. Moreover, long-held clinical assumptions such as the impact of age and tumor grade on the outcome of patients with diffuse glioma are challenged in the era of integrated histomolecular glioma classification based on IDH mutation and 1p/19q codeletion status.16 New, contemporary cohort studies on patients with IDH-mutant gliomas are needed on all aspects of diagnosis and treatment to understand the outcome of patients.

Table 1.

The Range in Overall Survival in IDH-Mutant Glioma Grades 2 and 3 After Radiotherapy With Alkylating Chemotherapy as Reported in Several Larger Series (Source: Bell et al.,12 van den Bent et al.,15 Chang et al.,14 Lassman et al.,11 Minniti et al.17)

Grade 2Grade 3
Astrocytoma IDHmt11.4–11.8 years7.9–9.7 years
Oligodendroglioma IDHmt, 1p/19q codelNR13.2–14.2 years
Grade 2Grade 3
Astrocytoma IDHmt11.4–11.8 years7.9–9.7 years
Oligodendroglioma IDHmt, 1p/19q codelNR13.2–14.2 years

NR, not reached.

Table 1.

The Range in Overall Survival in IDH-Mutant Glioma Grades 2 and 3 After Radiotherapy With Alkylating Chemotherapy as Reported in Several Larger Series (Source: Bell et al.,12 van den Bent et al.,15 Chang et al.,14 Lassman et al.,11 Minniti et al.17)

Grade 2Grade 3
Astrocytoma IDHmt11.4–11.8 years7.9–9.7 years
Oligodendroglioma IDHmt, 1p/19q codelNR13.2–14.2 years
Grade 2Grade 3
Astrocytoma IDHmt11.4–11.8 years7.9–9.7 years
Oligodendroglioma IDHmt, 1p/19q codelNR13.2–14.2 years

NR, not reached.

Clinical Factors Associated With Outcome of Patients With IDHmt Tumors

Several recent studies reported on the outcome of patients with IDH-mutant gliomas. Table 2 summarizes several studies that combined clinical factors with imaging, resection, or pathology data.11,17–29 Most focused on specific features (eg, associations with extent of resection, specific pathological and molecular findings, grade, and imaging) and did not co-analyze other important (clinical) variables. Another limitation of these studies is the uniformly retrospective design with highly heterogeneous treatment patterns, in particular, the variable use of chemotherapy or radiotherapy depending on perceived risk factors. Still, they provide useful information on the prognostic significance of clinical, tumor, and treatment factors in patients with IDH-mutant glioma.

Table 2.

Overview of Clinical Series on IDH-Mutant Glioma and Identified Prognostic Factors, Including the Hazard Ratio for Overall Survival for Identified Significant Factors

AuthorTumor type, gradenAgePerformance statusgradePreoperative
Size/volume
Surgery/Postoperative sizePost-op treatmentlocation
Aoki20OD 2,31412.25 (0.89, 5.70)NRNSNR3.44 (1.59, 7.47) (Q)NRNS
A 2,3109NSNRNSNRNS (P = .07) (Q)NRNS
Carstam19OD21261.05 (1.02, 1.08)4.47 (1.70, 11.78)NA1.05 (1.02, 1.08)NS (Q)NSNS
Carstam18OD 2,379NSNSNS1.05 (1.01–1.09)NSNRNS
A 2, 389NSNSNSNS1.02 (1.01–1.03)NRNS
Minniti17A2103NSNSNANR0.27 (0.08, 0.87) (E)*NANS
Mair21OD 2,3183NSNSNSNRNS (Q)NSNR
Pal’a23OD, A 2144NSNRNANRNS (Q)20.2 (3.4–118.9)NR
Dao Trong23A, OD,2,3167NSNSNSNSNS (Q)NSNS
Tesileanu24A34322.30 (1.13 − 4.67)1.49 (1.06 − 2.08)NANR1.77 (1.07 − 2.91) (Q)NANR
Wijnenga25A, OD 2205NSNSNA1.01 (1.0–1.02)1.70 (1.06–2.75) (E)NSNR
Park26OD 2,3861.07 (1.04–1.11)NS5.2 (1.7–15.3)NRNS (Q)NSNS
A 2, 3211NS0.97 (0.95–1.00)NR 0.30 (0.16–0.59) (Q)2.99 (1.51–5.93)NS
EORTC 2695111OD 3800.36 (0.17, 0.69)NSNANRNS (Q)NANR
RTOG 940211OD 3125NS0.36 (0.20, 0.64)NANRNRNANR
Tran27A2, 3118NSNSSNS 1.02 (E)NANR
Appay28OD3,4834.0 (1.8–9.0)NR0.58NR 0.58 (0.31–1.09) (Q)NSNR
A3, 44281.6 (1.1–2.5)NR2.9 (1.8–4.8)NR 0.46 (0.28–0.75) (Q)0.29 (0.19–0.46)NR
Weller29A2-4258NSNS3.1 (1.4-7.0)**NR2.61 (1.5-4.6) (Q)NSNR
AuthorTumor type, gradenAgePerformance statusgradePreoperative
Size/volume
Surgery/Postoperative sizePost-op treatmentlocation
Aoki20OD 2,31412.25 (0.89, 5.70)NRNSNR3.44 (1.59, 7.47) (Q)NRNS
A 2,3109NSNRNSNRNS (P = .07) (Q)NRNS
Carstam19OD21261.05 (1.02, 1.08)4.47 (1.70, 11.78)NA1.05 (1.02, 1.08)NS (Q)NSNS
Carstam18OD 2,379NSNSNS1.05 (1.01–1.09)NSNRNS
A 2, 389NSNSNSNS1.02 (1.01–1.03)NRNS
Minniti17A2103NSNSNANR0.27 (0.08, 0.87) (E)*NANS
Mair21OD 2,3183NSNSNSNRNS (Q)NSNR
Pal’a23OD, A 2144NSNRNANRNS (Q)20.2 (3.4–118.9)NR
Dao Trong23A, OD,2,3167NSNSNSNSNS (Q)NSNS
Tesileanu24A34322.30 (1.13 − 4.67)1.49 (1.06 − 2.08)NANR1.77 (1.07 − 2.91) (Q)NANR
Wijnenga25A, OD 2205NSNSNA1.01 (1.0–1.02)1.70 (1.06–2.75) (E)NSNR
Park26OD 2,3861.07 (1.04–1.11)NS5.2 (1.7–15.3)NRNS (Q)NSNS
A 2, 3211NS0.97 (0.95–1.00)NR 0.30 (0.16–0.59) (Q)2.99 (1.51–5.93)NS
EORTC 2695111OD 3800.36 (0.17, 0.69)NSNANRNS (Q)NANR
RTOG 940211OD 3125NS0.36 (0.20, 0.64)NANRNRNANR
Tran27A2, 3118NSNSSNS 1.02 (E)NANR
Appay28OD3,4834.0 (1.8–9.0)NR0.58NR 0.58 (0.31–1.09) (Q)NSNR
A3, 44281.6 (1.1–2.5)NR2.9 (1.8–4.8)NR 0.46 (0.28–0.75) (Q)0.29 (0.19–0.46)NR
Weller29A2-4258NSNS3.1 (1.4-7.0)**NR2.61 (1.5-4.6) (Q)NSNR

Abbreviations: OD, oligodendroglioma; A, astrocytoma IDHmt, 2, grade 2; 3, grade 3; NA, not applicable; NS, not significant; NR, not reported.

Column surgery/postoperative size: (Q): qualitative = description extent of resection; (E): exact: measurement volume after resection; * Postoperative volume ≤ versus > 1 cm3 **grade 2 versus 3.

+ all treated with chemo-radiation, “study on elderly patients.

Table 2.

Overview of Clinical Series on IDH-Mutant Glioma and Identified Prognostic Factors, Including the Hazard Ratio for Overall Survival for Identified Significant Factors

AuthorTumor type, gradenAgePerformance statusgradePreoperative
Size/volume
Surgery/Postoperative sizePost-op treatmentlocation
Aoki20OD 2,31412.25 (0.89, 5.70)NRNSNR3.44 (1.59, 7.47) (Q)NRNS
A 2,3109NSNRNSNRNS (P = .07) (Q)NRNS
Carstam19OD21261.05 (1.02, 1.08)4.47 (1.70, 11.78)NA1.05 (1.02, 1.08)NS (Q)NSNS
Carstam18OD 2,379NSNSNS1.05 (1.01–1.09)NSNRNS
A 2, 389NSNSNSNS1.02 (1.01–1.03)NRNS
Minniti17A2103NSNSNANR0.27 (0.08, 0.87) (E)*NANS
Mair21OD 2,3183NSNSNSNRNS (Q)NSNR
Pal’a23OD, A 2144NSNRNANRNS (Q)20.2 (3.4–118.9)NR
Dao Trong23A, OD,2,3167NSNSNSNSNS (Q)NSNS
Tesileanu24A34322.30 (1.13 − 4.67)1.49 (1.06 − 2.08)NANR1.77 (1.07 − 2.91) (Q)NANR
Wijnenga25A, OD 2205NSNSNA1.01 (1.0–1.02)1.70 (1.06–2.75) (E)NSNR
Park26OD 2,3861.07 (1.04–1.11)NS5.2 (1.7–15.3)NRNS (Q)NSNS
A 2, 3211NS0.97 (0.95–1.00)NR 0.30 (0.16–0.59) (Q)2.99 (1.51–5.93)NS
EORTC 2695111OD 3800.36 (0.17, 0.69)NSNANRNS (Q)NANR
RTOG 940211OD 3125NS0.36 (0.20, 0.64)NANRNRNANR
Tran27A2, 3118NSNSSNS 1.02 (E)NANR
Appay28OD3,4834.0 (1.8–9.0)NR0.58NR 0.58 (0.31–1.09) (Q)NSNR
A3, 44281.6 (1.1–2.5)NR2.9 (1.8–4.8)NR 0.46 (0.28–0.75) (Q)0.29 (0.19–0.46)NR
Weller29A2-4258NSNS3.1 (1.4-7.0)**NR2.61 (1.5-4.6) (Q)NSNR
AuthorTumor type, gradenAgePerformance statusgradePreoperative
Size/volume
Surgery/Postoperative sizePost-op treatmentlocation
Aoki20OD 2,31412.25 (0.89, 5.70)NRNSNR3.44 (1.59, 7.47) (Q)NRNS
A 2,3109NSNRNSNRNS (P = .07) (Q)NRNS
Carstam19OD21261.05 (1.02, 1.08)4.47 (1.70, 11.78)NA1.05 (1.02, 1.08)NS (Q)NSNS
Carstam18OD 2,379NSNSNS1.05 (1.01–1.09)NSNRNS
A 2, 389NSNSNSNS1.02 (1.01–1.03)NRNS
Minniti17A2103NSNSNANR0.27 (0.08, 0.87) (E)*NANS
Mair21OD 2,3183NSNSNSNRNS (Q)NSNR
Pal’a23OD, A 2144NSNRNANRNS (Q)20.2 (3.4–118.9)NR
Dao Trong23A, OD,2,3167NSNSNSNSNS (Q)NSNS
Tesileanu24A34322.30 (1.13 − 4.67)1.49 (1.06 − 2.08)NANR1.77 (1.07 − 2.91) (Q)NANR
Wijnenga25A, OD 2205NSNSNA1.01 (1.0–1.02)1.70 (1.06–2.75) (E)NSNR
Park26OD 2,3861.07 (1.04–1.11)NS5.2 (1.7–15.3)NRNS (Q)NSNS
A 2, 3211NS0.97 (0.95–1.00)NR 0.30 (0.16–0.59) (Q)2.99 (1.51–5.93)NS
EORTC 2695111OD 3800.36 (0.17, 0.69)NSNANRNS (Q)NANR
RTOG 940211OD 3125NS0.36 (0.20, 0.64)NANRNRNANR
Tran27A2, 3118NSNSSNS 1.02 (E)NANR
Appay28OD3,4834.0 (1.8–9.0)NR0.58NR 0.58 (0.31–1.09) (Q)NSNR
A3, 44281.6 (1.1–2.5)NR2.9 (1.8–4.8)NR 0.46 (0.28–0.75) (Q)0.29 (0.19–0.46)NR
Weller29A2-4258NSNS3.1 (1.4-7.0)**NR2.61 (1.5-4.6) (Q)NSNR

Abbreviations: OD, oligodendroglioma; A, astrocytoma IDHmt, 2, grade 2; 3, grade 3; NA, not applicable; NS, not significant; NR, not reported.

Column surgery/postoperative size: (Q): qualitative = description extent of resection; (E): exact: measurement volume after resection; * Postoperative volume ≤ versus > 1 cm3 **grade 2 versus 3.

+ all treated with chemo-radiation, “study on elderly patients.

The prognostic effect of age was analyzed in a number of studies, with various age cutoffs. In most studies on patients with astrocytoma, IDHmt failed to identify age as a prognostic indicator.17,27,29,30 One study of grade 3 astrocytoma patients found an association only in patients over 60 years, while another study reported a modest association in patients over 50 years.16,24 In contrast, 3 studies on IDHmt and 1p/19q-codeleted oligodendroglioma found an association with age: Age over 60 (cutoff identified with regression analysis), age as a continuous variable, and age over 40.11,19,20 A study on grade 3 and 4 IDH-mutant glioma found a more pronounced association with age in grade 3 oligodendroglioma patients, using the observed median (49.5 years).28 Another study that investigated IDHmt and 1p/19q-codeleted oligodendroglioma patients over 60 years found no difference in comparison with patients under 60 years. In contrast, a report from the French POLA network showed worse outcomes in patients with IDHmt grades 3 and 4 tumors who were over 70 years old; most had been diagnosed with grade 3 IDH-mutant and 1p/19q-codeleted oligodendroglioma.23,31

Performance status was associated with outcome in some of the cohorts on IDHmt and 1p/19q-codeleted oligodendroglioma (either performance status or neurological function) but in only one study on grade 3 IDH-mutant astrocytoma.11,19,24,26 The French POLA network study on elderly patients reported significant associations with outcome and a variety of clinical factors basically reflecting neurological function.31

Most of these series incorporated either tumor volume, extent of resection, and/or postoperative volume, mostly using only descriptive, categorical methodology for the description of extent of resection (eg, biopsy versus resection or versus partial or complete resection) without an attempt to quantify the post-resection tumor volume. Still, the association of tumor size and type of surgery with the outcome of patients was noted in many studies and emerged as significant in multivariate analysis especially if a quantitative assessment of postoperative volume was part of the study.17,18,25 With respect to postoperative treatments, several papers reported unfavorable outcomes after adjuvant treatment (be it radiotherapy, chemotherapy, or both).22,26 Others found no association impact with outcome.19,25 However, invariably, these series reveal a bias towards the selection of postoperative treatment for patients with unfavorable risk factors (especially less than complete resections).17,21,22,26,32,33 A series on patients with grades 3 and 4 tumors reported improved outcomes if postoperative treatment had been given.28 Some studies are limited to patients having undergone radiotherapy with or without chemotherapy.11,15,17 Three prospective randomized studies found improved outcomes if chemotherapy was added to radiotherapy.2,11,15 Two studies addressed immediate postoperative treatment versus delayed treatment and found no effect on survival; but here also patient selection appears to have played a major role in the choice for early treatment.17,21 None of the studies that investigated tumor location reported an association with survival. Importantly, a series on conservatively managed resected grade 3 glioma observed a 3.4-year median interval between surgery and the next oncological treatment.34 Codeletion status, pre-and postoperative volume, and TGR were associated with the time to the next treatment.

Association With Tumor Size, Extent of Resection and Outcome

Maximal safe microsurgical resection is the standard of care for patients with IDH-mutant gliomas with and without 1p/19q-codeletion and has been associated with improved outcome.1 In the absence of randomized studies, one might argue that extent of resection reflects an indirect marker for tumor localization, preoperative tumor size, or invasiveness all with an inherently worse prognosis. Preoperative tumor size may be prognostic for patient outcome as larger tumors may increase the probability of infiltration into eloquent areas, limit resectability, and signify a greater risk of malignant transformation.18,25,32,35 To assess the relationship between the extent of resection and patient outcome, recent studies on grade 2 IDH-mutant glioma patients focused on the quantification of residual tumor volume (measured in cm3) which came out of the analyses of higher relevance than the relative -percentage- of tumor volume reduction.18,25,32,35–37 These studies show that preoperative tumor size was prognostic in both IDHmt astrocytoma and IDHmt and 1p/19q-codeleted oligodendroglioma patients, whereas residual tumor volume was predominantly associated with prognosis in IDHmt astrocytoma patients. When astrocytoma IDH-mutant patients were stratified by residual tumor volume, survival curves of astrocytoma patients were split within the first 5 years following initial resection.25,32,35 In contrast, patients with IDH-mutant and 1p/19q-codeleted oligodendroglioma seem to derive a survival benefit from smaller postoperative tumor volumes only after more prolonged follow-up (>5–10 years).25,32 The role of supramaximal resection in IDH-mutant glioma remains controversial, due in part to the lack of a reliable method to quantify the extent of resection beyond the T2/FLAIR-hyperintense tumor borders. Functional and anatomical borders might be key to guide resection in selected cases but the patient benefit of further increasing the extent of resection will need to be weighed against the risk for neurologic deficits.32,38–40 While it remains unclear whether longer observation periods or larger series will reveal a trend towards a survival benefit after supramaximal resection of IDH-mutant and 1p/19q-codeleted oligodendroglioma, the yet considerable median follow-up over 11.7 years in a recent study supports the notion that the benefit of resection beyond the tumor borders is likely to be limited in patients with IDHmt and 1p/19q-codeleted oligodendroglioma.32 Studies on the prognostic role of extent of resection for grades 3 or 4 IDHmt astrocytomas and grade 3 IDHmt and. 1p/19q-codeleted oligodendroglioma patients are rare. More complete resection with lower residual T2-weighted tumor volumes was also associated with favorable survival in 113 patients with IDHmt astrocytomas, of which 86 patients had grade 3 histology (and the remaining 27 patients had grade 4 histology); this finding was confirmed in other retrospective studies on astrocytomas grade 3 and 4.37,38,41 A small single-institutional study on patients with grade 3 IDH-mutant and 1p/19q-codeleted oligodendroglioma failed to detect an association between the extent of resection and survival.42

Imaging and Contrast Enhancement

Relation Between Tumor Grade and Enhancement, Patient Outcome

Survival of patients with IDHmt glioma correlated with contrast enhancement on MR imaging in several series.43–46 In patients with IDHmt and 1p/19q-codeleted oligodendroglioma, a positive correlation has been reported between MR contrast enhancement and grade.47,48 Neo-angiogenesis and mitotic counts were independently associated with TGRs (TGR) ≥ 8 mm/year.49 Contrast enhancement in grade 3 IDH-mutant and 1p/19q-codeleted oligodendroglioma patients was associated with worse outcome, with larger tumor volumes and with several molecular factors.50,51 Contrast enhancement had a sensitivity of about 60% in identifying grade 3 IDH-mutant and 1p/19q-codeleted oligodendroglioma, but up to 50% of grade 2 IDHmt and 1p/19q-codeleted oligodendroglioma may show some enhancement and a substantial proportion of grade 3 IDHmt and 1p/19q-codeleted oligodendroglioma do not.52,53 In IDH-mutant astrocytoma, a positive association has also been reported between MR contrast enhancement and grade.47,53–55 Still, on initial brain MRI only 60% of grade 3 IDH-mutant astrocytomas showed patchy and faint enhancement, whereas 20% to 50% of grade 2 IDH-mutant astrocytomas showed some enhancement.43,47 Ring enhancement has been associated with grade 4 IDH-mutant astrocytomas, and marked enhancement was associated with the presence of homozygous CDKN2A deletion.47,56 However, the identification of homozygous CDKN2A deletion based on MRI had limited sensitivity (80%) and specificity (58%).56 Nodular and ring enhancement patterns on contrast-enhanced MRI have also been reported in grade 3 IDH-mutant astrocytomas, and were associated with worse outcomes.43,47,51 In previously non-enhancing IDH-mutant gliomas, contrast enhancement at progression typically indicates tumor progression to a higher grade of malignancy, associated with more aggressive behavior of the tumor and a worse prognosis.57 Both temozolomide and radiotherapy have been associated with the induction of novel genetic alterations (hypermutation, small and large DNA deletions) associated with poor outcome, implying a change in biology induced by oncolytic treatment.57–59 However, as a word of caution, studies have shown that 25%–30% of patients with IDH-mutant gliomas may develop pseudoprogression following radiotherapy.60,61

TGR as a Measure of Prognosis

Quantitative longitudinal studies of imaging growth patterns in patients with IDH-mutant glioma have confirmed the continuous tumor growth without treatment and established correlations with molecular status.49,62–64 Growth was slower in IDH-mutant and 1p/19q-codeleted oligodendroglioma than in IDHmt astrocytomas, and was also slower in grade 2 IDH-mutant and 1p/19q-oligodendroglioma codeleted than in grade 3 oligodendroglioma, IDH-mutant and 1p/19q-codeleted.49,62 Similarly, TGR correlated with grade in IDH-mutant astrocytoma (grades 4 > 3 > 2). In a cohort of IDHmt glioma patients on active surveillance (n = 128) a continuous percentage tumor volume growth rate per 6 months of 10.46% (95% CI: [9.11%, 11.83%]) and a doubling time of 3.5 years (95% CI: [3.10–3.98]) was noted, with higher rates in the presence of homozygous CDKN2A deletion.63 Each tumor volume increase of one natural logarithm was associated with a more than 3-fold increase in risk of death. When quantifying the tumor growth by the evolution of the mean tumor diameter over time, a cutoff ≥ 8 mm/year was systematically associated with IDHmt and 1p/19q-codeleted oligodendroglioma grade 3 rather than grade 2, and with either IDHmt astrocytomas grade 3 or 4.65 Using the current WHO classification, spontaneous tumor growth was observed to be a predictor of tumor progression requiring further oncological treatment in oligodendrogliomas and being a predictor of time to malignant transformation and of overall survival of patients with IDH-mutant glioma.49,62,63,65,66

Histologic Parameters for Distinguishing Grades 2 and 3 IDH-Mutant Gliomas

The traditional method for distinguishing histologic grade 2 from grade 3 in diffuse gliomas is based on the microscopic assessment of features of focal or dispersed anaplasia (such as increased cell density and nuclear atypia), and mitotic activity. This grading system is however subject to considerable interobserver variation.67

Grading of IDH-Mutant Astrocytoma

The 2021 WHO classification of CNS tumors states that in contrast to IDH-mutant astrocytomas grade 2, IDHmt astrocytomas grade 3 “exhibit focal or dispersed anaplasia and display significant mitotic activity.”68 IDHmt astrocytoma grade 3 may also feature atypical mitoses and/or multinucleated tumor cells, but microvascular proliferation, necrosis, and homozygous CDKN2A/CDKN2B deletion, ie, criteria for CNS WHO grade 4, are absent.68 Studies performed in the pre-IDH era indicated that diffuse astrocytomas with ≥ 2 mitoses per 10 high power fields (HPF) were associated with shorter survival than those with 0 or 1 mitoses and this threshold has been used by neuropathologists for the designation of WHO grade 3.69,70 In several recent studies of IDH-mutant astrocytoma cohorts, these thresholds for mitotic activity were not corroborated and no difference in survival between grades 2 and 3 tumors were observed.30,71–73 However, others have demonstrated that WHO grading schemes can stratify risk among patients with grades 2–4 IDH-mutant astrocytomas, yet with opportunity for improvement.29,74–77 A study based on selected patient cohorts included in EORTC trials 26053 (CATNON) and 22033-26033 reported that ≤ 2 mitoses per 10 microscopic HPF was significantly associated with longer PFS in patients with IDHmt astrocytoma without homozygous CDKN2A/CDKN2B deletion.78 A population-based report on clinical outcomes of IDHmt astrocytoma patients who were diagnosed according to the 2016 WHO classification of CNS tumors demonstrated that patients with WHO grade 2 IDH-mutant astrocytomas had a modest, but statistically significant, higher survival rate at 1 year than patients with IDH-mutant grade 3 astrocytomas (97.9% and 94.4%).79 A study of 118 IDH-mutant astrocytoma patients demonstrated that mitotic count (≥ 6/3mm2) was associated with a shorter time to postoperative treatment.27 Patients with tumors with mitotic activity lower than the cutoff and a post-surgical residual volume < 1 cm3 appeared to be the optimal candidates for observational follow-up. Studies of the proliferative index (eg, based on Ki-67 immunostaining) have not identified a robust cutoff to distinguish grade 2 from grade 3 IDHmt astrocytomas, although several studies reported worse outcomes in patients whose tumors displayed high proliferative activity.27,71

Grading of Oligodendroglioma IDHmt and 1p/19q Codeleted

Similar to IDHmt astrocytomas, IDHmt, and 1p/19q-codeleted oligodendroglioma represent a continuous spectrum ranging from indolent and well-differentiated tumors to those that are rapidly progressing. Here the grading scheme is also based on morphologic features. The histological criteria for IDHmt and 1p/19q-codeleted oligodendroglioma grading were established in the pre-IDH era and have been maintained in the 2021 WHO classification of CNS tumors. These criteria were largely based on work which indicated that grade 3 tumors should be distinguished from grade 2 tumors by either the presence of brisk mitotic activity (≥ 6 mitoses per HPF; ≥ 2.5 mitoses/mm2), microvascular proliferation, or necrosis.80 However, the 2021 WHO CNS classification emphasized that “data defining a clear cutoff point for a mitotic count that distinguish CNS WHO grade 2 from CNS WHO grade 3 of IDHmt 1p/19q co-deleted oligodendroglioma are not available.”68 Multiple studies demonstrated that patients with grade 2 IDHmt and 1p/19q-codeleted oligodendroglioma have significantly longer survival compared to patients with grade 3 tumors based on the above histologic criteria30,74,76 However, recent studies of IDHmt- and 1p/19q-codeleted oligodendroglioma grade 3 indicate that patients with tumors displaying elevated mitotic activity (≥6/10 HPF; 0.24 mm2) but no microvascular proliferation or necrosis have significantly longer PFS and OS than those whose tumors show microvascular proliferation or necrosis.81,82 This suggests that elevated mitotic activity alone is not a strong prognostic marker for aggressive clinical behavior in this tumor type. The variability noted in study outcomes based on mitotic thresholds may be due to an inherent lack of reproducibility in the microscopic assessment of mitotic count, reflecting challenges in consistently recognizing mitoses, in the variable selection of microscopic fields, and in non-standardized counting techniques.

Prognostic Genetic Markers

With the recent advances in molecular diagnostics, an important clinical question is whether molecular markers can provide a better way of risk stratification for patients with IDH-mutant glioma. More potential markers have been identified in IDH-mutant astrocytoma than in IDH-mutant and 1p/19q codeleted oligodendroglioma patients.

Prognostic Genetic Markers: IDHmt Astrocytomas

Non-canonical IDH mutations have been associated with a better outcome in astrocytoma, possibly related to different levels of 2-HG production.83

CDKN2A/B Hemizygous Deletion and Mutation

CDKN2A/B homozygous deletion is now recognized as a criterion for establishing the diagnosis of IDH-mutant astrocytoma grade 4, based on the finding of short overall survival associated with loss of both alleles.75,84–86 However, the presence of hemizygous deletion of CDKN2A/B was also shown to be a marker of less favorable outcome prognosis in patients with IDH-mutant astrocytomas when compared to patients with CDKN2A/B non-deleted tumors. Recent investigations found a significantly shorter survival among patients with IDHmt astrocytomas (all grades included) with hemizygous deletion of CDKN2A/B on multivariate analysis in independent datasets, with an intermediate overall survival of patients with hemizygous deletion of CDKN2A/B compared to patients with CDKN2A/B homozygously deleted tumors and patients whose tumors lacked copy number losses of CDKN2A/B.87,88 Hemizygous deletion was identified in 10% of grade 2, 27% of grade 3, and 33% of grade 4 IDHmt astrocytomas. The finding of any allelic loss of CDKN2A/B was associated with shorter survival on multivariate analysis, which included histologic grade. Two studies identified mutation of CDKN2A/B, an uncommon event in IDH-mutant astrocytoma (2.6%), to be associated with a poor prognosis similar to CDKN2A/B homozygous deletions on univariable analysis.89 The prognostic role of CDKN2A/B promoter methylation remains unclear.

Alteration of Other RB Pathway Genes

Analyses of large cohorts of patients with IDH-mutant astrocytoma grades 2–4 have shown that CDK4 amplification is associated with shorter survival on multivariate analysis.88,90 Other studies have concluded that CDK4 amplification, when considered by itself, was not associated with poor prognosis on univariate analysis.75,91 Since CDK4 is a member of the RB pathway (Figure 1) and its amplification is mutually exclusive with CDKN2A/B homozygous deletion and RB1 mutation, some investigators have explored alterations of RB pathway members as a single risk factor. Multivariate analysis of sizable patient cohorts reported that when altered RB pathway genes (CDKN2A/B homozygous deletion, CDK4 amplification, or RB1 mutation) were considered together, it was a strong and statistically significant predictor of poor prognosis among patients with grade 2 or 3 IDHmt astrocytoma.92,93 Others have used this approach to combine CDK4 amplification and CDKN2A/B homozygous as a single risk variable and demonstrated a significant association with shorter overall survival when either one of these findings was present.77 Similarly, a copy number analysis model for predicting outcomes in histologic grades 2 and 3 IDHmt astrocytomas indicated that the presence of either homozygous CDKN2A/B homozygous deletion or CDK4 amplification was associated with shorter survival and that the additional finding of chromosome 14 loss predicted an even shorter survival.76,94 Homozygous deletion of RB1 was strongly associated with inferior overall survival among IDH-mutant astrocytomas on univariate analysis.75 Analysis of 2 cohorts of histologic grades 2–4 IDH-mutant astrocytomas have demonstrated shorter survival associated with CCND2 amplification.75,95 Both mutual exclusivity with homozygous deletion of CDKN2A and the association with grade are suggestive of a role for CDK6 amplification in tumor malignancy. Similar correlations with tumor grade are found for all members of the RB pathway mentioned in this paragraph (Table 3).

Table 3.

The Frequency of Specific Alterations in Astrocytoma, IDHmt of Various Grades

genedatasetIIWHO2016 grade
III
IV
CDKN2ATCGA4/111(4%)7/104(7%)4/16(25%)
MSK0/44 (0%)13/92 (14%)11/38 (29%)
Korshunov42/97 (43%)
Lee0/16 (0%)0/43 (0%)14/36 (39%)
CDK4TCGA1/111(1%)5/104(5%)5/16(31%)
MSK0/49 (0%)5/100 (5%)5/42 (12%)
Korshunov21/97 (22%)
Wong15/53(28%)
Lee0/16 (0%)0/43 (0%)5/36 (14%)
CDK6TCGA0/111(0%)3/104(3%)2/16(13%)
MSK1/49 (2%)2/100 (2%)1/42 (2%)
Wong5/53(9%)
Lee0/16 (0%)1/43 (2%)0/36 (0%)
RB1TCGA2/111(2%)2/104(2%)0/16(0%)
MSK0/49 (0%)1/100 (1%)4/42 (10%)
Korshunov11/97(11%)
PDGFRATCGA1/111(1%)9/104(9%)1/16(6%)
MSK0/49 (0%)8/100 (8%)5/42 (12%)
Korshunov18/97 (19%)
Lee0/16 (0%)0/43 (0%)6/36 (17%)
PIK3CATCGA2/111(2%)1/104(1%)1/16(6%)
MSK0/57 (0%)8/117 (7%)8/46 (17%)
CGGA1/20(5%)0/22(0%)2/13(15%)
Korshunov12/97(12%)
Wong2/53 (4%)
Lee0/16 (0%)1/43 (2%)3/36 (8%)
PIK3R1TCGA2/111(2%)3/104(3%)3/16(19%)
MSK0/57 (0%)6/117 (5%)5/46 (11%)
CGGA0/20(0%)1/22 (5%)3/13(23%)
Korshunov1/97(1%)
Wong3/53 (6%)
MYCNTCGA2/111(2%)0/104(0%)2/16(13%)
MSK1/49 (2%)5/100 (5%)5/42 (12%)
Korshunov12/97(12%)
Wong4/53 (8%)
Lee0/16 (0%)0/43 (0%)3/36 (8%)
CCND2TCGA9/111(8%)8/104(8%)3/16(19%)
MSK1/49 (2%)4/100 (4%)4/42 (10%)
Korshunov21/97 (22%)
Lee0/16 (0%)0/43 (0%)2/36 (6%)
genedatasetIIWHO2016 grade
III
IV
CDKN2ATCGA4/111(4%)7/104(7%)4/16(25%)
MSK0/44 (0%)13/92 (14%)11/38 (29%)
Korshunov42/97 (43%)
Lee0/16 (0%)0/43 (0%)14/36 (39%)
CDK4TCGA1/111(1%)5/104(5%)5/16(31%)
MSK0/49 (0%)5/100 (5%)5/42 (12%)
Korshunov21/97 (22%)
Wong15/53(28%)
Lee0/16 (0%)0/43 (0%)5/36 (14%)
CDK6TCGA0/111(0%)3/104(3%)2/16(13%)
MSK1/49 (2%)2/100 (2%)1/42 (2%)
Wong5/53(9%)
Lee0/16 (0%)1/43 (2%)0/36 (0%)
RB1TCGA2/111(2%)2/104(2%)0/16(0%)
MSK0/49 (0%)1/100 (1%)4/42 (10%)
Korshunov11/97(11%)
PDGFRATCGA1/111(1%)9/104(9%)1/16(6%)
MSK0/49 (0%)8/100 (8%)5/42 (12%)
Korshunov18/97 (19%)
Lee0/16 (0%)0/43 (0%)6/36 (17%)
PIK3CATCGA2/111(2%)1/104(1%)1/16(6%)
MSK0/57 (0%)8/117 (7%)8/46 (17%)
CGGA1/20(5%)0/22(0%)2/13(15%)
Korshunov12/97(12%)
Wong2/53 (4%)
Lee0/16 (0%)1/43 (2%)3/36 (8%)
PIK3R1TCGA2/111(2%)3/104(3%)3/16(19%)
MSK0/57 (0%)6/117 (5%)5/46 (11%)
CGGA0/20(0%)1/22 (5%)3/13(23%)
Korshunov1/97(1%)
Wong3/53 (6%)
MYCNTCGA2/111(2%)0/104(0%)2/16(13%)
MSK1/49 (2%)5/100 (5%)5/42 (12%)
Korshunov12/97(12%)
Wong4/53 (8%)
Lee0/16 (0%)0/43 (0%)3/36 (8%)
CCND2TCGA9/111(8%)8/104(8%)3/16(19%)
MSK1/49 (2%)4/100 (4%)4/42 (10%)
Korshunov21/97 (22%)
Lee0/16 (0%)0/43 (0%)2/36 (6%)

Datasets: TCGA95; MSK96; CGGA97; Korshunov98; Wong99; Lee.100 Grading was extracted from the manuscripts/datasets and was done according to presented (WHO2016), except for Lee et al (WHO2021).

Table 3.

The Frequency of Specific Alterations in Astrocytoma, IDHmt of Various Grades

genedatasetIIWHO2016 grade
III
IV
CDKN2ATCGA4/111(4%)7/104(7%)4/16(25%)
MSK0/44 (0%)13/92 (14%)11/38 (29%)
Korshunov42/97 (43%)
Lee0/16 (0%)0/43 (0%)14/36 (39%)
CDK4TCGA1/111(1%)5/104(5%)5/16(31%)
MSK0/49 (0%)5/100 (5%)5/42 (12%)
Korshunov21/97 (22%)
Wong15/53(28%)
Lee0/16 (0%)0/43 (0%)5/36 (14%)
CDK6TCGA0/111(0%)3/104(3%)2/16(13%)
MSK1/49 (2%)2/100 (2%)1/42 (2%)
Wong5/53(9%)
Lee0/16 (0%)1/43 (2%)0/36 (0%)
RB1TCGA2/111(2%)2/104(2%)0/16(0%)
MSK0/49 (0%)1/100 (1%)4/42 (10%)
Korshunov11/97(11%)
PDGFRATCGA1/111(1%)9/104(9%)1/16(6%)
MSK0/49 (0%)8/100 (8%)5/42 (12%)
Korshunov18/97 (19%)
Lee0/16 (0%)0/43 (0%)6/36 (17%)
PIK3CATCGA2/111(2%)1/104(1%)1/16(6%)
MSK0/57 (0%)8/117 (7%)8/46 (17%)
CGGA1/20(5%)0/22(0%)2/13(15%)
Korshunov12/97(12%)
Wong2/53 (4%)
Lee0/16 (0%)1/43 (2%)3/36 (8%)
PIK3R1TCGA2/111(2%)3/104(3%)3/16(19%)
MSK0/57 (0%)6/117 (5%)5/46 (11%)
CGGA0/20(0%)1/22 (5%)3/13(23%)
Korshunov1/97(1%)
Wong3/53 (6%)
MYCNTCGA2/111(2%)0/104(0%)2/16(13%)
MSK1/49 (2%)5/100 (5%)5/42 (12%)
Korshunov12/97(12%)
Wong4/53 (8%)
Lee0/16 (0%)0/43 (0%)3/36 (8%)
CCND2TCGA9/111(8%)8/104(8%)3/16(19%)
MSK1/49 (2%)4/100 (4%)4/42 (10%)
Korshunov21/97 (22%)
Lee0/16 (0%)0/43 (0%)2/36 (6%)
genedatasetIIWHO2016 grade
III
IV
CDKN2ATCGA4/111(4%)7/104(7%)4/16(25%)
MSK0/44 (0%)13/92 (14%)11/38 (29%)
Korshunov42/97 (43%)
Lee0/16 (0%)0/43 (0%)14/36 (39%)
CDK4TCGA1/111(1%)5/104(5%)5/16(31%)
MSK0/49 (0%)5/100 (5%)5/42 (12%)
Korshunov21/97 (22%)
Wong15/53(28%)
Lee0/16 (0%)0/43 (0%)5/36 (14%)
CDK6TCGA0/111(0%)3/104(3%)2/16(13%)
MSK1/49 (2%)2/100 (2%)1/42 (2%)
Wong5/53(9%)
Lee0/16 (0%)1/43 (2%)0/36 (0%)
RB1TCGA2/111(2%)2/104(2%)0/16(0%)
MSK0/49 (0%)1/100 (1%)4/42 (10%)
Korshunov11/97(11%)
PDGFRATCGA1/111(1%)9/104(9%)1/16(6%)
MSK0/49 (0%)8/100 (8%)5/42 (12%)
Korshunov18/97 (19%)
Lee0/16 (0%)0/43 (0%)6/36 (17%)
PIK3CATCGA2/111(2%)1/104(1%)1/16(6%)
MSK0/57 (0%)8/117 (7%)8/46 (17%)
CGGA1/20(5%)0/22(0%)2/13(15%)
Korshunov12/97(12%)
Wong2/53 (4%)
Lee0/16 (0%)1/43 (2%)3/36 (8%)
PIK3R1TCGA2/111(2%)3/104(3%)3/16(19%)
MSK0/57 (0%)6/117 (5%)5/46 (11%)
CGGA0/20(0%)1/22 (5%)3/13(23%)
Korshunov1/97(1%)
Wong3/53 (6%)
MYCNTCGA2/111(2%)0/104(0%)2/16(13%)
MSK1/49 (2%)5/100 (5%)5/42 (12%)
Korshunov12/97(12%)
Wong4/53 (8%)
Lee0/16 (0%)0/43 (0%)3/36 (8%)
CCND2TCGA9/111(8%)8/104(8%)3/16(19%)
MSK1/49 (2%)4/100 (4%)4/42 (10%)
Korshunov21/97 (22%)
Lee0/16 (0%)0/43 (0%)2/36 (6%)

Datasets: TCGA95; MSK96; CGGA97; Korshunov98; Wong99; Lee.100 Grading was extracted from the manuscripts/datasets and was done according to presented (WHO2016), except for Lee et al (WHO2021).

Schematic representation of the retinoblastoma (Rb) and receptor tyrosine kinase (RTK) pathway.
Figure 1.

Schematic representation of the retinoblastoma (Rb) and receptor tyrosine kinase (RTK) pathway.

Tyrosine Kinase Receptor/PI3K/PTEN Pathway

In several studies, PI3K pathway alterations (Figure 1), as defined by either PIK3CA or PIK3R1 mutation were a marker of poor prognosis among patients with grades 2 and 3 IDH-mutant astrocytomas but not invariably.90,92,93,100 The positive correlation between tumor grade and mutation frequency in both PIK3CA and PIK3R1 is also suggestive of a negative prognostic impact (Table 3).96–99,101,102 Multiple studies of large cohorts of patients with IDH-mutant astrocytomas have demonstrated that PDGFRA amplification is associated with poor prognosis, even in histological grade 2 tumors (Figure 1).75,77,90,93 The increased frequency of PDGFRA amplification in higher-grade tumors, and the high frequency in grade 4 IDH-mutant astrocytomas also points towards a negative prognostic impact (Table 3).

MYCN Amplification

MYCN amplification has been shown to be associated with shorter survival in patients with IDH-mutant astrocytomas (grades 2–4)75,93,96 The frequency of MYCN-amplification increases with WHO 2016 tumor grade (Table 3), and was relatively high (8%–12%) in 2 separate cohorts of grade 4 IDHmt astrocytomas.99,101

Chromosomal Instability and Tumor Mutational Burden in IDH-Mutant Astrocytoma

Increased aneupleudy and/or total CNV load is associated with poorer prognosis and poorer prognostic methylation classes in IDH-mutant astrocytomas.24,73,75,103,104 High mutational burden has been described in recurrent IDHmt astrocytoma in particular following treatment with temozolomide due to defects in mismatch repair genes, which is associated with enhancing recurrences and a worse prognosis and development of discontiguous disease.57,105

Prognostic Genetic Markers: IDHmt and 1p/19q-codeleted Oligodendroglioma

Chromosomal Arm 9p Loss and CDKN2A/B Homozygous Deletion

Deletions on 9p have been associated with histological grade 3 and contrast enhancement on MRI in patients with IDHmt and 1p/19q-codeleted oligodendroglioma.106,107 Several studies have linked deletions on 9p encompassing the CDKN2A/B locus on 9p21 to shorter survival in patients with grade 3 IDHmt and 1p/19q-codeleted oligodendroglioma, although not invariably.107,108 Homozygous deletion of CDKN2A has been found in approximately 10% of grade 3 oligodendroglioma, IDH-mutant, and 1p/19q-codeleted, but was not detected in grade 2 tumors (Table 4).28,108 Tumors with homozygous CDKN2A/B deletion typically demonstrate contrast enhancement upon imaging, and within grade 3 IDH-mutant and 1p/19q-codeleted oligodendroglioma, patients with tumors with a homozygous CDKN2A deletion had shorter survival.28,50,91 In addition, CDKN2A/B homozygous deletions are frequent in the recently identified aggressive oligosarcoma subgroup.109 Point mutations in CDKN2A or CDKN2B, amplification of CDK4 or CCND1, and homozygous deletion of RB1 are rare or absent in oligodendroglioma, IDH-mutant and 1p/19q-codeleted (Table 4), and so far no prognostic roles for CDK4 amplification and homozygous deletion of RB1 in grade 3 IDHmt and 1p/19q-codeleted oligodendroglioma patients has been identified.28

Table 4.

The Frequency of Alterations in IDH-Mutant and 1p/19q-Codeleted Oligodendroglioma Grades 2 and 3

GeneDatasetGrade 2grade 3
CDKN2A homodelTCGA0/48 (0%)0/37 (0%)
MSK0/36 (0%)0/43 (0%)
POLA33/483 (7%)
CICTCGA27/48 (56%)20/37 (54%)
MSK25/36 (69%)34/43 (79%)
FUBP1TCGA12/48 (35%)11/37 (29%)
MSK8/36 (22%)16/43 (37%)
NOTCH1TCGA4/48 (8%)14/37 (38%)
MSK2/36 (5%)14/43 (32%)
PIK3CATCGA10/48 (21%)6/37 (16%)
MSK3/36 (8%)14/43 (32%)
PIK3R1TCGA2/48 (4%)4/37 (11%)
MSK5/36 (14%)7/43 (16%)
CDK4amplPOLA1/483
GeneDatasetGrade 2grade 3
CDKN2A homodelTCGA0/48 (0%)0/37 (0%)
MSK0/36 (0%)0/43 (0%)
POLA33/483 (7%)
CICTCGA27/48 (56%)20/37 (54%)
MSK25/36 (69%)34/43 (79%)
FUBP1TCGA12/48 (35%)11/37 (29%)
MSK8/36 (22%)16/43 (37%)
NOTCH1TCGA4/48 (8%)14/37 (38%)
MSK2/36 (5%)14/43 (32%)
PIK3CATCGA10/48 (21%)6/37 (16%)
MSK3/36 (8%)14/43 (32%)
PIK3R1TCGA2/48 (4%)4/37 (11%)
MSK5/36 (14%)7/43 (16%)
CDK4amplPOLA1/483

TCGA: data extracted from Gliovis.

POLA: reference89.

Table 4.

The Frequency of Alterations in IDH-Mutant and 1p/19q-Codeleted Oligodendroglioma Grades 2 and 3

GeneDatasetGrade 2grade 3
CDKN2A homodelTCGA0/48 (0%)0/37 (0%)
MSK0/36 (0%)0/43 (0%)
POLA33/483 (7%)
CICTCGA27/48 (56%)20/37 (54%)
MSK25/36 (69%)34/43 (79%)
FUBP1TCGA12/48 (35%)11/37 (29%)
MSK8/36 (22%)16/43 (37%)
NOTCH1TCGA4/48 (8%)14/37 (38%)
MSK2/36 (5%)14/43 (32%)
PIK3CATCGA10/48 (21%)6/37 (16%)
MSK3/36 (8%)14/43 (32%)
PIK3R1TCGA2/48 (4%)4/37 (11%)
MSK5/36 (14%)7/43 (16%)
CDK4amplPOLA1/483
GeneDatasetGrade 2grade 3
CDKN2A homodelTCGA0/48 (0%)0/37 (0%)
MSK0/36 (0%)0/43 (0%)
POLA33/483 (7%)
CICTCGA27/48 (56%)20/37 (54%)
MSK25/36 (69%)34/43 (79%)
FUBP1TCGA12/48 (35%)11/37 (29%)
MSK8/36 (22%)16/43 (37%)
NOTCH1TCGA4/48 (8%)14/37 (38%)
MSK2/36 (5%)14/43 (32%)
PIK3CATCGA10/48 (21%)6/37 (16%)
MSK3/36 (8%)14/43 (32%)
PIK3R1TCGA2/48 (4%)4/37 (11%)
MSK5/36 (14%)7/43 (16%)
CDK4amplPOLA1/483

TCGA: data extracted from Gliovis.

POLA: reference89.

CIC and FUBP Mutations

Up to 70% of IDH-mutant and 1p/19q-codeleted oligodendroglioma carry inactivating mutations in the homolog of the Drosophila capicua gene (CIC) on 19q13.2.110,111CIC inactivation has been shown to cooperate with mutant IDH in fostering increased production of 2-hydroxyglutatrate (2-HG), and has been linked to aberrant activation of MAPK/Ras signaling.112,113 Despite some studies reporting an association between CIC mutation or loss of protein expression and outcome, most studies in patients with grade 2 or 3 IDH-mutant and 1p/19q-codeleted oligodendroglioma found no prognostic association of CIC mutations.20,108,114,115 Although FUBP1 is frequently mutated in IDH-mutant and 1p/19q-codeleted oligodendroglioma, there is no clear association with survival.108 This is despite the mutation being subclonal (similar to CIC mutations) which suggests selection for FUBP1 mutant clones during tumor evolution.

Other Genetic Alterations

A few additional genetic alterations present in minor subsets of oligodendroglioma, IDH-mutant, and 1p/19q-codeleted each have been linked to unfavorable prognosis. These include mutations in PIK3CA and NOTCH1 (Table 4).20,116,117 The rare absence of TERT promoter mutations in IDH-mutant and 1p/19q-codeleted oligodendroglioma (occurring in < 5% of cases) has been associated with a worse prognosis in one study.118 In addition, increased MYC signaling has been found in a clinically more aggressive subgroup of IDHmt and 1p/19q-codeleted oligodendroglioma demonstrating an oligodendrocyte precursor-like gene expression signature, and MYC gain has been shown to be associated with the risk of developing a post-TMZ hypermutated phenotype.119,120PTEN alterations have been associated with shorter survival of patients with grade 2 IDHmt and 1p/19q-codeleted oligodendroglioma in one study.108

Chromosomal Instability and Tumor Mutational Burden in IDH-Mutant 1p/19q Codeleted Oligodendroglioma

Several retrospective studies reported that 1q and 19p polysomy is detectable in subsets of IDH-mutant and 1p/19q-codeleted oligodendroglioma and is associated with earlier recurrence and shorter survival.121 Chromosomal copy number variations in addition to 1p/19q codeletion increase significantly from grades 2 to 3 in IDH-mutant and 1p/19q-codeleted oligodendroglioma.20,122 In addition to a distinct DNA methylome profile, the recently reported prognostically unfavorable oligosarcoma also features increased chromosomal copy number variations.109 Apart from this rare subgroup, studies have reported that copy number burden was associated with a less favorable outcome in patients with IDH-mutant and 1p/19q-codeleted oligodendroglioma.20,123 In addition, a study based on TCGA data sets from 169 IDHmt and 1p/19q-codeleted oligodendroglioma patients revealed that high tumor mutational burden, defined by ≥ 0.69 mutations/megabase of DNA, was significantly associated with shorter survival.124 This study found a similar trend when using GLASS data sets from a small cohort of 25 IDHmt and 1p/19q-codeleted oligodendroglioma patients. Treatment with temozolomide has been associated with recurrences with high tumor mutational burden, high grade, and contrast-enhancing recurrences with poor prognosis.57,59

Gene Expression Profiles

Microarray-based mRNA expression profiling of 68 IDH-mutant and 1p/19q-codeleted oligodendroglioma treated with radio- +/− chemotherapy revealed an 8-gene signature (ST3GAL6, QPCT, NQO1, EPHX1, CST3, S100A8, CHI3L1, and OSBPL3) whose overexpression was significantly associated with shorter PFS.81,125 Another study reported on a prognostic 35-gene signature that identified high-risk and low-risk subgroups of 1p/19q codeleted glioma patients.126 A more recent study based on TCGA datasets from 137 oligodendroglioma patients and 2 independent validation cohorts of 218 patients reported on gene expression-based distinction of 2 prognostically distinct subtypes of oligodendroglioma, IDH-mutant and 1p/19q-codeleted.127 The prognostically unfavorable subtype displayed a proliferative phenotype with enrichment of histologically grade 3 tumors and higher mutation frequency in EGFR, MET, and NOTCH1. Using integrated analysis of the transcriptome, genome, and methylome data from 156 IDHmt and 1p/19q-codeleted oligodendroglioma patients, 3 subgroups were identified with distinct gene expression patterns corresponding to oligodendrocyte, oligodendrocyte precursor cell, and neuronal lineage cells.120 Among these, the oligodendrocyte precursor cell-like subgroup showed aberrant MYC activation and significantly worse outcomes independently of histological grade.

Methylation Analysis of IDH-Mutant Gliomas

Recent advances in glioma research underscore the pivotal role of epigenomic characteristics, particularly DNA methylation, allowing for refinement of the classification of gliomas, in particular in n IDH-mutant astrocytomas.128 Within the IDH-mutant astrocytoma tumor subgroup, 2 major methylation subgroups have been identified by genome-wide DNA methylation profiling: Glioma-CpG Island Methylator Phenotype (G-CIMP)-low and G-CIMP-high, with G-CIMP-low tumors displaying lower levels of genome-wide DNA-methylation levels than G-CIMP-high tumors.98 Patients with an initial diagnosis of G-CIMP-low tumors exhibited shorter overall survival compared to those with G-CIMP-high, accompanied by notable alterations in cell cycle pathways, CDKN2A/B deletions, and MET amplifications.90,98,104,129 The recently described LINE-1 methylation sequencing can be used as proxy for genome-wide DNA-methylation levels of a sample, and samples with low LINE-1 methylation levels are associated with grade 4 histology.29 Longitudinal analyses of paired samples showed that some G-CIMP-high tumors transitioned to G-CIMP-low at the time of tumor progression, indicating genome-wide DNA- demethylation.130,131 Tumors without malignant progression did not show such large differences, and a third methylation profile “G-CIMP-high at risk to low” subsequently identified patients with an intermediate prognosis.132,133 In parallel to these findings, a German group developed a classifier to aid the typing of brain tumors.134 This “Heidelberg methylation classifier” distinguishes an “Astrocytoma, IDH-mutant; high grade” class (A IDH HG) from an “Astrocytoma, IDH-mutant; lower grade” (A IDH) class, with different clinical outcomes.135 The prognostic significance of this distinction was confirmed in independent datasets, both in in grade 3 and in grades 2–4 astrocytomas.24,73 Multivariate analysis including grade, homozygous deletion of CDKN2A/B and methylation class showed that the latter 2 were independent prognostic factors, but not histological grade.73 Hypermethylation of a set of 7 HOX genes has been associated with survival in both IDH-mutant astrocytoma and IDH-mutant and 1p/19q codeleted oligodendroglioma, independent from CIMP status.104

MGMT promoter methylation is frequent in IDH-mutant glioma, and is present in nearly all oligodendrogliomas IDHmt and 1p/19q-codeleted.136 In grade 4 astrocytoma IDHmt, MGMT methylation status was associated with improved outcome but in the CATNON trial on anaplastic astrocytoma IDHmt, it failed to predict outcome to the addition of temozolomide to radiotherapy.137,138 In a study comparing radiotherapy to temozolomide chemotherapy in grade 2 gliomas MGMT status appeared to be associated with a longer PFS in astrocytoma IDHmt after temozolomide treatment, but whether MGMT status has an effect on survival in grade 2 astrocytoma IDHmt is unclear.139 In IDH-mutant low-grade gliomas, the MGMT methylation level in the tumor at first resection was associated with a hypermutational status at tumor recurrence after temozolomide treatment.140 To conclude, MGMT promoter assessment is not useful in oligodendroglioma IDHmt and 1p/19q codeleted, its clinical value in IDHmt astrocytoma remains to be demonstrated and is most likely less relevant than CIMP status.

IDH-mutant, 1p/19q codeleted oligodendrogliomas represent a distinct methylation class, and to date, methylation analysis with respect to tumor grade or along the disease course is not well understood. A recently identified and prognostic unfavorable methylation subclass “oligosarcoma” contains a high proportion of IDH-mutant and 1p19q codeleted oligodendrogliomas.109 Several of these “oligosarcomas” were recurrences of prior oligodendrogliomas which suggests this subclass represents malignant progression of oligodendrogliomas.

Overall, the analyses to date lead to biological insights that shed light on the role of global DNA methylation changes and demethylation in specific genes during glioma progression. While the utility of these classifiers and biomarkers in treatment strategies awaits further validation, their emergence marks a significant step towards personalized and risk-adapted approaches in the management of gliomas.

Clinical Experience With IDH Inhibitors in Grades 2 and 3 IDH-Mutant Glioma Patients

The importance of IDH mutations in gliomagenesis has made it a target of interest for the treatment of IDH-mutant gliomas.141,142 Multiple IDH inhibitors have been developed, some specific for mutant IDH1, while others inhibit both mutant IDH1 and IDH2. These inhibitors have been evaluated in several studies with heterogeneous populations of patients with IDH-mutant grades 2–4 gliomas, both in patients with enhancing and non-enhancing tumors (Table 5).4–6,143,144 All the initial studies of IDH inhibitors were conducted in patients with recurrent/progressive gliomas where the histology was usually based on prior resections rather than on tumor samples obtained immediately before study entry.5,6,143–146

Table 5.

Summary of the various clinical studies on IDH inhibitors, the objective response rate and the median progression-free survival in enhancing and unenhancing glioma.

StudyIDH inhibitorPatient population investigatedNumber of patients includedGlioma types/gradesORR and mPFS in enhancing gliomasORR and mPFS in non-enhancing gliomas
Mellinghoff et al133IvosidenibRecurrent IDH1-mutant gliomas66:
31 enhancing
35 non-enhancing
32 grade 2
18 grade 3
12 grade 4
0 (0%)
14 SD (45.2%)
mPFS 1.4 m
2.9% (1 PR)
30 SD (85.7%)
mPFS 13.6 m
Mellinghoff et al5VorasidenibRecurrent IDH1/2 mutant gliomas52:
30 enhancing
22 non-enhancing
25 grade 2
22 grade 3
4 grade 4
0 (0%)
17 SD (56.7%)
mPFS 3.0 m for grade 2; 3.7 m for grade 3
18%
1 PR (4.5%)
3 MR (13.6%)
16 SD (72.7%)
mPFS 19.6 m for grade 2; 40.8 m for grade 3
Mellinghoff et al131Vorasidenib and IvosidenibRecurrent, non-enhancing IDH1-mutant gliomas49, all non-enhancing43 grade 2
6 grade 3
N/AVorasidenib 50 mg qd: 42.9% (2 PR, 4 MR); SD 42.9%
Vorasidenib 10 mg qd: 10% (1 MR); SD 80%
Ivosidenib 500 mg qd: 35.7% (3 PR, 2 MR); SD 64.3%
Ivosidenib 250 mg BID: 12.5% (1 MR); SD 62.5%
Mellinghoff et al7VorasidenibResidual or recurrent non-enhancing gliomas168, all non-enhancingGrade 2N/A
Natsume et al6DS-1001Recurrent IDH1-mutant gliomas47:
35 enhancing
12 non-enhancing
16 grade 2
22 grade 3
9 grade 4
17.1% (2 CR, 4 PR)
11 SD (31.4%)
mPFS 10.4 weeks
33.3% (1 PR, 3 MR)
7 SD (58.3%)
mPFS not reached
De la Fuente et al132OlutasidenibRecurrent IDH1-mutant gliomas26:
23 enhancing
3 non-enhancing
4 grade 2
15 grade 3
7 grade 4
8.7% (2 PR)
10 SD (38.5%) in the entire cohort
0%
10 SD (38.5%) in the entire cohort
Wick et al134BAY1436032Recurrent IDH1-mutant gliomas49:
35 LGG, 33 of whom had measurable enhancing disease
14 enhancing grade 4 astrocytoma
LGG: 6.1% (1 CR, 1 PR)
15 SD (42.9%)
Grade 4 astrocytoma: 0%
4 SD (29%)
LGG: 100% (2 PR)
StudyIDH inhibitorPatient population investigatedNumber of patients includedGlioma types/gradesORR and mPFS in enhancing gliomasORR and mPFS in non-enhancing gliomas
Mellinghoff et al133IvosidenibRecurrent IDH1-mutant gliomas66:
31 enhancing
35 non-enhancing
32 grade 2
18 grade 3
12 grade 4
0 (0%)
14 SD (45.2%)
mPFS 1.4 m
2.9% (1 PR)
30 SD (85.7%)
mPFS 13.6 m
Mellinghoff et al5VorasidenibRecurrent IDH1/2 mutant gliomas52:
30 enhancing
22 non-enhancing
25 grade 2
22 grade 3
4 grade 4
0 (0%)
17 SD (56.7%)
mPFS 3.0 m for grade 2; 3.7 m for grade 3
18%
1 PR (4.5%)
3 MR (13.6%)
16 SD (72.7%)
mPFS 19.6 m for grade 2; 40.8 m for grade 3
Mellinghoff et al131Vorasidenib and IvosidenibRecurrent, non-enhancing IDH1-mutant gliomas49, all non-enhancing43 grade 2
6 grade 3
N/AVorasidenib 50 mg qd: 42.9% (2 PR, 4 MR); SD 42.9%
Vorasidenib 10 mg qd: 10% (1 MR); SD 80%
Ivosidenib 500 mg qd: 35.7% (3 PR, 2 MR); SD 64.3%
Ivosidenib 250 mg BID: 12.5% (1 MR); SD 62.5%
Mellinghoff et al7VorasidenibResidual or recurrent non-enhancing gliomas168, all non-enhancingGrade 2N/A
Natsume et al6DS-1001Recurrent IDH1-mutant gliomas47:
35 enhancing
12 non-enhancing
16 grade 2
22 grade 3
9 grade 4
17.1% (2 CR, 4 PR)
11 SD (31.4%)
mPFS 10.4 weeks
33.3% (1 PR, 3 MR)
7 SD (58.3%)
mPFS not reached
De la Fuente et al132OlutasidenibRecurrent IDH1-mutant gliomas26:
23 enhancing
3 non-enhancing
4 grade 2
15 grade 3
7 grade 4
8.7% (2 PR)
10 SD (38.5%) in the entire cohort
0%
10 SD (38.5%) in the entire cohort
Wick et al134BAY1436032Recurrent IDH1-mutant gliomas49:
35 LGG, 33 of whom had measurable enhancing disease
14 enhancing grade 4 astrocytoma
LGG: 6.1% (1 CR, 1 PR)
15 SD (42.9%)
Grade 4 astrocytoma: 0%
4 SD (29%)
LGG: 100% (2 PR)

IDH, isocitrate dehydrogenase; LGG, low-grade glioma; mPFS, median progression-free survival; MR, minor response; ORR, objective response rate; PR, partial response; SD, stable disease.

Table 5.

Summary of the various clinical studies on IDH inhibitors, the objective response rate and the median progression-free survival in enhancing and unenhancing glioma.

StudyIDH inhibitorPatient population investigatedNumber of patients includedGlioma types/gradesORR and mPFS in enhancing gliomasORR and mPFS in non-enhancing gliomas
Mellinghoff et al133IvosidenibRecurrent IDH1-mutant gliomas66:
31 enhancing
35 non-enhancing
32 grade 2
18 grade 3
12 grade 4
0 (0%)
14 SD (45.2%)
mPFS 1.4 m
2.9% (1 PR)
30 SD (85.7%)
mPFS 13.6 m
Mellinghoff et al5VorasidenibRecurrent IDH1/2 mutant gliomas52:
30 enhancing
22 non-enhancing
25 grade 2
22 grade 3
4 grade 4
0 (0%)
17 SD (56.7%)
mPFS 3.0 m for grade 2; 3.7 m for grade 3
18%
1 PR (4.5%)
3 MR (13.6%)
16 SD (72.7%)
mPFS 19.6 m for grade 2; 40.8 m for grade 3
Mellinghoff et al131Vorasidenib and IvosidenibRecurrent, non-enhancing IDH1-mutant gliomas49, all non-enhancing43 grade 2
6 grade 3
N/AVorasidenib 50 mg qd: 42.9% (2 PR, 4 MR); SD 42.9%
Vorasidenib 10 mg qd: 10% (1 MR); SD 80%
Ivosidenib 500 mg qd: 35.7% (3 PR, 2 MR); SD 64.3%
Ivosidenib 250 mg BID: 12.5% (1 MR); SD 62.5%
Mellinghoff et al7VorasidenibResidual or recurrent non-enhancing gliomas168, all non-enhancingGrade 2N/A
Natsume et al6DS-1001Recurrent IDH1-mutant gliomas47:
35 enhancing
12 non-enhancing
16 grade 2
22 grade 3
9 grade 4
17.1% (2 CR, 4 PR)
11 SD (31.4%)
mPFS 10.4 weeks
33.3% (1 PR, 3 MR)
7 SD (58.3%)
mPFS not reached
De la Fuente et al132OlutasidenibRecurrent IDH1-mutant gliomas26:
23 enhancing
3 non-enhancing
4 grade 2
15 grade 3
7 grade 4
8.7% (2 PR)
10 SD (38.5%) in the entire cohort
0%
10 SD (38.5%) in the entire cohort
Wick et al134BAY1436032Recurrent IDH1-mutant gliomas49:
35 LGG, 33 of whom had measurable enhancing disease
14 enhancing grade 4 astrocytoma
LGG: 6.1% (1 CR, 1 PR)
15 SD (42.9%)
Grade 4 astrocytoma: 0%
4 SD (29%)
LGG: 100% (2 PR)
StudyIDH inhibitorPatient population investigatedNumber of patients includedGlioma types/gradesORR and mPFS in enhancing gliomasORR and mPFS in non-enhancing gliomas
Mellinghoff et al133IvosidenibRecurrent IDH1-mutant gliomas66:
31 enhancing
35 non-enhancing
32 grade 2
18 grade 3
12 grade 4
0 (0%)
14 SD (45.2%)
mPFS 1.4 m
2.9% (1 PR)
30 SD (85.7%)
mPFS 13.6 m
Mellinghoff et al5VorasidenibRecurrent IDH1/2 mutant gliomas52:
30 enhancing
22 non-enhancing
25 grade 2
22 grade 3
4 grade 4
0 (0%)
17 SD (56.7%)
mPFS 3.0 m for grade 2; 3.7 m for grade 3
18%
1 PR (4.5%)
3 MR (13.6%)
16 SD (72.7%)
mPFS 19.6 m for grade 2; 40.8 m for grade 3
Mellinghoff et al131Vorasidenib and IvosidenibRecurrent, non-enhancing IDH1-mutant gliomas49, all non-enhancing43 grade 2
6 grade 3
N/AVorasidenib 50 mg qd: 42.9% (2 PR, 4 MR); SD 42.9%
Vorasidenib 10 mg qd: 10% (1 MR); SD 80%
Ivosidenib 500 mg qd: 35.7% (3 PR, 2 MR); SD 64.3%
Ivosidenib 250 mg BID: 12.5% (1 MR); SD 62.5%
Mellinghoff et al7VorasidenibResidual or recurrent non-enhancing gliomas168, all non-enhancingGrade 2N/A
Natsume et al6DS-1001Recurrent IDH1-mutant gliomas47:
35 enhancing
12 non-enhancing
16 grade 2
22 grade 3
9 grade 4
17.1% (2 CR, 4 PR)
11 SD (31.4%)
mPFS 10.4 weeks
33.3% (1 PR, 3 MR)
7 SD (58.3%)
mPFS not reached
De la Fuente et al132OlutasidenibRecurrent IDH1-mutant gliomas26:
23 enhancing
3 non-enhancing
4 grade 2
15 grade 3
7 grade 4
8.7% (2 PR)
10 SD (38.5%) in the entire cohort
0%
10 SD (38.5%) in the entire cohort
Wick et al134BAY1436032Recurrent IDH1-mutant gliomas49:
35 LGG, 33 of whom had measurable enhancing disease
14 enhancing grade 4 astrocytoma
LGG: 6.1% (1 CR, 1 PR)
15 SD (42.9%)
Grade 4 astrocytoma: 0%
4 SD (29%)
LGG: 100% (2 PR)

IDH, isocitrate dehydrogenase; LGG, low-grade glioma; mPFS, median progression-free survival; MR, minor response; ORR, objective response rate; PR, partial response; SD, stable disease.

Ivosidenib, the first-in-class IDH 1 inhibitor, was evaluated in a multicenter, open-label, phase 1 study in 66 patients with recurrent IDH1-mutant gliomas, 32 of whom had grade 2 gliomas, 18 had grade 3 gliomas, and 12 had grade 4 gliomas.145 Only one partial response was observed, in a patient with grade 3 a non-enhancing tumor. Stable disease as the best response was seen in 85.7% of patients with non-enhancing tumors compared to 45.2% with enhancing tumors. Disease progression was more prevalent in enhancing tumors (54.8% vs. 11.4%), and median PFS was longer in non-enhancing tumors (13.6 months) versus enhancing tumors (1.4 months). More recent analysis of data from the ivosidenib trial by tumor grade showed no objective responses, neither in enhancing nor in non-enhancing tumors.4 For patients treated at the dose expansion cohort with non-enhancing tumors the median PFS was 19.4 months for grade 2 tumors (18 patients) and 23 months for grade 3 gliomas (4 patients; data on file). These data suggest that some patients with recurrent grade 3 non-enhancing gliomas may benefit from ivosidenib.

Another phase 1 study evaluated the brain-penetrant IDH1/2 inhibitor vorasidenib in recurrent IDH1/2-mutant gliomas. Seventeen of the twenty-five grade 2 gliomas, 5 of the 22 grade 3 gliomas, and none of the 4 grade 4 gliomas were non-enhancing (data on file).5 The objective response rate (ORR) was 18% in non-enhancing gliomas (1 partial response and 3 minor responses), and 3.3% in enhancing gliomas (1 patient with a grade 3 tumor). The ORR was 8% for grade 2 gliomas and 13.6% for grade 3 gliomas. Median PFS was again longer in patients with non-enhancing tumors (36.8 vs. 3.6 months). Of the 22 non-enhancing tumors, there were 17 grade 2, 5 grade 3, and no grade 4. Median PFS was 19.6 months for grade 2 tumors and 40.8 months for grade 3 tumors. There were 30 patients with enhancing tumors, 8 grade 2, 17 grade 3, and 4 grade 4 (1 grade unknown). Median PFS was 3.0 months for grade 2, 3.7 months for grade 3, and 1.1 months for grade 4 gliomas. These data, while limited, suggest that the response to vorasidenib is not substantially different between grades 2 and 3 gliomas, especially when they are non-enhancing.

Ivosidenib and vorasidenib were both evaluated in a surgical window-of-opportunity trial in patients with recurrent non-enhancing grade 2 (43 patients) and 3 (6 patients) IDH1-mt gliomas.143 Two doses of vorasidenib and ivosidenib were used. Five patients had a partial response, and 8 patients had a minor response. Most responders had grade 2 tumors, although one patient with grade 3 astrocytoma had a partial response. Stable disease was documented in 31 patients. Vorasidenib was associated with a slightly tighter reduction in intra-tumoral 2-HG levels and was selected for evaluation in the phase 3 placebo-controlled INDIGO study. This trial enrolled patients with grade 2 IDH-mutant tumors with measurable non-enhancing disease, not in immediate need of chemotherapy/radiation, which study found that vorasidenib led to significant improvement in PFS and time-to-next intervention compared to placebo.7

A multicenter, open-label, dose-escalation, phase 1 study evaluated the IDH1 inhibitor DS-1001 (safusidenib, AB-218) in patients with recurrent IDH-mutant gliomas of any grade.6 Of the 45 patients, 4 had grade 2 oligodendroglioma, 11 had grade 3 oligodendroglioma, 12 had grade 2 astrocytoma, 11 had grade 3 astrocytoma, and 7 had grade 4 astrocytoma. Thirty-five patients had enhancing tumors and 12 had non-enhancing tumors. ORR was 33.3% in non-enhancing tumors and 17.1% in enhancing gliomas.6 A complete response was observed in an enhancing grade 4 astrocytoma and an enhancing grade 3 oligodendroglioma, and partial responses were observed in enhancing astrocytomas (2 of the 21) and oligodendrogliomas (2 of the 9). Stable disease was reported in 52.3% of enhancing astrocytomas and 22.2% of enhancing oligodendrogliomas. Median PFS was longer in non-enhancing tumors (not reached) versus 10.4 weeks in enhancing tumors. Although the paper did not breakdown the data by tumor grade, since the majority of patients had grade 3 tumors, DS-1001 (safusidenib) appears to be active for both enhancing and non-enhancing grade 2 and 3 IDH-mutant gliomas.

Another multicenter phase 1 study evaluated BAY1436032 in 29 patients with recurrent IDH-1 mutant gliomas.146 There was a complete response in one grade 3 astrocytoma patient, and a partial response in a grade 3 astrocytoma and 2 grade 3 oligodendroglioma patients. In this study, 33 of the 35 patients with grades 2 and 3 glioma had enhancing disease, among whom 1 had a complete response, and another had a partial response, while 2 had stable disease. Twenty-nine percent of grade 4 astrocytoma patients had stable disease, but no responses were observed among these patients. PFS rate at 3 months was higher in grades 2 or 3 IDH-mutant gliomas compared to grade 4 IDH-mutant astrocytomas patients (31% vs. 22%).

Finally, the IDH1 inhibitor olutasidenib was evaluated in a phase Ib/II study, in 26 patients with recurrent IDH-1 mutant gliomas of whom 88% had enhancing tumors.144 Fifty-eight percent of the patients had a grade 3 glioma, and 27% had a grade 4 astrocytoma. A partial response was observed in 2 patients, who both had enhancing gliomas (1 grade 3 and 1 grade 4). Upon central assessment of response, 4 partial responses were reported, and 5 patients had a reduction in tumor size that did not reach 50%. PFS rate at 6 months was 23% in enhancing gliomas.

All patients with enhancing and higher-grade gliomas included in the above studies had recurrent diseases and usually had received multiple lines of treatment prior to enrollment. Prior radiation was given to 73% to 100%, and prior chemotherapy to 75% to 88% of the enrolled patients, with most patients having received more than 2 prior lines of therapy.5,6,144–146 At advanced stages, tumors can acquire additional genomic alterations that can bypass the need for the mutant IDH enzyme to maintain gliomagenesis and the IDH mutation can even be lost due to copy number alterations.97,142 Despite this, a number of patients with grade 3 IDH-mutant gliomas appear to have benefited from therapy with IDH inhibitors, challenging the perception that IDH inhibitors would be efficacious only in grade 2 non-enhancing IDH-mutant gliomas. It is possible that the benefit of IDH inhibitors will be even greater in newly diagnosed tumors grade 3 IDH-mutant gliomas where activation of alternate molecular drivers may be less than in recurrent disease.

Conclusions

While some series show an association between tumor grade and outcome others fail to do so, and many patients with a grade 3 IDH-mutant astrocytoma or IDH-mutant and 1p/19q codeleted oligodendroglioma experience a survival well beyond 10 years. The wide overlap in the survival range of grades 2 and 3 IDH-mutant glioma patients appears in part due to the subjective nature of the histological grading of IDH-mutant tumors: There are no sharp and objective criteria distinguishing grade 2 form grade 3 tumors. Nonetheless, many factors associated with the outcome of patients with IDH-mutant glioma (enhancement on imaging, molecular findings) show some association with histological grade. Still, the overall limited association of outcome of patients with grade 2 versus 3 3 IDH-mutant glioma patients reflects the biological continuum of these tumor grades implying that a sharp distinction between these grades is artificial.

Genetic analysis analyses may allow for better prognostication of IDH-mutant glioma patients than histological grading and mitotic counts. An update of the WHO classification especially for IDHmt astrocytomas is needed. Apart from homozygous deletion of CDKN2A/B several other well-defined alterations have been associated with poor outcome. In contrast, there are currently no well-validated prognostic molecular markers in IDH-mutant and 1p/19q-codeleted oligodendroglioma patients. Similar to mutational analysis, genome-wide methylation analysis holds promise for risk stratification. Several studies have shown that specific methylation patterns are associated with outcome, may allow risk stratification within tumor grades, and may be more associated with outcome than tumor grade.

The modest association between age and outcome of patients with IDH-mutant glioma does not warrant the use of guidelines of a strict age criterion of 40 years for the identification of patients at risk for a poor outcome. The available evidence shows that a cutoff in the range of 50-60 years is more appropriate for that purpose. Importantly, most series show that the prognostic effect of clinical factors decreases once the extent of resection and in particular the quantitative postoperative tumor volume are considered. This is in particular true for patients with IDH-mutant astrocytoma, in whom postoperative residual tumor has a major association with survival.

Although the presence of contrast enhancement on MR imaging is associated with tumor grade in IDH-mutant gliomas, this finding is neither sensitive or specific, and a substantial percentage of grade 2 oligodendrogliomas and IDH-mutant astrocytomas will demonstrate some level of enhancement. Contrast enhancement at the time of progression of lower-grade IDH-mutant gliomas suggests transformation towards a higher grade of malignancy, which carries a different clinical significance compared to the presence of contrast enhancement at first diagnosis. For prognostication, spontaneous TGR may help in predicting the clinical behavior of IDH-mutant glioma. The evaluation of changes in tumor growth curves may be a better way of identifying patients with non-enhancing tumors that benefit from medical treatments than classical response assessment using RANO criteria, and needs further confirmatory studies. Clinical trials with IDH inhibitors have shown some activity in recurrent tumors, in enhancing tumors, and in grade 3 tumors. From a biological perspective, there is no a priori reason why newly diagnosed grade 3 tumors cannot be responsive to IDH inhibitors and other clinical, radiological, and molecular factors need consideration (like growth rate, histology, other molecular findings, and pre-and postoperative tumor volume). Real-world studies should prospectively collect data to provide guidance for future patient counseling.

Acknowledgments

This invited review was prepared without external support. All authors have contributed equally to this manuscript.

Conflict of interest statement

MvdB has received honoraria for consultancy from Anheart Therapeutics, Boehringer Ingelheim, Fore Biotherapeutics, Genenta, Incyte, Mundipharm, Chimerix, Roche, and Servier and support for travel to meetings by Servier. PJF is providing consultancy to Servier. JCT has received honoraria for consultancy, lecture or research grants from the following for-profit companies: Novocure, Novartis, Munich Surgical imaging, Seagen. DB has nothing to declare. MT received honoraria for lectures, consultation or advisory board participation from Medscape, Novocure, Ono, Servier. BME is a Paid Consultant and Advisor Alpheus Medical, Chimerix, Erasca, Global Coalition for Adaptive Research, Imaging Endpoints, Medicenna, Voiant, Monteris, Neosoma, Orbus Therapeutics, Sagimet Biosciences, Sapience Therapeutics, Servier Pharmaceuticals, SonALAsense, and Sumitomo Dianippon Pharma Oncology. BME also has a research grant from Siemens and is a paid member of the data monitoring board for Carthera. RJY: Is a consultant for NordicNeuroImaging, Turing Medical, Olea Medical, ICON plc, RadMD. JP has nothing to declare. AvD: Shareholder and cofounder of Heidelberg Epignostix GmbH, honoraria from Illumina. FS is shareholder and cofounder of Heidelberg Epignostix GmbH, honoraria from Illumina.

DFB has nothing to declare. RYH has consulting/advisory board activities for Vysioneer and Nuvation Bio. MW has received research grants from Novartis, Quercis and Versameb, and honoraria for lectures or advisory board participation or consulting from Anheart Therapeutics, Bayer, Curevac, Medac, Neurosense, Novartis, Novocure, Orbus, Pfizer, Philogen, Roche and Servier. IKM has received research funding from General Electric, Agios, and Lilly and honoraria from Roche for a presentation. IKM has served in advisory roles for Agios, Amgen, Debiopharm, Novartis, Puma Biotechnology, and Voyager Therapeutics. TFC is cofounder, major stock holder, consultant and board member of Katmai. Pharmaceuticals, holds stock for Erasca, member of the board and paid consultant for the 501c3 Global Coalition for Adaptive Research, holds stock in Chimerix and receives milestone payments and possible future royalties, member of the scientific advisory board for Break Through Cancer, member of the scientific advisory board for Cure Brain Cancer Foundation, has provided paid consulting services to Symbio, Mundipharma, Tango BlueRock, Vida Ventures, Lista Therapeutics, Stemline, Novartis, Roche, Sonalasense, Sagimet, Clinical Care Options, Ideology Health, Servier, Jubilant, Immvira, Gan & Lee, BrainStorm, Katmai, Sapience, Inovio, Vigeo Therapeutics, DNATrix, Tyme, SDP, Kintara, Bayer, Merck, Boehinger Ingelheim, VBL, Amgen, Kiyatec, Bayer, Abbvie, VBI, Deciphera, VBL, Agios, Novocure, Medscape and has contracts with UCLA for the Brain Tumor Program with Roche, VBI, Merck, Novartis, BMS. The Regents of the University of California (T.F.C. employer) has licensed intellectual property co-invented by TFC to Katmai Pharmaceuticals. JTH has nothing to disclose. KMA has nothing to disclose. GR has nothing to disclose. GY has nothing to disclose. PK has nothing to disclose. HN has nothing to disclose. KBP has received research support: Biomimetix, LLC, Servier, Ono Pharmaceutical. Advisory Board/Consultant: Anheart, Rigel, Sapience, Servier, Telix, Varian. PYW has received research Support from Astra Zeneca, Black Diamond, Bristol Meyers Squibb, Chimerix, Eli Lily, Erasca, Global Coalition For Adaptive Research, Kazia, MediciNova, Merck, Novartis, Quadriga, Servier, VBI Vaccines. Advisory Board/Consultant: Anheart, Astra Zeneca, Black Diamond, Celularity, Chimerix, Day One Bio, Genenta, Glaxo Smith Kline, Kintara, Merck, Mundipharma, Novartis, Novocure, Prelude Therapeutics, Sagimet, Sapience, Servier, Symbio, Tango, Telix, VBI Vaccines. FD has received honoraria for consultancy and lectures from Servier and Novocure. MP has received honoraria for lectures, consultation or advisory board participation from the following for-profit companies: Bayer, Bristol-Myers Squibb, Novartis, Gerson Lehrman Group (GLG), CMC Contrast, GlaxoSmithKline, Mundipharma, Roche, BMJ Journals, MedMedia, Astra Zeneca, AbbVie, Lilly, Medahead, Daiichi Sankyo, Sanofi, Merck Sharp & Dome, Tocagen, Adastra, Gan & Lee Pharmaceuticals, Janssen, Servier, Miltenyi, Böhringer-Ingelheim, Telix, Medscape.

References

1.

Weller
M
,
van den Bent
M
,
Preusser
M
, et al. .
EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood
.
Nat Rev Clin Oncol.
2020
;
18
(
3
):
170
186
.

2.

Buckner
JC
,
Shaw
EG
,
Pugh
SL
, et al. .
Radiation plus Procarbazine, CCNU, and Vincristine in Low-Grade Glioma
.
N Engl J Med.
2016
;
374
(
14
):
1344
1355
.

3.

Pignatti
F
,
van den Bent
MJ
,
Curran
D
, et al. ;
European Organization for Research and Treatment of Cancer Brain Tumor Cooperative Group
.
Prognostic factors for survival in adult patients with cerebral low-grade glioma
.
J Clin Oncol.
2002
;
20
(
8
):
2076
2084
.

4.

Mellinghoff
IK
,
Ellingson
BM
,
Touat
M
, et al. .
Ivosidenib in IDH1-mutated advanced glioma
.
J Clin Oncol.
2020
;
38
(
29
):
3398
3406
.

5.

Mellinghoff
IK
,
Penas-Prado
M
,
Peters
KB
, et al. .
Vorasidenib, 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
(
16
):
4491
4499
.

6.

Natsume
A
,
Arakawa
Y
,
Narita
Y
, et al. .
The 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
(
2
):
326
336
.

7.

Mellinghoff
IK
,
van den Bent
MJ
,
Blumenthal
DT
, et al. ;
INDIGO Trial Investigators
.
Vorasidenib in IDH1- or IDH2-Mutant Low-Grade Glioma
.
N Engl J Med.
2023
;
389
(
7
):
589
601
.

8.

Pallud
J
,
Mandonnet
E
,
Duffau
H
, et al. .
Prognostic value of initial magnetic resonance imaging growth rates for World Health Organization grade II gliomas
.
Ann Neurol.
2006
;
60
(
3
):
380
383
.

9.

Brasil
CG
,
Ciccarelli
O
,
Altmann
DR
, et al. .
Low-grade gliomas: six-month tumor growth predicts patient outcome better than admission tumor volume, relative cerebral blood volume, and apparent diffusion coefficient
.
Radiology.
2009
;
253
(
2
):
505
512
.

10.

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
(
6
):
803
820
.

11.

Lassman
AB
,
Hoang-Xuan
K
,
Polley
MC
, et al. .
Joint Final Report of EORTC 26951 and RTOG 9402: Phase III Trials with Procarbazine, Lomustine, and Vincristine Chemotherapy for Anaplastic Oligodendroglial Tumors
.
J Clin Oncol.
2022
;
40
(
23
):
2539
2545
.

12.

Bell
EH
,
Zhang
P
,
Shaw
EG
, et al. .
Comprehensive Genomic Analysis in NRG Oncology/RTOG 9802: A Phase III Trial of radiation versus radiation plus procarbazine, lomustine (CCNU), and vincristine in high-risk low-grade glioma
.
J Clin Oncol.
2020
;
38
(
29
):
3407
3417
.

13.

Donahue
B
,
Scott
CB
,
Nelson
JS
, et al. .
Influence of an oligodendroglial component on the survival of patients with anaplastic astrocytomas: A report of radiation therapy oncology group 83-02
.
Int J Radiat Oncol Biol Phys.
1997
;
38
(
5
):
911
914
.

14.

Chang
S
,
Zhang
P
,
Cairncross
JG
, et al. .
Phase III randomized study of radiation and temozolomide versus radiation and nitrosourea therapy for anaplastic astrocytoma: results of NRG Oncology RTOG 9813
.
Neuro Oncol
.
2017
;
19
(
2
):
252
258
.

15.

van den Bent
MJ
,
Tesileanu
CMS
,
Wick
W
, et al. .
Adjuvant and concurrent temozolomide for 1p/19q non-co-deleted anaplastic glioma (CATNON; EORTC study 26053-22054): Second interim analysis of a randomised, open-label, phase 3 study
.
Lancet Oncol.
2021
;
22
(
6
):
813
823
.

16.

Reuss
DE
,
Mamatjan
Y
,
Schrimpf
D
, et al. .
IDH mutant diffuse and anaplastic astrocytomas have similar age at presentation and little difference in survival: a grading problem for WHO
.
Acta Neuropathol.
2015
;
129
(
6
):
867
873
.

17.

Minniti
G
,
Paolini
S
,
Antonelli
M
, et al. .
Long-term treatment outcomes of temozolomide-based chemoradiation in patients with adult-type diffuse IDH-mutant grade 2 astrocytoma
.
J Neurooncol.
2023
;
164
(
2
):
331
339
.

18.

Carstam
L
,
Corell
A
,
Smits
A
, et al. .
WHO grade loses its prognostic value in molecularly defined diffuse lower-grade gliomas
.
Front Oncol.
2021
;
11
:
803975
.

19.

Carstam
L
,
Latini
F
,
Solheim
O
, et al. .
Long-term follow up of patients with WHO grade 2 oligodendroglioma
.
J Neurooncol.
2023
;
164
(
1
):
65
74
.

20.

Aoki
K
,
Nakamura
H
,
Suzuki
H
, et al. .
Prognostic relevance of genetic alterations in diffuse lower-grade gliomas
.
Neuro Oncol
.
2018
;
20
(
1
):
66
77
.

21.

Mair
MJ
,
Leibetseder
A
,
Heller
G
, et al. .
Early postoperative treatment versus initial observation in CNS WHO Grade 2 and 3 Oligodendroglioma: Clinical Outcomes and DNA Methylation Patterns
.
Clin Cancer Res.
2022
;
28
(
20
):
4565
4573
.

22.

Paľa
A
,
Coburger
J
,
Scherer
M
, et al. .
To treat or not to treat? A retrospective multicenter assessment of survival in patients with IDH-mutant low-grade glioma based on adjuvant treatment
.
J Neurosurg.
2019
;
133
(
2
):
273
280
.

23.

Dao Trong
P
,
Gluszak
M
,
Reuss
D
, et al. .
Isocitrate-dehydrogenase-mutant lower grade glioma in elderly patients: Treatment and outcome in a molecularly characterized contemporary cohort
.
J Neurooncol.
2023
;
161
(
3
):
605
615
.

24.

Tesileanu
CMS
,
van den Bent
MJ
,
Sanson
M
, et al. .
Prognostic significance of genome-wide DNA methylation profiles within the randomised, phase 3, EORTC CATNON trial on non-1p/19q deleted anaplastic glioma
.
Neuro Oncol
.
2021
;
23
(
9
):
1547
1559
.

25.

Wijnenga
MMJ
,
French
PJ
,
Dubbink
HJ
, et al. .
The impact of surgery in molecularly defined low-grade glioma: An integrated clinical, radiological, and molecular analysis
.
Neuro Oncol
.
2018
;
20
(
1
):
103
112
.

26.

Park
YW
,
Kim
S
,
Han
K
, et al. .
Rethinking extent of resection of contrast-enhancing and non-enhancing tumor: Different survival impacts on adult-type diffuse gliomas in 2021 World Health Organization classification
.
Eur Radiol.
2023
;
34
(
2
):
1376
1387
.

27.

Tran
S
,
Thomas
A
,
Aliouat
I
, et al. .
A threshold for mitotic activity and post-surgical residual volume defines distinct prognostic groups for astrocytoma IDH-mutant
.
Neuropathol Appl Neurobiol.
2023
;
49
(
4
):
e12928
.

28.

Appay
R
,
Dehais
C
,
Maurage
CA
, et al. ;
POLA Network
.
CDKN2A homozygous deletion is a strong adverse prognosis factor in diffuse malignant IDH-mutant gliomas
.
Neuro. Oncol
.
2019
;
21
(
12
):
1519
1528
.

29.

Weller
M
,
Felsberg
J
,
Hentschel
B
, et al. .
Improved prognostic stratification of patients with isocitrate dehydrogenase-mutant astrocytoma
.
Acta Neuropathol.
2024
;
147
(
1
):
11
.

30.

Olar
A
,
Wani
KM
,
Alfaro-Munoz
KD
, et al. .
IDH mutation status and role of WHO grade and mitotic index in overall survival in grade II-III diffuse gliomas
.
Acta Neuropathol.
2015
;
129
(
4
):
585
596
.

31.

Montégut
C
,
Guillamo
JS
,
Ducray
F
, et al. .
Characteristics, patterns of care and predictive geriatric factors in elderly patients treated for high-grade idh-mutant gliomas: A french POLA Network Study
.
Cancers (Basel)
.
2022
;
14
(
22
):
5509
.

32.

Hervey-Jumper
SL
,
Zhang
Y
,
Phillips
JJ
, et al. .
Interactive effects of molecular, therapeutic, and patient factors on outcome of diffuse low-grade glioma
.
J Clin Oncol.
2023
;
41
(
11
):
2029
2042
.

33.

Weller
J
,
Katzendobler
S
,
Blobner
J
, et al. .
Limited efficacy of temozolomide alone for astrocytoma, IDH-mutant, CNS WHO grades 2 or 3
.
J Neurooncol.
2022
;
160
(
1
):
149
158
.

34.

Darlix
A
,
Rigau
V
,
Fraisse
J
, et al. .
Postoperative follow-up for selected diffuse low-grade gliomas with WHO grade III/IV foci
.
Neurology.
2020
;
94
(
8
):
e830
e841
.

35.

Kavouridis
VK
,
Boaro
A
,
Dorr
J
, et al. .
Contemporary assessment of extent of resection in molecularly defined categories of diffuse low-grade glioma: A volumetric analysis
.
J Neurosurg.
2019
;
133
(
5
):
1291
1301
.

36.

Karschnia
P
,
Vogelbaum
MA
,
van den Bent
M
, et al. .
Evidence-based recommendations on categories for extent of resection in diffuse glioma
.
Eur J Cancer.
2021
;
149
:
23
33
.

37.

Kawaguchi
T
,
Sonoda
Y
,
Shibahara
I
, et al. .
Impact of gross total resection in patients with WHO grade III glioma harboring the IDH 1/2 mutation without the 1p/19q co-deletion
.
J Neurooncol.
2016
;
129
(
3
):
505
514
.

38.

Molinaro
AM
,
Hervey-Jumper
S
,
Morshed
RA
, et al. .
Association of maximal extent of resection of contrast-enhanced and non-contrast-enhanced tumor with survival within molecular subgroups of patients with newly diagnosed glioblastoma
.
JAMA Oncol
.
2020
;
6
(
4
):
495
503
.

39.

Duffau
H.
Higher-order surgical questions for diffuse low-grade gliomas: Supramaximal resection, neuroplasticity, and screening
.
Neurosurg Clin N Am.
2019
;
30
(
1
):
119
128
.

40.

Cahill
DP
,
Dunn
GP.
Considering the extent of resection in diffuse glioma
.
Neuro Oncol
.
2023
;
25
(
12
):
2134
2135
.

41.

Beiko
J
,
Suki
D
,
Hess
KR
, et al. .
IDH1 mutant malignant astrocytomas are more amenable to surgical resection and have a survival benefit associated with maximal surgical resection
.
Neuro Oncol
.
2014
;
16
(
1
):
81
91
.

42.

Bush
NAO
,
Young
JS
,
Zhang
Y
, et al. .
A single institution retrospective analysis on survival based on treatment paradigms for patients with anaplastic oligodendroglioma
.
J Neurooncol.
2021
;
153
(
3
):
447
454
.

43.

Suchorska
B
,
Schuller
U
,
Biczok
A
, et al. .
Contrast enhancement is a prognostic factor in IDH1/2 mutant, but not in wild-type WHO grade II/III glioma as confirmed by machine learning
.
Eur J Cancer.
2019
;
107
:
15
27
.

44.

Lasocki
A
,
Buckland
ME
,
Molinaro
T
, et al. .
Correlating MRI features with additional genetic markers and patient survival in histological grade 2-3 IDH-mutant astrocytomas
.
Neuroradiology.
2023
;
65
(
8
):
1215
1223
.

45.

Castet
F
,
Alanya
E
,
Vidal
N
, et al. .
Contrast-enhancement in supratentorial low-grade gliomas: A classic prognostic factor in the molecular age
.
J Neurooncol.
2019
;
143
(
3
):
515
523
.

46.

Wang
YY
,
Wang
K
,
Li
SW
, et al. .
Patterns of tumor contrast enhancement predict the prognosis of anaplastic gliomas with IDH1 Mutation
.
AJNR Am J Neuroradiol.
2015
;
36
(
11
):
2023
2029
.

47.

Joyner
DA
,
Garrett
J
,
Batchala
PP
, et al. .
MRI features predict tumor grade in isocitrate dehydrogenase (IDH)-mutant astrocytoma and oligodendroglioma
.
Neuroradiology.
2023
;
65
(
1
):
121
129
.

48.

Lin
Y
,
Xing
Z
,
She
D
, et al. .
IDH mutant and 1p/19q co-deleted oligodendrogliomas: tumor grade stratification using diffusion-, susceptibility-, and perfusion-weighted MRI
.
Neuroradiology.
2017
;
59
(
6
):
555
562
.

49.

Roux
A
,
Tauziede-Espariat
A
,
Zanello
M
, et al. .
Imaging growth as a predictor of grade of malignancy and aggressiveness of IDH-mutant and 1p/19q-codeleted oligodendrogliomas in adults
.
Neuro Oncol
.
2020
;
22
(
7
):
993
1005
.

50.

Reyes-Botero
G
,
Dehais
C
,
Idbaih
A
, et al. ;
POLA Network
.
Contrast enhancement in 1p/19q-codeleted anaplastic oligodendrogliomas is associated with 9p loss, genomic instability, and angiogenic gene expression
.
Neuro Oncol
.
2014
;
16
(
5
):
662
670
.

51.

Wang
K
,
Wang
Y
,
Fan
X
, et al. .
Regional specificity of 1p/19q co-deletion combined with radiological features for predicting the survival outcomes of anaplastic oligodendroglial tumor patients
.
J Neurooncol.
2018
;
136
(
3
):
523
531
.

52.

Giantini-Larsen
AM
,
Abou-Mrad
Z
,
Yu
KK
, et al. .
Treatment and outcomes of IDH1-mutant gliomas in elderly patients
.
J Neurosurg.
2023
;
140
(
2
):
367
376
.

53.

Juratli
TA
,
Tummala
SS
,
Riedl
A
, et al. .
Radiographic assessment of contrast enhancement and T2/FLAIR mismatch sign in lower grade gliomas: Correlation with molecular groups
.
J Neurooncol.
2019
;
141
(
2
):
327
335
.

54.

Yamauchi
T
,
Ohno
M
,
Matsushita
Y
, et al. .
Radiological characteristics based on isocitrate dehydrogenase mutations and 1p/19q codeletion in grade II and III gliomas
.
Brain Tumor Pathol.
2018
;
35
(
3
):
148
158
.

55.

Darvishi
P
,
Batchala
PP
,
Patrie
JT
, et al. .
Prognostic value of preoperative MRI Metrics for diffuse lower-grade glioma molecular subtypes
.
AJNR Am J Neuroradiol.
2020
;
41
(
5
):
815
821
.

56.

Park
YW
,
Park
KS
,
Park
JE
, et al. .
Qualitative and quantitative magnetic resonance imaging phenotypes may predict CDKN2A/B homozygous deletion status in isocitrate dehydrogenase-mutant astrocytomas: A multicenter study
.
Korean J Radiol.
2023
;
24
(
2
):
133
144
.

57.

Yu
Y
,
Villanueva-Meyer
J
,
Grimmer
MR
, et al. .
Temozolomide-induced hypermutation is associated with distant recurrence and reduced survival after high-grade transformation of low-grade IDH-mutant gliomas
.
Neuro Oncol
.
2021
;
23
(
11
):
1872
1884
.

58.

Kocakavuk
E
,
Anderson
KJ
,
Varn
FS
, et al. .
Radiotherapy is associated with a deletion signature that contributes to poor outcomes in patients with cancer
.
Nat Genet.
2021
;
53
(
7
):
1088
1096
.

59.

Touat
M
,
Li
YY
,
Boynton
AN
, et al. .
Mechanisms and therapeutic implications of hypermutation in gliomas
.
Nature.
2020
;
580
(
7804
):
517
523
.

60.

Jaspers
JPM
,
Taal
W
,
van Norden
Y
, et al. .
Early and late contrast enhancing lesions after photon radiotherapy for IDH mutated grade 2 diffuse glioma
.
Radiother Oncol.
2023
;
184
:
109674
.

61.

Wetzel
EA
,
Farrell
MJ
,
Eldred
BSC
, et al. .
Retrospective examination of pseudoprogression in IDH mutant gliomas
.
Neurooncol. Adv..
2023
;
5
(
1
):
vdad028
.

62.

Huang
RY
,
Young
RJ
,
Ellingson
BM
, et al. .
Volumetric analysis of IDH-mutant lower-grade glioma: A natural history study of tumor growth rates before and after treatment
.
Neuro Oncol
.
2020
;
22
(
12
):
1822
1830
.

63.

Bhatia
A
,
Moreno
R
,
Reiner
AS
, et al. .
Tumor volume growth rates and doubling times during active surveillance of IDH-mutant low-grade glioma
.
Clin Cancer Res.
2023
;
30
(
1
):
106
115
.

64.

Gui
C
,
Kosteniuk
SE
,
Lau
JC
,
Megyesi
JF.
Tumor growth dynamics in serially-imaged low-grade glioma patients
.
J Neurooncol.
2018
;
139
(
1
):
167
175
.

65.

Leclerc
A
,
Roux
A
,
Elia
A
, et al. .
Radiographic growth rate as a predictor of aggressiveness of diffuse gliomas without 1p19q codeletion
.
Neurosurg Focus.
2024
;
56
(
2
):
E4
.

66.

Aoki
K
,
Suzuki
H
,
Yamamoto
T
, et al. .
Mathematical modeling and mutational analysis reveal optimal therapy to prevent malignant transformation in grade II IDH-Mutant Gliomas
.
Cancer Res.
2021
;
81
(
18
):
4861
4873
.

67.

van den Bent
MJ.
Interobserver variation of the histopathological diagnosis in clinical trials on glioma: A clinician’s perspective
.
Acta Neuropathol.
2010
;
120
(
3
):
297
304
.

68.

Louis
DN
,
Perry
A
,
Wesseling
P
, et al. .
The 2021 WHO classification of tumors of the central nervous system: A summary
.
Neuro Oncol
.
2021
;
23
(
8
):
1231
1251
.

69.

Giannini
C
,
Scheithauer
BW
,
Burger
PC
, et al. .
Cellular proliferation in pilocytic and diffuse astrocytomas
.
J Neuropathol Exp Neurol.
1999
;
58
(
1
):
46
53
.

70.

Coons
SW
,
Pearl
DK.
Mitosis identification in diffuse gliomas: Implications for tumor grading
.
Cancer.
1998
;
82
(
8
):
1550
1555
.

71.

Duregon
E
,
Bertero
L
,
Pittaro
A
, et al. .
Ki-67 proliferation index but not mitotic thresholds integrates the molecular prognostic stratification of lower grade gliomas
.
Oncotarget
.
2016
;
7
(
16
):
21190
21198
.

72.

Yoda
RA
,
Marxen
T
,
Longo
L
, et al. .
Mitotic index thresholds do not predict clinical outcome for IDH-mutant astrocytoma
.
J Neuropathol Exp Neurol.
2019
;
78
(
11
):
1002
1010
.

73.

Kling
T
,
Ferreyra Vega
S
,
Suman
M
, et al. .
Refinement of prognostication for IDH-mutant astrocytomas using DNA methylation-based classification
.
Brain Pathol.
2024
:
e13233
. doi:10.1111/bpa.13233.

74.

Franceschi
E
,
Tosoni
A
,
Bartolini
S
, et al. .
Histopathological grading affects survival in patients with IDH-mutant grade II and grade III diffuse gliomas
.
Eur J Cancer.
2020
;
137
:
10
17
.

75.

Shirahata
M
,
Ono
T
,
Stichel
D
, et al. .
Novel, improved grading system(s) for IDH-mutant astrocytic gliomas
.
Acta Neuropathol.
2018
;
136
(
1
):
153
166
.

76.

Cimino
PJ
,
Zager
M
,
McFerrin
L
, et al. .
Multidimensional scaling of diffuse gliomas: Application to the 2016 World Health Organization classification system with prognostically relevant molecular subtype discovery
.
Acta Neuropathol Commun
.
2017
;
5
(
1
):
39
.

77.

Yang
RR
,
Shi
ZF
,
Zhang
ZY
, et al. .
IDH mutant lower grade (WHO Grades II/III) astrocytomas can be stratified for risk by CDKN2A, CDK4 and PDGFRA copy number alterations
.
Brain Pathol.
2020
;
30
(
3
):
541
553
.

78.

Kros
JM
,
Rushing
E
,
Uwimana
AL
, et al. .
Mitotic count is prognostic in IDH mutant astrocytoma without homozygous deletion of CDKN2A/B. Results of consensus panel review of EORTC trial 26053 (CATNON) and EORTC trial 22033-26033
.
Neuro Oncol
.
2023
;
25
(
8
):
1443
1449
.

79.

Ostrom
QT
,
Shoaf
ML
,
Cioffi
G
, et al. .
National-level overall survival patterns for molecularly-defined diffuse glioma types in the United States
.
Neuro Oncol
.
2023
;
25
(
4
):
799
807
.

80.

Giannini
C
,
Scheithauer
BW
,
Weaver
A
, et al. .
Oligodendrogliomas: Reproducibility and prognostic value of histologic diagnosis and grading
.
J Neuropathol Exp Neurol.
2001
;
60
(
3
):
248
262
.

81.

Figarella-Branger
D
,
Mokhtari
K
,
Dehais
C
, et al. ;
POLA Network
.
Mitotic index, microvascular proliferation, and necrosis define 3 groups of 1p/19q codeleted anaplastic oligodendrogliomas associated with different genomic alterations
.
Neuro. Oncol
.
2014
;
16
(
9
):
1244
1254
.

82.

Figarella-Branger
D
,
Mokhtari
K
,
Dehais
C
, et al. ;
POLA Network
.
Mitotic index, microvascular proliferation, and necrosis define 3 pathological subgroups of prognostic relevance among 1p/19q co-deleted anaplastic oligodendrogliomas
.
Neuro. Oncol
.
2016
;
18
(
6
):
888
890
.

83.

Tesileanu
CMS
,
Vallentgoed
WR
,
Sanson
M
, et al. .
Non-IDH1-R132H IDH1/2 mutations are associated with increased DNA methylation and improved survival in astrocytomas, compared to IDH1-R132H mutations
.
Acta Neuropathol.
2021
;
141
(
6
):
945
957
.

84.

Brat
DJ
,
Aldape
K
,
Colman
H
, et al. .
cIMPACT-NOW update 5: Recommended grading criteria and terminologies for IDH-mutant astrocytomas
.
Acta Neuropathol.
2020
;
139
(
3
):
603
608
.

85.

Louis
DN
,
Perry
A
,
Wesseling
P
, et al. .
The 2021 WHO classification of tumors of the central nervous system: A summary
.
Neuro-Oncology.
2021
;
23
(
8
):
1231
1251
.

86.

Tesileanu
CMS
,
Vallentgoed
WR
,
French
PJ
,
van den Bent
MJ.
Molecular markers related to patient outcome in patients with IDH-mutant astrocytomas grade 2 to 4: A systematic review
.
Eur J Cancer.
2022
;
175
:
214
223
.

87.

Kocakavuk
E
,
Johnson
KC
,
Sabedot
TS
, et al. .
Hemizygous CDKN2A deletion confers worse survival outcomes in IDHmut-noncodel gliomas
.
Neuro Oncol
.
2023
;
25
(
9
):
1721
1723
.

88.

Hickman
RA
,
Gedvilaite
E
,
Ptashkin
R
, et al. .
CDKN2A/B mutations and allele-specific alterations stratify survival outcomes in IDH-mutant astrocytomas
.
Acta Neuropathol.
2023
;
146
(
6
):
845
847
.

89.

Yokoda
RT
,
Cobb
WS
,
Yong
RL
, et al. .
CDKN2A mutations have equivalent prognostic significance to homozygous deletion in IDH-mutant astrocytoma
.
J Neuropathol Exp Neurol.
2023
;
82
(
10
):
845
852
.

90.

Tesileanu
CMS
,
van den Bent
MJ
,
Sanson
M
, et al. .
Prognostic significance of genome-wide DNA methylation profiles within the randomized, phase 3, EORTC CATNON trial on non-1p/19q deleted anaplastic glioma
.
Neuro-Oncology.
2021
;
23
(
9
):
1547
1559
.

91.

Appay
R
,
Dehais
C
,
Maurage
CA
, et al. ;
POLA Network
.
CDKN2A homozygous deletion is a strong adverse prognosis factor in diffuse malignant IDH-mutant gliomas
.
Neuro Oncol.
2019
;
21
(
12
):
1519
1528
.

92.

Aoki
K
,
Nakamura
H
,
Suzuki
H
, et al. .
Prognostic relevance of genetic alterations in diffuse lower-grade gliomas
.
Neuro Oncol.
2018
;
20
(
1
):
66
77
.

93.

Richardson
TE
,
Walker
JM.
The prognostic significance of RB and PI3K pathway alterations in IDH-mutant grade II/III astrocytomas
.
J Neuropathol Exp Neurol.
2020
;
79
(
9
):
1019
1023
.

94.

Cimino
PJ
,
Holland
EC.
Targeted copy number analysis outperforms histologic grading in predicting patient survival for WHO grades II/III IDH-mutant astrocytomas
.
Neuro-Oncology.
2019
;
21
(
6
):
819
821
.

95.

Richardson
TE
,
Walker
JM.
CCND2 amplification is an independent adverse prognostic factor in IDH-mutant lower-grade astrocytoma
.
Clin Neuropathol.
2021
;
40
(
4
):
209
214
.

96.

Lee
K
,
Kim
SI
,
Kim
EE
, et al. .
Genomic profiles of IDH-mutant gliomas: MYCN-amplified IDH-mutant astrocytoma had the worst prognosis
.
Sci Rep.
2023
;
13
(
1
):
6761
.

97.

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
(
18
):
5537
5547
.

98.

Ceccarelli
M
,
Barthel
FP
,
Malta
TM
, et al. ;
TCGA Research Network
.
Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma
.
Cell.
2016
;
164
(
3
):
550
563
.

99.

Korshunov
A
,
Casalini
B
,
Chavez
L
, et al. .
Integrated molecular characterization of IDH-mutant glioblastomas
.
Neuropathol Appl Neurobiol.
2019
;
45
(
2
):
108
118
.

100.

Draaisma
K
,
Wijnenga
MM
,
Weenink
B
, et al. .
PI3 kinase mutations and mutational load as poor prognostic markers in diffuse glioma patients
.
Acta Neuropathol Commun
.
2015
;
3
(
1
):
88
.

101.

Wong
QH
,
Li
KK
,
Wang
WW
, et al. .
Molecular landscape of IDH-mutant primary astrocytoma Grade IV/glioblastomas
.
Mod Pathol.
2021
;
34
(
7
):
1245
1260
.

102.

Zhao
Z
,
Zhang
KN
,
Wang
Q
, et al. .
Chinese Glioma Genome Atlas (CGGA): A comprehensive resource with functional genomic data from chinese glioma patients
.
Genomics, Proteomics Bioinform
.
2021
;
19
(
1
):
1
12
.

103.

Barthel
FP
,
Johnson
KC
,
Varn
FS
, et al. ;
GLASS Consortium
.
Longitudinal molecular trajectories of diffuse glioma in adults
.
Nature.
2019
;
576
(
7785
):
112
120
.

104.

Mamatjan
Y
,
Voisin
MR
,
Nassiri
F
, et al. .
Integrated molecular analysis reveals hypermethylation and overexpression of HOX genes to be poor prognosticators in isocitrate dehydrogenase mutant glioma
.
Neuro Oncol
.
2023
;
25
(
11
):
2028
2041
.

105.

Prost
D
,
Bielle
F
,
Ligon
KL
,
Touat
M.
Mutational burden and immune recognition of gliomas
.
Curr Opin Oncol.
2021
;
33
(
6
):
626
634
.

106.

Michaud
K
,
de Tayrac
M
,
D’Astous
M
, et al. .
Contribution of 1p, 19q, 9p and 10q automated analysis by FISH to the diagnosis and prognosis of oligodendroglial tumors according to WHO 2016 guidelines
.
PLoS One.
2016
;
11
(
12
):
e0168728
.

107.

Alentorn
A
,
Dehais
C
,
Ducray
F
, et al. ;
POLA Network
.
Allelic loss of 9p21.3 is a prognostic factor in 1p/19q codeleted anaplastic gliomas
.
Neurology.
2015
;
85
(
15
):
1325
1331
.

108.

Wijnenga
MMJ
,
French
PJ
,
Dubbink
HJ
, et al. .
Prognostic relevance of mutations and copy number alterations assessed with targeted next generation sequencing in IDH mutant grade II glioma
.
J Neurooncol.
2018
;
139
(
2
):
349
357
.

109.

Suwala
AK
,
Felix
M
,
Friedel
D
, et al. .
Oligosarcomas, IDH-mutant are distinct and aggressive
.
Acta Neuropathol.
2022
;
143
(
2
):
263
281
.

110.

Yip
S
,
Butterfield
YS
,
Morozova
O
, et al. .
Concurrent CIC mutations, IDH mutations, and 1p/19q loss distinguish oligodendrogliomas from other cancers
.
J Pathol.
2012
;
226
(
1
):
7
16
.

111.

Bettegowda
C
,
Agrawal
N
,
Jiao
Y
, et al. .
Mutations in CIC and FUBP1 contribute to human oligodendroglioma
.
Science.
2011
;
333
(
6048
):
1453
1455
.

112.

LeBlanc
VG
,
Firme
M
,
Song
J
, et al. .
Comparative transcriptome analysis of isogenic cell line models and primary cancers links capicua (CIC) loss to activation of the MAPK signalling cascade
.
J Pathol.
2017
;
242
(
2
):
206
220
.

113.

Chittaranjan
S
,
Chan
S
,
Yang
C
, et al. .
Mutations in CIC and IDH1 cooperatively regulate 2-hydroxyglutarate levels and cell clonogenicity
.
Oncotarget
.
2014
;
5
(
17
):
7960
7979
.

114.

Gleize
V
,
Alentorn
A
,
Connen de
KL
, et al. .
CIC inactivating mutations identify aggressive subset of 1p19q codeleted gliomas
.
Ann Neurol.
2015
;
78
(
3
):
355
374
.

115.

Dubbink
HJ
,
Atmodimedjo
PN
,
Kros
JM
, et al. .
Molecular classification of anaplastic oligodendroglioma using next generation sequencing. A report of the prospective randomized EORTC Brain Tumor Group 26951 phase III trial
.
Neuro Oncol
.
2016
;
18
(
3
):
388
400
.

116.

Halani
SH
,
Yousefi
S
,
Vega
JV
, et al. .
Multi-faceted computational assessment of risk and progression in oligodendroglioma implicates NOTCH and PI3K pathways
.
NPJ Precis Oncol.
2018
;
2
:
24
.

117.

Dono
A
,
Alfaro-Munoz
K
,
Yan
Y
, et al. .
Molecular, histological, and clinical characteristics of oligodendrogliomas: A multi-institutional retrospective study
.
Neurosurgery.
2022
;
90
(
5
):
515
522
.

118.

Pekmezci
M
,
Rice
T
,
Molinaro
AM
, et al. .
Adult infiltrating gliomas with WHO 2016 integrated diagnosis: Additional prognostic roles of ATRX and TERT
.
Acta Neuropathol.
2017
;
133
(
6
):
1001
1016
.

119.

Mu
Q
,
Chai
R
,
Pang
B
, et al. .
Identifying predictors of glioma evolution from longitudinal sequencing
.
Sci Transl Med.
2023
;
15
(
716
):
eadh4181
.

120.

Kamoun
A
,
Idbaih
A
,
Dehais
C
, et al. ;
POLA network
.
Integrated multi-omics analysis of oligodendroglial tumours identifies three subgroups of 1p/19q co-deleted gliomas
.
Nat Commun.
2016
;
7
:
11263
.

121.

Chen
H
,
Thomas
C
,
Munoz
FA
, et al. .
Polysomy is associated with poor outcome in 1p/19q codeleted oligodendroglial tumors
.
Neuro Oncol
.
2019
;
21
(
9
):
1164
1174
.

122.

Richardson
TE
,
Williams
M
,
Galbraith
K
, et al. .
Total copy number variation, somatic mutation burden, and histologic grade correlate with clinical outcome in oligodendroglioma
.
Clin Neuropathol.
2020
;
39
(
5
):
238
242
.

123.

Garnier
L
,
Vidal
C
,
Chinot
O
, et al. .
Characteristics of anaplastic oligodendrogliomas short-term survivorS: A POLA network study
.
Oncologist.
2022
;
27
(
5
):
414
423
.

124.

Alghamri
MS
,
Thalla
R
,
Avvari
RP
, et al. .
Tumor mutational burden predicts survival in patients with low-grade gliomas expressing mutated IDH1
.
Neurooncol. Adv..
2020
;
2
(
1
):
vdaa042
.

125.

Gilhodes
J
,
Meola
A
,
Cabarrou
B
, et al. .
A multigene signature associated with progression-free survival after treatment for IDH Mutant and 1p/19q Codeleted Oligodendrogliomas
.
Cancers (Basel)
.
2023
;
15
(
12
):
3067
.

126.

Hu
X
,
Martinez-Ledesma
E
,
Zheng
S
, et al. .
Multigene signature for predicting prognosis of patients with 1p19q co-deletion diffuse glioma
.
Neuro Oncol
.
2017
;
19
(
6
):
786
795
.

127.

Wu
F
,
Yin
YY
,
Fan
WH
, et al. .
Immunological profiles of human oligodendrogliomas define two distinct molecular subtypes
.
EBioMedicine
.
2023
;
87
:
104410
.

128.

Malta
TM
,
de Souza
CF
,
Sabedot
TS
, et al. .
Glioma CpG island methylator phenotype (G-CIMP): Biological and clinical implications
.
Neuro Oncol
.
2018
;
20
(
5
):
608
620
.

129.

Li
KK
,
Shi
ZF
,
Malta
TM
, et al. .
Identification of subsets of IDH-mutant glioblastomas with distinct epigenetic and copy number alterations and stratified clinical risks
.
Neurooncol. Adv..
2019
;
1
(
1
):
vdz015
.

130.

Mazor
T
,
Pankov
A
,
Johnson
BE
, et al. .
DNA methylation and somatic mutations converge on the cell cycle and define similar evolutionary histories in brain tumors
.
Cancer Cell
.
2015
;
28
(
3
):
307
317
.

131.

Bai
H
,
Harmanci
AS
,
Erson-Omay
EZ
, et al. .
Integrated genomic characterization of IDH1-mutant glioma malignant progression
.
Nat Genet.
2016
;
48
(
1
):
59
66
.

132.

de Souza
CF
,
Sabedot
TS
,
Malta
TM
, et al. .
A Distinct DNA methylation shift in a subset of glioma CpG island methylator phenotypes during tumor recurrence
.
Cell Rep
.
2018
;
23
(
2
):
637
651
.

133.

Malta
TM
,
Sabedot
TS
,
Morosini
NS
, et al. .
The epigenetic evolution of glioma is determined by the IDH1 mutation status and treatment regimen
.
Cancer Res.
2024
;
84
(
5
):
741
756
.

134.

Capper
D
,
Stichel
D
,
Sahm
F
, et al. .
Practical implementation of DNA methylation and copy-number-based CNS tumor diagnostics: The Heidelberg experience
.
Acta Neuropathol.
2018
;
136
(
2
):
181
210
.

135.

Capper
D
,
Jones
DTW
,
Sill
M
, et al. .
DNA methylation-based classification of central nervous system tumours
.
Nature.
2018
;
555
(
7697
):
469
474
.

136.

Bady
P
,
Kurscheid
S
,
Delorenzi
M
, et al. .
The DNA methylome of DDR genes and benefit from RT or TMZ in IDH mutant low-grade glioma treated in EORTC 22033
.
Acta Neuropathol.
2018
;
135
(
4
):
601
615
.

137.

Molenaar
RJ
,
Verbaan
D
,
Lamba
S
, et al. .
The combination of IDH1 mutations and MGMT methylation status predicts survival in glioblastoma better than either IDH1 or MGMT alone
.
Neuro Oncol
.
2014
;
16
(
9
):
1263
1273
.

138.

Tesileanu
CMS
,
Gorlia
T
,
Golfinopoulos
V
,
French
PJ
,
van den Bent
MJ.
MGMT promoter methylation determined by the MGMT-STP27 algorithm is not predictive for outcome to temozolomide in IDH-mutant anaplastic astrocytomas
.
Neuro Oncol
.
2022
;
24
(
4
):
665
667
.

139.

Horbinski
C
,
McCortney
K
,
Stupp
R.
MGMT promoter methylation is associated with patient age and 1p/19q status in IDH-mutant gliomas
.
Neuro Oncol
.
2021
;
23
(
5
):
858
860
.

140.

Mathur
R
,
Zhang
Y
,
Grimmer
MR
, et al. .
MGMT promoter methylation level in newly diagnosed low-grade glioma is a predictor of hypermutation at recurrence
.
Neuro Oncol
.
2020
;
22
(
11
):
1580
1590
.

141.

Koivunen
P
,
Lee
S
,
Duncan
CG
, et al. .
Transformation by the (R)-enantiomer of 2-hydroxyglutarate linked to EGLN activation
.
Nature.
2012
;
483
(
7390
):
484
488
.

142.

Johannessen
TA
,
Mukherjee
J
,
Viswanath
P
, et al. .
Rapid Conversion of Mutant IDH1 from driver to passenger in a model of human gliomagenesis
.
Mol Cancer Res.
2016
;
14
(
10
):
976
983
.

143.

Mellinghoff
IK
,
Lu
M
,
Wen
PY
, et al. .
Vorasidenib and ivosidenib in IDH1-mutant low-grade glioma: a randomized, perioperative phase 1 trial
.
Nat Med.
2023
;
29
(
3
):
615
622
.

144.

de la Fuente
MI
,
Colman
H
,
Rosenthal
M
, et al. .
Olutasidenib (FT-2102) in patients with relapsed or refractory IDH1-mutant glioma: a multicenter, open-label, phase Ib/II trial
.
Neuro Oncol
.
2023
;
25
(
1
):
146
156
.

145.

Mellinghoff
IK
,
Ellingson
BM
,
Touat
M
, et al. .
Ivosidenib in isocitrate dehydrogenase 1–mutated advanced glioma
.
J Clin Oncol.
2020
;
38
(
29
):
3398
3406
.

146.

Wick
A
,
Bähr
O
,
Schuler
M
, et al. .
Phase I assessment of safety and therapeutic activity of BAY1436032 in patients with IDH1-mutant solid tumors
.
Clin Cancer Res.
2021
;
27
(
10
):
2723
2733
.

Author notes

Matthias Preusser and Patrick Y Wen Equal contribution

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.