2,149
Views
12
CrossRef citations to date
0
Altmetric
Editorial

Determining optimal treatment strategy for diffuse glioma: the emerging role of IDH mutations

, &

Abstract

The isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) genes mutate frequently in gliomas, and it has become increasingly apparent that IDH mutation status accounts for much of the prognostic information previously rendered by histological grading. Most glioblastomas (90–95%) are IDH wild-type and most lower-grade diffuse gliomas (80%) are IDH-mutant. We examine here how IDH mutation status interacts with treatments known to influence survival (surgery, chemotherapy and radiotherapy) in patients with gliomas, and the impact of the IDH mutations on patients’ survival after such treatments. IDH mutations is associated with more complete surgical resection of enhancing disease, and with a better response to RT. In addition, there is increasing clinical evidence that, in certain contexts, IDH mutations predict chemotherapeutic sensitivity. Mutations in IDH and other genes are beginning to drive decisions on therapy for diffuse gliomas and will likely allow tailoring of treatment by molecular profile in the future.

Our knowledge about molecular markers in gliomas has undergone a rapid evolution in the past few years. This new era began with the publication of a landmark genetic sequencing study in 2008, in which isocitrate dehydrogenase 1 (IDH1) was identified as a frequently mutated gene in secondary glioblastoma Citation[1]. Since that time, much work has been carried out to further determine the clinical value of mutations in IDH1 and close family member IDH2 in gliomas. Simultaneously, assessment of IDH mutation status has rapidly become incorporated as a standard component in the neuropathological assessment of brain tumors. Detection methods include immunohistochemistry and PCR analysis with DNA sequencing.

Numerous further genomic alterations are strongly associated with IDH mutations – overlapping to define distinct subgroups of gliomas, including mutations of the TP53 gene and the α-thalassemia/mental retardation syndrome X-linked gene in astrocytic gliomas Citation[2], whereas oligodendroglial tumors harbor more frequent mutations in the homolog of the Drosophila capicua gene, mutations of the far upstream element binding protein 1 (FUBP 1), co-deletion of chromosome arms 1p and 19q Citation[3,4] and mutations in promoter of the telomerase reverse transcriptase gene Citation[5,6].

Neuro-oncologists now face the challenge of incorporating these new markers into the treatment of glioma patients since many unanswered questions remain about the evidence base for using these markers in clinical decision making. Whether the favorable prognosis seen in association with IDH mutation in gliomas is due to improved natural history or better response to therapy is unknown. Until now, treatment decision making has been driven by histological type and malignancy grade. IDH-mutant gliomas comprise 80% of diffuse low-grade gliomas (WHO grade II), 60% of anaplastic gliomas (WHO grade III) and 5–10% of glioblastomas (WHO grade IV) Citation[7]. IDH-mutant status therefore offers the basis for an alternative method of classification. Here, we will focus on the emerging role of the IDH mutations in gliomas, with regards to how they might influence treatment decisions.

Surgical management

Surgery is the first essential step in the classification and management of gliomas. In general, patients with suspected glioma undergo surgical resection to establish a histological and molecular diagnosis and to alleviate neurological symptoms. Due to the close linkage between surgical specimen size and accurate histologic grading Citation[8], the choice for tumor biopsy instead of resection should be reserved for cases where surgical resection is associated with high risk of morbidity (due to location of the tumor or impaired clinical condition of the patient). If a biopsy is indicated, sufficient amount of tissue should be obtained for molecular testing Citation[9].

In glioblastomas, which are largely IDH wild-type, maximum safe resection of contrast-enhancing tumor is associated with improved survival Citation[10]. In the management of IDH-mutant gliomas, aggressive surgery targeting all nonenhancing areas of disease remains the standard first-line treatment since, in low-grade gliomas (which again are largely IDH-mutant), a significant correlation has been demonstrated between maximal tumor resection and overall survival Citation[11]. IDH-mutant high-grade gliomas are more amenable to complete resection of enhancing tumor, a fact that may contribute to the better prognosis observed after aggressive surgery in this patient population Citation[12]. Possible explanations for this phenomenon are the frequent location of IDH-mutant gliomas in the frontal lobe, the less functional disturbance of adjacent normal brain, or that they may have a less fibrovascular tumor consistency Citation[13], facilitating more extensive removal. Nevertheless, the lack of Class I evidence – due to lack of prospective randomized surgical trials with determination of IDH mutations – prevents certainty in assessing the influence of extent of resection.

Radiation therapy

Radiotherapy (RT) is a mainstay in the initial treatment of patients with de novo high-grade gliomas, which are primarily IDH wild-type. It is administered in the early postoperative phase and is often combined with CT. In diffuse (low-grade) gliomas, which are largely IDH-mutant, the European Organization for Research and Treatment of Cancers (EORTC) trial 22845 showed a significant benefit of early RT in relation to longer progression-free survival and seizure control Citation[14]. However, the trial was conducted before the detection of IDH mutations as a molecular marker in gliomas became standard. Therefore, prospectively collected data on the impact of IDH mutations with regard to RT are still lacking. On the other hand, an increased radiosensitivity of IDH-mutant diffuse gliomas in patients treated with RT has been reported in a small patient cohort Citation[15]. This effect was clinically translated as improved survival. Furthermore, recent in vitro studies Citation[16] and clinical data from EORTC trial 26951 Citation[17] support the hypothesis that IDH-mutant gliomas are highly sensitive to RT. This sensitivity might be due the fact that wild-type IDH enzymes protect cells against gamma irradiation by maintaining NADPH levels that buffer against irradiation-induced reactive oxygen species, thus protecting cells from apoptosis Citation[18].

Chemotherapy

Although CT with alkylating agents, for example, temozolomide or lomustine, improves survival in IDH wild-type gliomas Citation[19], few studies have investigated the role of CT in IDH-mutant gliomas Citation[17,20]. In this context, the chromosomal 1p/19q-deletion status should be considered as well because virtually all 1p/19q-deleted gliomas are also IDH-mutant Citation[21]. Long-term follow-up data of the randomized Phase III Radiation Therapy Oncology Group (RTOG) trial 9402 Citation[22] and the EORTC trial 26951 Citation[23] demonstrated an improved outcome – in both progression-free and overall survival – in 1p/19q-co-deleted anaplastic oligodendroglial tumors when procarbazine, lomustine and vincristine chemotherapy (PCV) was added to RT; for instance, in the RTOG 9402 trial, the median survival of those with 1p/19q co-deleted tumors treated with PCV plus RT was twice that of patients receiving RT (14.7 vs 7.3 years). Consequently, 1p/19q codeletion is now incorporated in treatment decision making in anaplastic oligodendroglial tumors.

Moreover, both aforementioned trials have retrospectively established IDH mutations as a prognostic marker. In the RTOG 9402 trial, survival of anaplastic oligodendroglial and mixed oligoastrocytic tumor patients exhibiting an IDH-mutation treated with PCV and RT was significantly longer than patients without an IDH mutation (9.4 vs 5.7 years) Citation[20]. Thus, the role of IDH as a predictive marker of response to CT with PCV may be dependent upon its linkage with 1p/19q codeletion and oligodendroglial histology. Accordingly, IDH status was prognostic but not predictive in the retrospective analysis of the German Glioma Network NOA-04 trial, in which patients with anaplastic gliomas were randomized to receive first either RT or CT (PCV or temozolomide) and upon disease progression to receive the other treatment modality Citation[24]. Nevertheless, a recently published study demonstrated that IDH-mutant glioblastomas have a more pronounced radiographic response to chemoradiation therapy by serial quantitative MR volumetry than do their IDH wild-type counterparts Citation[25]. The predictive value of IDH mutation, separate from 1p/19q co-deletions, in relation to CT response has not yet been investigated prospectively. The role of adjuvant temozolomide in newly diagnosed IDH-mutant malignant gliomas also remains with a less developed evidence base than that of PCV Citation[26].

Also unclear is the value of IDH mutation for predicting response to CT in diffuse low-grade tumors. Contradictory results have been obtained in small retrospective studies. A larger prospective trial, the Phase III RTOG 9802 trial, was conducted to compare the addition of PCV to RT versus treatment with RT alone for ‘high-risk’ WHO grade II diffuse glioma patients. This ‘high-risk’ cohort included patients with subtotal tumor resection or aged >40 years. The long-term follow-up data demonstrate that PCV + RT prolongs both progression-free and overall survival compared with RT alone Citation[27]. However, in this trial, multivariable models that incorporate 1p/19q co-deletion and IDH mutational analyses have not yet been reported. Hence, no statements can be made about the predictive implication of molecular markers in this patient group. A further limitation is that all patients in RTOG 9802 received RT, precluding determination of the value of CT alone in this patient group. Finally, no conclusions can be drawn on the effect of treatment with temozolomide as patients in RTOG 9802 were treated with PCV.

On the other hand, there is increasing evidence that IDH-mutant gliomas might follow a common mechanism of resistance to CT. In a recently published study, the exomes of 23 IDH-mutant low-grade gliomas and their subsequent recurrent tumors were sequenced Citation[28]. In 6 of 10 patients treated with temozolomide, these tumors were hypermutated at recurrence and harbored driver mutations as a consequence of temozolomide-induced mutagenesis, indicative of an alternative evolutionary path to high-grade gliomas. Thus, a connection between temozolomide treatment, driver mutations in oncogenic signaling pathways, and malignant progression was suggested. A further study aimed to investigate the possible causative relationship between IDH mutation and response to temozolomide in vitro Citation[29]. In this study, IDH mutations introduced into immortalized untransformed human astrocytes initiated expression alterations in DNA repair genes that facilitated repair of temozolomide-induced DNA damage, resulting in temozolomide.

The results of the ongoing CODEL Citation[30] and CATNON Citation[31], multicenter Phase III trials, may well clear these uncertainties. The CODEL trial addresses newly diagnosed anaplastic gliomas with 1p/19q codeletion – mainly oligodendroglial tumors – and the CATNON trial is designed for patients with newly diagnosed anaplastic gliomas without 1p/19q codeletion – primarily astrocytic tumors. Once available, the results from these clinical studies can hopefully determine the influence of molecular stratification by IDH mutation on resistance to temozolomide.

Finally, novel methods directly targeting the IDH mutation itself hold significant promise for improving treatment of these gliomas. Inhibitors of the mutant enzyme can slow the growth of an IDH-mutant glioma in mice Citation[32], and a vaccination strategy targeting the mutant IDH1 epitope has been shown to induce antitumor immunity in a humanized murine model Citation[33]. Clinical trials of these experimental therapies are underway.

Conclusion

IDH mutation status is emerging as a useful guide for treatment decision making in gliomas. To optimize the application of existing therapies, we would propose the following conclusions based on the available evidence: enhancing high-grade IDH wild-type gliomas should be managed with an aim toward complete surgical resection of enhancing disease, and combined/concurrent chemoradiation therapy with temozolomide according to the Stupp glioblastoma regimen, whereas in nonenhancing or diffuse lower-grade IDH wild-type gliomas, surgical resection followed by RT alone is the current standard of care, awaiting the results of the ongoing EORTC/RTOG Phase III trials; IDH-mutant 1p/19q-co-deleted gliomas should be managed with maximal safe surgical resection of both enhancing and nonenhancing disease, followed by sequential radiation therapy and PCV CT; IDH-mutant 1p/19q-nondeleted glioma management can be controversial, given the conflicting available evidence. These gliomas should be managed with maximal safe surgical resection of enhancing and nonenhancing disease, which can be followed by radiation alone, or sequential radiation and CT, or enrollment in clinical trials of CT or IDH mutant-directed agents.

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

References

  • Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008;321(5897):1807-12
  • Kannan K, Inagaki A, Silber J, et al. Whole-exome sequencing identifies ATRX mutation as a key molecular determinant in lower-grade glioma. Oncotarget 2012;3(10):1194-203
  • Bettegowda C, Agrawal N, Jiao Y, et al. Mutations in CIC and FUBP1 contribute to human oligodendroglioma. Science 2011;333(6048):1453-5
  • 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
  • Jiao Y, Killela PJ, Reitman ZJ, et al. Frequent ATRX, CIC, FUBP1 and IDH1 mutations refine the classification of malignant gliomas. Oncotarget 2012;3(7):709-22
  • Killela PJ, Pirozzi CJ, Healy P, et al. Mutations in IDH1, IDH2, and in the TERT promoter define clinically distinct subgroups of adult malignant gliomas. Oncotarget 2014;5(6):1515-25
  • Hartmann C, Meyer J, Balss J, et al. Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a study of 1,010 diffuse gliomas. Acta Neuropathol 2009;118(4):469-74
  • Glantz MJ, Burger PC, Herndon JE2nd, et al. Influence of the type of surgery on the histologic diagnosis in patients with anaplastic gliomas. Neurology 1991;41(11):1741-4
  • Kim BY, Jiang W, Beiko J, et al. Diagnostic discrepancies in malignant astrocytoma due to limited small pathological tumor sample can be overcome by IDH1 testing. J Neurooncol 2014;118(2):405-12
  • Lacroix M, Abi-Said D, Fourney DR, et al. A multivariate analysis of 416 patients with glioblastoma multiforme: prognosis, extent of resection, and survival. J Neurosurg 2001;95(2):190-8
  • Smith JS, Chang EF, Lamborn KR, et al. Role of extent of resection in the long-term outcome of low-grade hemispheric gliomas. J Clin Oncol 2008;26(8):1338-45
  • 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
  • Lai A, Kharbanda S, Pope WB, et al. Evidence for sequenced molecular evolution of IDH1 mutant glioblastoma from a distinct cell of origin. J Clin Oncol 2011;29(34):4482-90
  • van den Bent MJ, Afra D, de Witte O, et al. Long-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: the EORTC 22845 randomised trial. Lancet 2005;366(9490):985-90
  • Juratli TA, Kirsch M, Robel K, et al. IDH mutations as an early and consistent marker in low-grade astrocytomas WHO grade II and their consecutive secondary high-grade gliomas. J Neurooncol 2012;108(3):403-10
  • Li S, Chou AP, Chen W, et al. Overexpression of isocitrate dehydrogenase mutant proteins renders glioma cells more sensitive to radiation. Neuro Oncol 2013;15(1):57-68
  • van den Bent MJ, Dubbink HJ, Marie Y, et al. IDH1 and IDH2 mutations are prognostic but not predictive for outcome in anaplastic oligodendroglial tumors: a report of the European Organization for Research and Treatment of Cancer Brain Tumor Group. Clin Cancer Res 2010;16(5):1597-604
  • Bleeker FE, Atai NA, Lamba S, et al. The prognostic IDH1 (R132) mutation is associated with reduced NADP+-dependent IDH activity in glioblastoma. Acta Neuropathol 2010;119(4):487-94
  • Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Eng J Med 2005;352(10):987-96
  • Cairncross JG, Wang M, Jenkins RB, et al. Benefit from procarbazine, lomustine, and vincristine in oligodendroglial tumors is associated with mutation of IDH. J Clin Oncol 2014;32(8):783-90
  • Labussiere M, Idbaih A, Wang XW, et al. All the 1p19q codeleted gliomas are mutated on IDH1 or IDH2. Neurology 2010;74(23):1886-90
  • Cairncross G, Wang M, Shaw E, et al. Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402. J Clin Oncol 2013;31(3):337-43
  • van den Bent MJ, Brandes AA, Taphoorn MJ, et al. Adjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-term follow-up of EORTC brain tumor group study 26951. J Clin Oncol 2013;31(3):344-50
  • Wick W, Hartmann C, Engel C, et al. NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide. J Clin Oncol 2009;27(35):5874-80
  • Tran AN, Lai A, Li S, et al. Increased sensitivity to radiochemotherapy in IDH1 mutant glioblastoma as demonstrated by serial quantitative MR volumetry. Neuro Oncol 2014;16(3):414-20
  • Lassman AB, Iwamoto FM, Cloughesy TF, et al. International retrospective study of over 1000 adults with anaplastic oligodendroglial tumors. Neuro Oncol 2011;13(6):649-59
  • van den Bent MJ. Practice changing mature results of RTOG study 9802: another positive PCV trial makes adjuvant chemotherapy part of standard of care in low-grade glioma. Neuro Oncol 2014;16(12):1570-4
  • Johnson BE, Mazor T, Hong C, et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 2014;343(6167):189-93
  • Ohba S, Mukherjee J, See WL, et al. Mutant IDH1-driven cellular transformation increases RAD51-mediated homologous recombination and temozolomide resistance. Cancer Res 2014;74(17):4836-44
  • Radiation therapy with concomitant and adjuvant temozolomide or radiation therapy with adjuvant PCV or temozolomide alone in treating patients with anaplastic glioma. Available from: https://clinicaltrials.gov/ct2/show/NCT00887146
  • Radiation therapy with or without temozolomide in treating patients with anaplastic glioma. Available from: https://clinicaltrials.gov/ct2/show/NCT00626990
  • Rohle D, Popovici-Muller J, Palaskas N, et al. An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science 2013;340(6132):626-30
  • Schumacher T, Bunse L, Pusch S, et al. A vaccine targeting mutant IDH1 induces antitumour immunity. Nature 2014;512(7514):324-7

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.