Low grade glioma: a journey towards a cure

Similar documents
Systemic Treatment. Third International Neuro-Oncology Course. 23 May 2014

Radioterapia no Tratamento dos Gliomas de Baixo Grau

성균관대학교삼성창원병원신경외과학교실신경종양학 김영준. KNS-MT-03 (April 15, 2015)

21/03/2017. Disclosure. Practice Changing Articles in Neuro Oncology for 2016/17. Gliomas. Objectives. Gliomas. No conflicts to declare

Neuro-Oncology Program

Neuro-Oncology. Martin J. van den Bent. Department of Neuro-oncology/Neurology, Erasmus M.C. Cancer Institute, Rotterdam, Netherlands

UPDATES ON CHEMOTHERAPY FOR LOW GRADE GLIOMAS

Chemotherapy plus Radiotherapy versus Radiotherapy Alone for Patients with Anaplastic Oligodendroglioma: Long Term Results of RTOG 9402

Prior to 1993, the only data available in the medical

Precision medicine for gliomas

PROCARBAZINE, lomustine, and vincristine (PCV) is

Scottish Medicines Consortium

Collection of Recorded Radiotherapy Seminars

Carmustine implants and Temozolomide for the treatment of newly diagnosed high grade glioma

Response to postoperative radiotherapy as a prognostic factor for patients with low-grade gliomas

Contemporary Management of Glioblastoma

Survival of High Grade Glioma Patients Treated by Three Radiation Schedules with Chemotherapy: A Retrospective Comparative Study

PRINCESS MARGARET CANCER CENTRE CLINICAL PRACTICE GUIDELINES

Concepts for a personalized neurosurgical oncology. XXIV Annual Conference Pietro Paoletti 27. November 2015

Neurocognitive Assessment in Patients with Brain Metastases. Martin Klein VU University Medical Center Amsterdam, The Netherlands

PRINCESS MARGARET CANCER CENTRE CLINICAL PRACTICE GUIDELINES

EORTC (RTOG 0834 Endorsed) Opened: July 22, 2009

Surgical resection versus watchful waiting in low-grade gliomas

Adjuvant treatment of high grade gliomas

Oncological Management of Brain Tumours. Anna Maria Shiarli SpR in Clinical Oncology 15 th July 2013

Role of Prophylactic Cranial Irradiation in Small Cell Lung Cancer

LOW GRADE ASTROCYTOMAS

CNAJ12TZRT. Protocol Code. Neuro-Oncology. Tumour Group. Dr. Brian Thiessen. Contact Physician

Glioblastoma: Current Treatment Approach 8/20/2018

Corporate Medical Policy

Oligodendroglial Tumors: A Review

Stereotactic Radiosurgery for Brain Metastasis: Changing Treatment Paradigms. Overall Clinical Significance 8/3/13

CNS SESSION 3/8/ th Multidisciplinary Management of Cancers: A Case based Approach

Management of Glioma: The Basics Glioma Update The clinical challenge. Glioma a malignant disease of the CNS

A clinical perspective on neuropathology and molecular genetics in brain tumors

NON-SURGICAL STRATEGY FOR ADULT EPENDYMOMA

Clinical Trials for Adult Brain Tumors - the Imaging Perspective

Hypofractionated radiation therapy for glioblastoma

Incidence of Early Pseudo-progression in a Cohort of Malignant Glioma Patients Treated With Chemoirradiation With Temozolomide

Case Presentation: USCAP Jason T. Huse, MD, PhD Assistant Member Department of Pathology Memorial Sloan Kettering Cancer Center

University of Zurich. Temozolomide and MGMT forever? Zurich Open Repository and Archive. Weller, M. Year: 2010

2015 EUROPEAN CANCER CONGRESS

Chemotherapy in Adults with Gliomas

The New WHO Classification and the Role of Integrated Molecular Profiling in the Diagnosis of Malignant Gliomas

Predictive Biomarkers in GBM

CNS Tumors: The Med Onc Perspective. Ronald J. Scheff, MD Associate Clinical Professor Weill Medical College of Cornell U.

Oligodendrogliomas & Oligoastrocytomas

Treatment with Tumor-Treating Fields therapy and pulse dose bevacizumab in patients with bevacizumab-refractory recurrent glioblastoma: A case series.

2011 Oncology Highlights News from ASCO 2011:

Going Past the Data for Temozolomide. J. Lee Villano, M.D., Ph.D., Nathalie Letarte, B.Pharm, M.Sc, Linda R. Bressler, Pharm. D.

Concomitant (without adjuvant) temozolomide and radiation to treat glioblastoma: A retrospective study

Related Policies None

MOLECULAR DIAGNOSTICS OF GLIOMAS

BRAIN TUMOURS: SUCCESSES AND CHALLENGES ON THE OTHER SIDE OF THE BLOOD-BRAIN BARRIER

Imaging for suspected glioma

Jennie W Taylor, MD 02/15/2019. Patient 1 Presentation

UNDERSTANDING MOLECULAR TESTING IN BRAIN TUMORS: HOW CLINICALLY USEFUL IS IT?

Zurich Open Repository and Archive. Long-term survival of glioblastoma patients treated with radiotherapy and lomustine plus temozolomide

RINDOPEPIMUT (CDX-110) IN GLIOBLASTOMA

Bevacizumab rescue therapy extends the survival in patients with recurrent malignant glioma

PRESURGICAL PLANNING. Strongly consider neuropsychological evaluation before functional imaging study Strongly consider functional imaging study

Integrating molecular markers into the World Health Organization classification of CNS tumors: a survey of the neuro-oncology community

Radiation and DCIS. The 16 th Annual Conference on A Multidisciplinary Approach to Comprehensive Breast Care and Imaging

Bevacizumab in combination with temozolomide and regional radiation therapy for up-front treatment of patients with newly-diagnosed glioblastoma

Adjuvant Therapy in Locally Advanced Head and Neck Cancer. Ezra EW Cohen University of Chicago. Financial Support

Glioblastoma: Adjuvant Treatment Abdulrazag Ajlan, MD, MSc, FRCSC, UCNS(D)

ACTR-46. Presented at the Society for Neuro-Oncology Annual Scientific Meeting, November 16-19, 2017; San Francisco, CA, USA

Examining large groups of cancer patients to identify ways of predicting which therapies cancers might respond to.

Procarbazine Lomustine and VinCRIStine (PCV) Therapy INDICATIONS FOR USE:

Long-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: the EORTC randomised trial

Clinical Policy: Electric Tumor Treating Fields (Optune) Reference Number: PA.CP.MP.145

Background. Central nervous system (CNS) tumours. High-grade glioma

Gliomas in the 2016 WHO Classification of CNS Tumors

Antiangiogenic drugs in unresectable glioblastoma. Dra. Carmen Balañá. /

VAL-083: Validated DNA-targeting Agent for Underserved Cancer Patients. September 2018

Zurich Open Repository and Archive. Long-term survival of glioblastoma patients treated with radiotherapy and lomustine plus temozolomide

Update on Limited Small Cell Lung Cancer. Laurie E Gaspar MD, MBA Prof/Chair Radiation Oncology University of Colorado Denver

Tristate Lung Meeting 2014 Pro-Con Debate: Surgery has no role in the management of certain subsets of N2 disease

Disclosures. Preoperative Treatment: Chemotherapy or ChemoRT? Adjuvant chemotherapy helps. so what about chemo first?

3-D conformal radiotherapy with concomitant and adjuvant temozolomide for patients with glioblastoma multiforme and evaluation of prognostic factors

Advances in Brain Tumor Research: Leveraging BIG data for BIG discoveries

Description. Section: Durable Medical Equipment Effective Date: January 15, 2016 Subsection: Original Policy Date: December 6, 2013 Subject:

CHMP Type II variation assessment report

Chemotherapy in malignant brain tumors

Chika Nwachukwu, Ph.D. MS IV Radiation Oncology Rotation

CNS pathology Third year medical students. Dr Heyam Awad 2018 Lecture 12: CNS tumours 2/3

5-hydroxymethylcytosine loss is associated with poor prognosis for

Epidemiology and outcome research of glioma patients in Southern Switzerland: A population based analysis

SYSTEMIC MANAGEMENT OF PEDIATRIC PRIMARY BRAIN TUMORS

IAN CROCKER = TIM HOWARD

Minesh Mehta, Northwestern University. Chicago, IL

Radiation plus Procarbazine, CCNU, and Vincristine in Low-Grade Glioma

LETTER INTENT

Brain Tumors: Radiologic Perspective

RADIATION THERAPY ONCOLOGY GROUP RTOG 0925

11/27/2017. Modern Treatment of Meningiomas. Disclosures. Modern is Better? No disclosures relevant to this presentation

Dose dense 1 week on/1 week off temozolomide in recurrent glioma: a retrospective study

What yield in the last decade about Molecular Diagnostics in Neuro

Temozolomide Concomitant and Adjuvant to Radiotherapy in Elderly Patients With Glioblastoma

Pioneering vaccines that transform lives.

Transcription:

Editorial Page 1 of 5 Low grade glioma: a journey towards a cure Ali K. Choucair SIU School of Medicine, Springfield, IL, USA Correspondence to: Ali K. Choucair, MD. Professor of Neurology, Director of Neuro-Oncology, SIU School of Medicine, Springfield, IL, USA. Email: akiahc@gmail.com. Submitted Jul 13, 2017. Accepted for publication Jul 20, 2017. doi: 10.21037/cco.2017.07.08 View this article at: http://dx.doi.org/10.21037/cco.2017.07.08 Data from recent clinical, translational, and basic science research provide us with a powerful propelling energy towards a new design of clinical trials. The updated survival data from the RTOG 9802 (see below) hint at the possibility that among the long-term survivors, there might be some with a cure. In all the fields of human oncology no entity has had the misnomer as that of a low grade glioma (LGG). Histologically it has been considered a benign or low grade tumor. Yet biologically in due time it almost always progresses towards a malignant killer. Furthermore LGG as a single histological grade contains heterogeneity of molecular entities each with its own biological behavior that calls for a personalized approach to treatment. For that reason there is a need to revisit data from past and recent clinical trials in order to adapt a new mindset in the design of LGG clinical trials. In order to adopt best evidence for treating the individual patient this new mindset must re-examine the applicability of past clinical data in light of the recent molecular data. Whereas past clinical trials have enrolled all LGG patients into one entity going forward the design of new clinical trials must take into consideration the incorporation and subsequent validation of the new molecular data. As clinicians we approach each patient with the mindset that we live in the era of personalized medicine with a duty and emphasis upon true informed consent and the patient-physician shared decision-making. In treating LGG, revised survival data from past clinical trials with incorporation of molecular profile has shown that there is no on size fits all when it comes to treatment. There is always the individual approach to the individual patient. The molecular era The Cancer Genome Atlas Research Network has provided us with an integrative genomic analysis of diffuse LGG subdividing them into three different groups with different but important prognosis: IDH mutation with co-deletion of 1p/19q, IDH mutation without 1p/19q co-deletion, and IDH wild type (1). The 2016 WHO classification of tumors of the central nervous system has integrated these molecular markers with the traditional histological grading system (2). Traditionally, in the design of clinical trials we have stratified patients into low-risk and high-risk LGG based upon the Pignatti criteria (3). These criteria have served us very well despite their two limitations: first there was no central pathological validation of the local pathology and second the analysis did not include molecular markers. Also keep in mind that EORTC (4) clinical trial that randomized patients with LGG between early vs. deferred radiation did not take into consideration either clinical, histological or molecular risk factors and it excluded patients with GTR. We now have biomarkers with important and different prognostication that will need to be incorporated in our treatment decisions and as such it would be unwise to indiscriminately recommend the watch-and-see approach. The role of the extent of resection (EOR) This is the one modality of treatment for LGG that is not the subject of controversy. In surgical neuro-oncology as important as it is how much the surgeon takes out is more important how much is left behind. In the RTOG 9802 trial the size of residual disease on the postoperative MRI correlated with the risk of progression (5) Although it remains to be answered whether GTR is equally important in all three LGG molecular subtypes, yet should radiotherapy become necessary it is wise that the volume to be radiated is as small as possible to minimize the late

Page 2 of 5 Choucair. Low grade glioma cognitive effects of radiotherapy. Although systemic reviews and meta-analysis to quantify the association of the EOR with the likelihood of survival found no prospective or high class I or II studies, yet there appears to be an association between the EOR and improved OS as well as PFS (6-8). Despite ongoing controversy (9,10), additional benefits of the EOR in the hands of the experienced neurosurgeon are the following improvements in: (I) Seizure control; (II) QOL; (III) PFS; (IV) Symptomatic relief; (V) Decadron-dependency; (VI) Delayed malignant transformation. Role of radiotherapy, chemotherapy or combination of radio-chemotherapy In the landmark trial of RTOG 9802, 254 patients with high-risk LGG as defined by the Pignatti criteria were randomized to radiotherapy with or without adjuvant PCV chemotherapy. Of the patients who had progressed on the radiation-only arm more than 70% were treated with PCV. The immediate combination therapy resulted in a gain of PFS (10.4 vs. 4.0 years) and of median OS (13.3 vs. 7.8 years). PFS at 10 years was 51% vs. 21% in favor of the combination therapy (11). This trial has proven that when the decision is made to proceed with radiotherapy it would be best that adjuvant chemotherapy be used immediately following the completion of radiotherapy rather than waiting to start the chemotherapy for the time of progression. Neither arm of the study showed deterioration in the neurocognitive function as measured by the ( insensitive ) MMSE. Molecular data was available in only 45% of the patients. The benefit was particularly in favor of the patients with oligodendroglioma molecular profile (IDH mutation and co-deletion of 1p/19q). In a single arm trial of RTOG 0424 newly diagnosed patients with high-risk LGG were treated with radiotherapy plus concurrent as well as adjuvant temozolomide (12). Although the 3-year OS at 73% and the PFS at 59.2%, both better than at the time best known historical controls, yet we should keep in mind the median PFS of 4.5 years was inferior to that of 10.4 years in the RTOG 9802. This study also has pending analysis of QOL as well as a battery for neurocognitive function as secondary endpoints. Biomarker analysis is pending. Recently the EORTC has reported on a randomized, phase 3, intergroup trial (EORTC 22033-26033) in which patients with high-risk LGG (using the Pignatti criteria) either newly diagnosed or progressive disease previously untreated after surgery were randomized to either radiotherapy or dose dense temozolomide (75 mg/m 2 /day for 21 days every 28 days for 12 cycles (13). This, at the time of study initiation, was the only study to prospectively stratify tumor by molecular subtypes before treatment randomization. The authors have made it clear, however, that the study was not empowered for the analysis of the molecular subgroups but only for hypothesis generation. The median PFS was 46M for RT group vs. 39M for TMZ group (P=0.22). PFS was 62, 48 and 20 months for patients with IDH mutation and 1p/19q co-deletion, IDH mutation and no co-deletion, and IDH wild type respectively. The PFS at 5 years showed significant difference between RT and TMZ only for the patients with IDH wild type: 42 months for RT vs. 19 months for temozolomide (P=0.004).The median OS, necessary for future individualized treatment choices based upon the predictive effects of different biomarker subtypes, had not been reached. Of note is that PFS for the RT only arm in this study (46 months) was similar to that of RTOG 9802 RT only arm (48 months). There are three take home messages from this study: (I) since the IDH mutation with 1p/19q co-deletion had the best outcome regardless of treatment there is a strong argument in favor of initiating patients with this combination biomarker on chemotherapy, especially if there is a large postoperative residual tumor. (II) The second take home message is since patients with the IDH wild type tumors had the worst prognosis regardless of treatment there is an argument, supported by data from RTOG 9802 as well as the Cancer Genomic Atlas, that these patients should be considered as pre-gbm. (III) The third take home message as pointed out by the authors is that the study could not establish a preferred treatment modality with high variability of the individual disease course where some patients showed progression within few months while others were symptom free for many years, once again emphasizing the need for an individualized approach to treatment. As for the MGMT status, while it is important to check it in patients with the IDH wild type it does not provide any additional predictive or prognostic value in patients with IDH mutation (13). There was no difference in the effects of RT or temozolomide upon the HRQOL or NCF as measured by

Chinese Clinical Oncology, Vol 6, No 4 August 2017 Page 3 of 5 the (insensitive) MMSE (14). Following the final report of RTOG 9802, the NCI suspended accrual to the one study, ECOG E3F05, which was designed for comparison of RT vs. RT plus TMZ. The role of temozolomide as single agent in newly diagnosed LGG with postoperative residual disease: the UCSF experience As mentioned above, the RTOG 9802 study showed significant improvement in both the OS and PFS for the patients who received PCV immediately following RT but not for those whose PCV was deferred to the time of progression. A natural question that arose from this trial was what would happen if alkylating chemotherapy was to be given at time of postoperative diagnosis and RT was deferred to the time of progression in the group with the most favorable molecular profile (the oligodendroglioma). In a single arm, phase II trial the UCSF group has investigated standard temozolomide as a single agent in 120 patients with newly diagnosed LGG with postoperative residual disease by MRI (15). The primary end point was objective radiological response rate. The study looked at three molecular subtypes: 1p/19q co-deletion (45%), IDH mutation with intact 1p/19q (38%) and IDH wild type (16%). The majority of the patients (53% had not received salvage RT at time of last follow-up. Although the partial response rate was low at 6% yet there was a high rate (86%) of stable or improved disease. The median PFS was 4.2 years with a median OS of 9.7 years, which compare favorably to 4.0 and 7.8 years in the RT only arm of RTOG 9802 and 4.5 years PFS on RTOG 0424. Molecular subtype was associated with rate of disease progression, PFS and OS. Pretreatment tumor volume was associated with PFS and OS. Compared to 8% of patients with IDH mutation and 56% of patients with wild type IDH, patients with co-deleted tumors showed no evidence of progression during treatment, thus raising the possibility that these patients may be candidates for deferred or even omitted RT in favor of chemotherapy. Summary for the future (I) Minimizing inter-observer variability: a strong argument in favor of molecular stratification of LGG is the considerable variability in both the typing and grading of LGGs (16,17). In the Gorlia et al. study, 21% of cases reviewed had to be excluded due to differing central pathology review diagnosis, 17% (II) (III) (IV) being qualified as HGGs (16). For the future clinical trials quality controls would, as much as resources would allow, be a major plus if efforts are made to minimize the same problem when it comes to molecular profiling. There is an obvious need to evaluate and validate risk profile for patients with LGG to combine both Pignatti clinical criteria as well as the newly established molecular subtypes. Along with this validation comes the consideration for a prognostic model. In the past the one prognostic model for PFS and OS that was reported by Gorlia et al. (16) used data from 339 patients from the EORTC clinical trials for the development of the prognostic model and 450 patients from North American cooperative groups for the validation of the model. The study established three risk groups: low, intermediate and high. Criteria used to develop the prognostic models included clinical, radiological and histological criteria. Inclusion of molecular markers in the development of future prognostic profiles will substantially improve their predictive power. It is very clear that future clinical trials will have to address some very important issues as to the timing and choice of therapy for patients with LGG. Considering the impressive survival data from the RTOG 9802 it is conceivable that there might have been some cures amongst those with longterm OS. With that in mind, future clinical trials must incorporate HRQOL and neurocognitive function as important outcomes. Hopefully, with long term survivors it will be lesser of a challenge to capture long-term neurocognitive function. The time might come when neurocognitive function becomes an important primary endpoint in this disease entity, keeping in mind that the MMSE is not sensitive enough to identify early and important neurocognitive changes. There is a need to best identify the subtype of LGG who are best candidates for watch-andsee approach. In this regard the data from both the EORTC (13) and the UCSF (15) in using temozolomide and deferring RT is an encouraging step in that direction. That being said, we need to keep in mind that 45% of patients with oligodendroglioma grade II and 74% of grade II astrocytoma recur as grade III-IV (18). Thus, the patients with favorable molecular subtypes will need

Page 4 of 5 Choucair. Low grade glioma a closer monitoring of clinical as well as radiological status. We should also seek for paired tissue to examine if and how often does alkylating chemotherapy result in super-mutated aggressive tumor at time of recurrence. (V) In the era of molecular data question arises if future clinical trials should combine both groups of lower grades II and III glioma with IDH mutation. Reuss et al. examined the relationship between age and histological grade vs. IDH mutation in predicting survival in patients with lower grades II and III. They combined three independent series containing 1,245 patients (out of a total of 1,360) with lower grades II and III and showed that IDH mutation type predicted survival much more robustly than WHO histological grading with overall survival of 10.9 and 9.3 years respectively for the patients with IDH-mutant grades II and III (19). In the same study, they also showed that age at presentation was identical for the same two groups of patients. These findings with far-reaching consequences for the practicing clinician need to be validated in prospective studies (19). In another study, Olar et al. examined the IDH mutation status and role of histological grade, age, and mitotic index in the overall survival in patients with glioma grades II and III (20). They found that the prognostic impact of the grade was modest in the patients with the IDH-mutation type in comparison to IDH-wild type (HR 1.21 vs. 1.74, respectively). Mitotic index was significantly associated with outcome in the group with IDH-wild but not the IDH-mutation (HR 4.41 vs. 1.10 and P<0.0001 versus P<0.5, respectively). Age was significantly associated with outcome only in the IDH-wild type (20). There is an obvious need to take consideration of this date in the design of future clinical trials. Lastly, we live in the age of personalized medicine with emphasis upon patient-physician shared decision making. Patient-centered goals are for improved survival, minimizing toxicity, improving HRQOL, and protecting neurocognitive function. I strongly believe these patient-centered goals and objectives are best pursued when patients are enrolled on clinical trials. Every patient with LGG is a candidate for a clinical trial until it is proven otherwise. Acknowledgements None. Footnote Conflicts of Interest: The author has no conflicts of interest to declare. References 1. Cancer Genome Atlas Research Network, Brat DJ, Verhaak RG, et al. Comprehensive, Integrative Genomic Analysis of Diffuse Lower-Grade Gliomas. N Engl J Med 2015;372:2481-98. 2. Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol 2016;131:803-20. 3. Pignatti F, van den Bent M, Curran D, et al. Prognostic factors for survival in adult patients with cerebral lowgrade glioma. J Clin Oncol 2002;20:2076-84. 4. 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;366:985-90. 5. Shaw EG, Berkey B, Coons SW, et al. Recurrence following neurosurgeon-determined gross-total resection of adult supratentorial low-grade glioma: results of a prospective clinical trial. J Neurosurg 2008;109:835-41. 6. Brown TJ, Bota DA, Maher EA, et al. Association of aggressive resection with survival and progression-free survival in adult low-grade glioma: A systematic review and meta-analysis with numbers needed to treat. J Clin Oncol 2017;35:suppl; abstr 2025. 7. 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;26:1338-45. 8. Jakola AS, Myrmel KS, Kloster R, et al. Comparison of a strategy favoring early surgical resection vs a strategy favoring watchful waiting in low-grade gliomas. JAMA 2012;308:1881-8. 9. Duffau H, Taillandier L. New concepts in the management of diffuse low-grade glioma: Proposal of a multistage and individualized therapeutic approach. Neuro Oncol 2015;17:332-42. 10. Zadeh G, Khan OH, Vogelbaum M, et al. Much debated controversies of diffuse low-grade gliomas. Neuro Oncol 2015;17:323-6. 11. Buckner JC, Shaw EG, Pugh SL, et al. Radiation plus Procarbazine, CCNU, and Vincristine in Low-Grade

Chinese Clinical Oncology, Vol 6, No 4 August 2017 Page 5 of 5 Glioma. N Engl J Med 2016;374:1344-55. 12. Fisher BJ, Hu C, Macdonald DR, et al. Phase 2 study of temozolomide-based chemoradiation therapy for highrisk low-grade gliomas: preliminary results of Radiation Therapy Oncology Group 0424. Int J Radiat Oncol Biol Phys 2015;91:497-504. 13. Baumert BG, Hegi ME, van den Bent MJ, et al. Temozolomide chemotherapy versus radiotherapy in high-risk low-grade glioma (EORTC 22033-26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol 2016;17:1521-32. 14. Reijneveld JC, Taphoorn MJ, Coens C, et al. Healthrelated quality of life in patients with high-risk low-grade glioma (EORTC 22033-26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol 2016;17:1533-42. 15. Wahl M, Phillips JJ, Molinaro AM, et al. Chemotherapy for adult low-grade gliomas: clinical outcomes by molecular subtype in a phase II study of adjuvant temozolomide. Neuro Oncol 2017;19:242-51. 16. Gorlia T, Wu W, Wang M, et al. New validated prognostic models and prognostic calculators in patients with lowgrade gliomas diagnosed by central pathology review: a pooled analysis of EORTC/RTOG/NCCTG phase III clinical trials. Neuro Oncol 2013;15:1568-79. 17. van den Bent MJ. Interobserver variation of the histopathological diagnosis in clinical trials on glioma: a clinician's perspective. Acta Neuropathol 2010;120:297-304. 18. Jaeckle KA, Decker PA, Ballman KV, et al. Transformation of low grade glioma and correlation with outcome: an NCCTG database analysis. J Neurooncol 2011;104:253-9. 19. 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;129:867-73. 20 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;129:585-96. Cite this article as: Choucair AK. Low grade glioma: a journey towards a cure.. doi: 10.21037/ cco.2017.07.08