section name header

Introduction

AHFS Class:

Generic Name(s):

Temozolomide, a prodrug that is converted in vivo to a cytotoxic alkylating metabolite, is an alkylating antineoplastic agent.1,2,3

Uses

[Section Outline]

Brain Tumors !!navigator!!

Temozolomide is used in adults for the treatment of newly diagnosed glioblastoma, concomitantly with radiotherapy and then as maintenance treatment.1 Temozolomide is also used in adults for the adjuvant treatment of newly diagnosed anaplastic astrocytoma and for the treatment of refractory disease.1

Glioblastoma

Temozolomide is used in adults concomitantly with radiation therapy for the treatment of newly diagnosed glioblastoma multiforme and then as maintenance treatment.1

Clinical Experience

Efficacy of temozolomide in patients with newly diagnosed glioblastoma is based on a randomized, multicenter, open-label trial (EORTC-NCIC 26981-22981; MK-7365-051) involving 573 patients.1,8 Patients were randomized to temozolomide (75 mg/m2 once daily) with standard fractionated radiation therapy (total dose of 60 Gy in 30 daily fractions of 2 Gy each) for 42 days (up to a maximum of 49 days) followed by a 4-week rest period and then adjuvant therapy with up to 6 cycles of temozolomide (150 or 200 mg/m2) on days 1-5 of every 28-day cycle or radiation therapy alone.1,8,20 The median age of patients was 56 years and 84% of patients had undergone debulking surgery.8 Salvage therapy with temozolomide was administered upon disease progression in 22% of patients who received initial therapy with temozolomide and radiation therapy and in 57% of patients who received initial therapy with radiation therapy alone.1

Overall survival was prolonged (unadjusted hazard ratio for death: 0.63) and median survival was increased by 2.5 months (14.6 versus 12.1 months) in patients receiving concomitant temozolomide and radiation therapy followed by adjuvant temozolomide compared with those receiving radiation therapy alone as initial therapy for glioblastoma multiforme.1,8 At a median follow-up of 61 months, overall survival benefit in patients receiving temozolomide and radiation therapy followed by adjuvant temozolomide compared with radiation therapy alone remained similar to the initial results.20 A subgroup analysis of tumor specimens in 206 patients for whom adequate tumor specimens were available indicated that methylation status of the MGMT promoter was a strong prognostic biomarker for overall survival benefit.20 Results of the subgroup analysis also suggested that the effect of temozolomide and radiation therapy followed by adjuvant temozolomide on overall survival was consistent regardless of MGMT promoter methylation status; however, progression-free survival benefit was observed only in temozolomide-treated patients with MGMT promoter methylated disease.20 Nausea (36 versus 16%), vomiting (20 versus 6%), anorexia (19 versus 9%), constipation (18 versus 6%), and thrombocytopenia (4 versus 1%) occurred more frequently in patients receiving concomitant temozolomide and radiation therapy than in those receiving radiation therapy alone.1

Among elderly patients with good performance status, the addition of concomitant and adjuvant temozolomide to hypofractionated radiation therapy appears to be safe and efficacious without impairing quality of life.28,30 In a randomized phase 3 study, 562 patients 65 years of age or older with newly diagnosed glioblastoma multiforme received temozolomide 75 mg/m2 once daily with hypofractionated radiation therapy (total dose of 40 Gy in 15 daily fractions) over a period of 3 weeks followed by maintenance therapy with up to 12 cycles of temozolomide (150 or 200 mg/m2) on days 1-5 of every 28-day cycle; or hypofractionated radiation therapy alone.29 Overall survival was prolonged (hazard ratio for death: 0.67; 95% confidence interval 0.56-0.80) and median survival was increased by 1.7 months (9.3 versus 7.6 months) in patients receiving concomitant temozolomide and hypofractionated radiation therapy followed by maintenance therapy with temozolomide compared with those receiving hypofractionated radiation therapy alone as initial therapy for glioblastoma multiforme.29 Although the magnitude of overall survival benefit was greatest in patients with methylated MGMT promoter receiving concomitant temozolomide and hypofractionated radiation therapy followed by adjuvant temozolomide compared with patients receiving hypofractionated radiation therapy alone, a numerical, but not statistically significant, overall survival benefit was observed in patients with unmethylated MGMT promoter (10.0 versus 7.9 months; hazard ratio of 0.75 with a 95% confidence interval 0.56-1.01).29

Clinical Perspective

Malignant gliomas often have inactivated MGMT (also referred to as O6-methylguanine DNA methyltransferase) due to aberrant methylation of its promoter region.17,19,20,21,22,28 The presence of MGMT promoter methylation has been used as a prognostic and predictive marker for response to therapy with alkylating agents.17,18,20,21,22,27 High expression of MGMT in cancer cells, such as glioma cells, account for the predominant mechanism of resistance to alkylating agents.17,18,20,21,22,27 Epigenetic silencing of MGMT by promoter methylation has been associated with improved survival in patients with glioblastoma multiforme receiving temozolomide, with or without radiation therapy.17,19,20,21,22,28 In the European Organisation for Research and Treatment of Cancer (EORTC) and National Cancer Institute of Canada Clinical Trials Group (NCIC) 26981-22981 study, overall survival benefit was observed regardless of MGMT promoter methylation status in patients with glioblastoma receiving concomitant temozolomide and radiation therapy; however, the magnitude of benefit was more pronounced in patients with methylated MGMT promoter.28

Lack of standardization in MGMT promoter methylation assays complicates interpretation of the prognostic and predictive impact of MGMT methylation status.17,21,23 Common methods for quantitating methylation status include methylation-specific polymerase chain reaction (MSP) and pyrosequencing (PSQ), both of which evaluate the MGMT promoter region, and immunohistochemistry (IHC), which evaluates MGMT protein expression.17,21,23 In a meta-analysis, MSP and PSQ appeared to be more prognostic for overall survival than IHC in patients with glioblastoma treated with temozolomide.23 The exact promoter regions (5'-cytosine-phosphate-guanine-3' [CpG]) and thresholds for interpretation are not well-defined; however, targeting multiple CpG promoter sites is likely to be more prognostic than targeting a single site.23

The current standard of treatment for newly diagnosed patients with glioblastoma includes maximum safe surgical resection followed by radiotherapy with concurrent and adjuvant chemotherapy with or without Tumor Treating Fields.31 Radiation therapy remains a key adjuvant therapy for most patients with glioblastoma given the locally recurrent pattern of disease spread after surgical resection.31 Temozolomide is the recommended systemic therapy in newly diagnosed glioblastoma due to its demonstrated survival benefit, with the greatest benefit seen in patients who harbor MGMT promoter methylation.31 Experts recommend conventional radiation therapy with concurrent and adjuvant temozolomide in glioblastoma patients up to 70 years of age with reasonable performance status.28,31 In elderly glioblastoma patients 70 years of age with reasonable performance status, hypofractionated radiation therapy with concurrent and adjuvant temozolomide may be considered.28

Anaplastic Astrocytoma

Temozolomide is used for the adjuvant treatment of adults with newly diagnosed anaplastic astrocytoma and in the treatment of refractory disease.1

Clinical Experience

The use of temozolomide as an adjuvant treatment in adults with newly diagnosed anaplastic astrocytoma is based on studies in the published literature in addition to interim results from the ongoing open-label, randomized, multicenter, phase 3 CATNON trial.1,32,33 Enrolled patients in CATNON had newly diagnosed anaplastic glioma without 1p/19q co-deletion, a World Health Organization (WHO) performance status score of 0-2, and adequate hematological, renal, and liver function, and were taking stable or decreasing corticosteroid doses.32,33 Patients were randomly assigned to radiotherapy alone, radiotherapy with adjuvant temozolomide (12 four-week cycles of 150-200 mg/m2 given on days 1-5), radiotherapy with concurrent temozolomide (75 mg/m2 per day), or radiotherapy and concurrent temozolomide plus adjuvant temozolomide.32,33 The primary endpoint was overall survival.32,33

At the initial interim analysis, the hazard ratio for overall survival for patients administered adjuvant temozolomide was 0.65 (99.145% confidence interval: 0.45-0.93); the median follow-up was 27 months.32 Median overall survival was not reached among patients who received adjuvant temozolomide and was 41.1 months in patients who did not receive adjuvant temozolomide.32 At 5 years, overall survival was 55.9% versus 44.1% in the respective groups.32 Similar beneficial results with the use of adjuvant temozolomide were observed at a second interim analysis conducted at a median follow-up of 55.7 months.33 Adjuvant temozolomide significantly improved median overall survival compared with no adjuvant temozolomide therapy (82.3 versus 46.9 months).33 However, administration of concurrent temozolomide did not result in a significant improvement in median overall survival when compared to no concurrent temozolomide therapy (66.9 months versus 60.4 months).33

The use of temozolomide in refractory anaplastic astrocytoma is based on tumor response rates in this population from an uncontrolled phase 2 study.1,4 In a single-arm, multicenter study in 162 patients with relapsed anaplastic astrocytoma who had a baseline Karnofsky performance status of 70 or greater, efficacy in a subgroup of 54 patients with refractory disease (i.e., progression following treatment with a nitrosourea and procarbazine) was demonstrated by an overall (complete plus partial) tumor response rate of 22% and a complete response rate of 9%.1,4 Median durations of all responses and complete responses were 50 and 64 weeks, respectively.1 Progression-free survival at 6 and 12 months was 45 and 29%, respectively; median progression-free survival was 4.4 months; 12-month overall survival was 65%; and median overall survival was 15.9 months.1

Dosage and Administration

[Section Outline]

General !!navigator!!

Pretreatment Screening

Patient Monitoring

Premedication and Prophylaxis

Dispensing and Administration Precautions

Cautions

[Section Outline]

Contraindications !!navigator!!

Warnings/Precautions !!navigator!!

Myelosuppression

Myelosuppression (e.g., pancytopenia, leukopenia, anemia) has been reported with temozolomide, with fatal outcomes observed in some patients.1 In certain cases, assessment has been complicated by exposure to concomitant medications (e.g., carbamazepine, phenytoin, co-trimoxazole) which may be associated with aplastic anemia.14,15,16

In a clinical study (MK-7365-006), myelosuppression with temozolomide usually occurred during the initial cycles of therapy and was not observed to be cumulative in nature.1 The median nadirs occurred for platelets at 26 days (range: 21 to 40 days) and for neutrophils at 28 days (range: 1 to 44 days).1 Myelosuppression resulted in hospitalization, blood transfusion, or discontinuation of temozolomide therapy in approximately 10% of patients.1 In clinical trials, women and geriatric patients have been shown to have an increased risk of developing myelosuppression.1

A complete blood count (CBC) should be obtained and absolute neutrophil count (ANC) and platelet count monitored before therapy initiation and as clinically indicated during treatment.1 When temozolomide is used concurrently with radiotherapy, a CBC should be obtained prior to initiation, weekly during treatment, and as clinically indicated.1

Temozolomide should be withheld if severe myelosuppression occurs.1 Temozolomide may be resumed at the same or reduced dose, or permanently discontinued, based on myelosuppression occurrence.1

Hepatotoxicity

Severe hepatotoxicity sometimes fatal, has occurred in patients receiving temozolomide.1 Reactivation of hepatitis B resulting in death has been reported in a patient receiving temozolomide for glioblastoma.9 Increased serum aminotransferase concentrations, hyperbilirubinemia, cholestasis, and hepatitis have been reported in postmarketing surveillance.1 Liver function tests should be evaluated at baseline, half-way through the first treatment cycle, prior to each subsequent cycle, and approximately 2-4 weeks following the last dose of temozolomide.1 Hepatitis screening and prophylactic therapy with antiviral agents as clinically indicated should be considered in patients receiving temozolomide.9

Pneumocystis Pneumonia

Pneumocystis pneumonia has been observed in patients administered temozolomide, with an increased risk seen in patients administered steroids or receiving longer treatment regimens of temozolomide.1

Prophylaxis against Pneumocystis pneumonia should be provided to all patients with newly diagnosed glioblastoma during the concomitant phase.1 Prophylaxis should be continued in patients with lymphopenia until resolution to grade 1 or less.1 All patients should be monitored for lymphopenia and Pneumocystis pneumonia development.1

Secondary Malignancies

Temozolomide-containing regimens are associated with an increased incidence of secondary malignancies.1 Myelodysplastic syndrome and secondary malignancies, including myeloid leukemia, have been observed.1

Fetal/Neonatal Morbidity and Mortality

May cause fetal harm based on its mechanism of action and animal findings; teratogenicity and embryolethality were demonstrated in animals at doses less than the maximum human dose based on body surface area (200 mg/m2).1 Spontaneous abortion and congenital malformations, including polymalformations with CNS, facial, cardiac, skeletal, and genitourinary system anomalies have been reported with exposure to temozolomide during pregnancy in postmarketing surveillance.1

Pregnancy should be avoided during therapy.1 The manufacturer states that a pregnancy test should be performed prior to starting temozolomide and females of reproductive potential should be advised to use effective contraceptive methods during and for 6 months after the last dose.1 In addition, male patients with such female partners should be advised to use effective methods of contraception during and for 3 months after the last dose.1 Male patients should not donate semen during therapy and for 3 months after the last dose.1 Patients should be apprised of the potential hazard to the fetus if temozolomide is used during pregnancy.1

Exposure to Opened Capsules

Recommended precautions should be observed to avoid exposure to opened capsules of temozolomide.1 (See Dosage and Administration.)

Specific Populations

Pregnancy

Temozolomide can cause fetal harm.1

Lactation

It is not known whether temozolomide or its metabolites are distributed in human milk.1 The effects of the drug on breast-fed infants or on the production of milk are also unknown.1 Patients should discontinue breast-feeding during therapy and for at least 1 week after the last dose because of potential risk, including myelosuppression, in breast-fed infants.1

Females and Males of Reproductive Potential

Temozolomide can cause fetal harm if administered during pregnancy.1 Females of reproductive potential should be advised to use effective contraceptive methods during and for 6 months after the last temozolomide dose.1 Male patients with such female partners should also be advised to use effective methods of contraception during and for 3 months after the last dose.1

Based on limited data, temozolomide may impair fertility in males.1 Changes in sperm parameters and genotoxic effects on sperm cells have occurred in men receiving temozolomide; however, the duration or reversibility of these effects has not been established.1 Male patients should not donate semen during therapy and for 3 months after the last dose.1

Pediatric Use

Safety and efficacy in pediatric patients have not been established.1 Efficacy was not demonstrated in open-label studies of children 3-18 years of age receiving temozolomide mostly for CNS tumors.1 Similar toxicity was observed in pediatric patients and adults.1

Geriatric Use

Experience in those 65 years of age and older with newly diagnosed glioblastoma is insufficient to determine whether they respond differently than younger adults.1

An increased incidence of grade 4 thrombocytopenia and/or neutropenia has been reported in patients 70 years of age or older compared with younger patients receiving temozolomide for refractory anaplastic astrocytoma.1

Similar toxicity was observed in patients 65 years of age or older and younger patients receiving temozolomide for newly diagnosed glioblastoma multiforme.1

Hepatic Impairment

Pharmacokinetics of temozolomide are not substantially altered in patients with mild to moderate hepatic impairment (Child-Pugh class A and B).1

Renal Impairment

Pharmacokinetics of temozolomide are not substantially altered in patients with creatinine clearance of 36-130 mL/minute per m2.1

Common Adverse Effects !!navigator!!

The most common adverse reactions (20%) include: alopecia, fatigue, nausea, vomiting, headache, constipation, anorexia, and convulsions.1

Drug Interactions

[Section Outline]

Drugs Affecting Hepatic Microsomal Enzymes !!navigator!!

Temozolomide and 5-(3-methyltriazen-1-yl)imidazole-4-carboxamide (MTIC) are only minimally metabolized by CYP isoenzymes.1

Carbamazepine !!navigator!!

Unlikely to affect temozolomide clearance.1 Possible additive hematologic toxicity (i.e., aplastic anemia); concomitant administration may complicate assessment of hematologic toxicity.14,15

Co-trimoxazole !!navigator!!

Possible additive hematologic toxicity (i.e., aplastic anemia); concomitant administration may complicate assessment of hematologic toxicity.16

Dexamethasone !!navigator!!

Unlikely to affect temozolomide clearance.1

Histamine H2-receptor Antagonists !!navigator!!

Unlikely to affect temozolomide clearance.1

Ondansetron !!navigator!!

Unlikely to affect temozolomide clearance.1

Phenobarbital !!navigator!!

Unlikely to affect temozolomide clearance.1

Phenytoin !!navigator!!

Unlikely to affect temozolomide clearance.1 Possible additive hematologic toxicity (i.e., aplastic anemia); concomitant administration may complicate assessment of hematologic toxicity.14,15,16

Prochlorperazine !!navigator!!

Unlikely to affect temozolomide clearance.1

Valproic Acid !!navigator!!

Pharmacokinetic interaction (decreased temozolomide clearance by about 5%).1 Clinical importance unknown.1

Other Information

Description

Temozolomide, an imidazotetrazine derivative, is an antineoplastic agent.1,2,3 Temozolomide is a prodrug and has little, if any, pharmacologic activity until hydrolyzed in vivo to 5-(3-methyltriazen-1-yl)imidazole-4-carboxamide (MTIC).1,2,3 Following administration of temozolomide, the drug undergoes rapid, nonenzymatic hydrolysis at physiologic pH to MTIC.1,3,4 MTIC is thought to exert its cytotoxic effects by acting as an alkylating agent at the O 6 and N 7 positions of guanine in DNA.1,2,3,4,10

Temozolomide is rapidly and completely absorbed after oral administration, with nearly 100% bioavailability.1,3,6,10 Peak plasma concentrations usually are attained within 1 hour.1,6,10 The bioequivalence of temozolomide (with respect to both peak plasma concentration and AUC) administered orally or as an IV infusion over 90 minutes at a dosage of 150 mg/m2 has been demonstrated.1 Food decreases the rate and extent of absorption after oral administration.1 A modified high fat breakfast decreased mean peak plasma temozolomide concentrations (32%) and AUC (9%).1

Temozolomide efficiently crosses the blood brain barrier.6,10 It is not known if temozolomide is distributed into human milk.1 Plasma protein binding is approximately 15%.1 MTIC, the main active metabolite, is further hydrolyzed to 5-amino-imidazole-4-carboxamide (AIC) and to methylhydrazine.1 CYP isoenzymes play only a minor role in metabolism of temozolomide and MTIC.1 About 38% of an administered dose is recovered over 7 days, principally in urine with <1% in feces.1 The mean elimination half-life of temozolomide is 1.8 hours.1,3,6,10 Apparent half-lives for metabolites MTIC and AIC are 2.1 and 2.6 hours, respectively.3

In patients with mild to moderate hepatic impairment, the pharmacokinetic profile resembles that in patients with normal hepatic function.1 Temozolomide has not been studied in patients with severe hepatic impairment.1 The pharmacokinetic profile of temozolomide is not affected by renal function in patients with creatinine clearance 36-130 mL/minute per m2.1 The drug has not been studied in patients with severe renal impairment (creatinine clearance <36 mL/minute per m2) or in patients with end-stage renal disease receiving dialysis.1

Advice to Patients

Additional Information

The American Society of Health-System Pharmacists, Inc. represents that the information provided in the accompanying monograph was formulated with a reasonable standard of care, and in conformity with professional standards in the field. Readers are advised that decisions regarding use of drugs are complex medical decisions requiring the independent, informed decision of an appropriate health care professional, and that the information contained in the monograph is provided for informational purposes only. The manufacturer's labeling should be consulted for more detailed information. The American Society of Health-System Pharmacists, Inc. does not endorse or recommend the use of any drug. The information contained in the monograph is not a substitute for medical care. For further information on the handling of antineoplastic agents, see the ASHP Guidelines on Handling Hazardous Drugs at [Web].

Preparations

Excipients in commercially available drug preparations may have clinically important effects in some individuals; consult specific product labeling for details.

Please refer to the ASHP Drug Shortages Resource Center for information on shortages of one or more of these preparations.

Temozolomide

Routes

Dosage Forms

Strengths

Brand Names

Manufacturer

Oral

Capsules

5 mg*

Temozolomide Capsules

Merck

20 mg*

Temozolomide Capsules

Merck

100 mg*

Temodar®

Merck

Temozolomide Capsules

140 mg*

Temodar®

Merck

Temozolomide Capsules

180 mg*

Temodar®

Merck

Temozolomide Capsules

250 mg*

Temodar®

Merck

Temozolomide Capsules

Parenteral

For injection, for IV infusion

100 mg

Temodar® for Injection

Merck

* available from one or more manufacturer, distributor, and/or repackager by generic (nonproprietary) name

Copyright

AHFS® Drug Information. © Copyright, 1959-2024, Selected Revisions October 10, 2024. American Society of Health-System Pharmacists, Inc., 4500 East-West Highway, Suite 900, Bethesda, MD 20814.

References

1. Merck & Co, Inc. Temodar® (temozolomide) capsules, Temodar (temozolomide) for injection® prescribing information. Whitehouse Station, NJ; 2023 Sept.

2. Anon. Temozolomide. Drugs Future . 1994; 19:746-9.

3. Baker SD, Wirth M, Statkevich P et al. Absorption, metabolism, and excretion of 14C-temozolomide following oral administration to patients with advanced cancer. Clin Cancer Res . 1999; 5:309-17. [PubMed 10037179]

4. Yung WKA, Prados MD, Yaya-Tur R et al. Multicenter phase II trial of temozolomide in patients with anaplastic astrocytoma or anaplastic oligoastrocytoma at first relapse. J Clin Oncol . 1999; 17:2762-71. [PubMed 10561351]

6. Baker SD, Wirth M, Statkevich P et al. Absorption, metabolism, and excretion of 14C-temozolomide following oral administration to patients with advanced cancer. Clin Cancer Res . 1999; 5:309-17. [PubMed 10037179]

8. Stupp R, Mason WP, van den Bent MJ et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med . 2005; 352:987-96. [PubMed 15758009]

9. Grewal J, Dellinger CA, Yung WK. Fatal reactivation of hepatitis B with temozolomide. N Engl J Med . 2007; 356:1591-2. [PubMed 17429098]

10. Baker SD, Wirth M, Statkevich P et al. Absorption, metabolism, and excretion of14C-temozolomide following oral administration to patients with advanced cancer. Clin Cancer Res . 1999; 5:309-17. [PubMed 10037179]

11. Agarwala SS, Kirkwood JM. Temozolomide, a novel alkylating agent with activity in the central nervous system, may improve the treatment of advanced metastatic melanoma. Oncologist . 2000; 5:144-51. [PubMed 10794805]

12. Merck & Co. Inc. Temodar® (temozolomide) capsules pharmacist information sheet. Whitehouse Station, NJ: 2020 Nov.

13. Merck & Co. Inc. Temodar® (temozolomide) Patient Information. Whitehouse Station, NJ; 2019 Nov.

14. Gilbar PJ, Pokharel K, Mangos HM. Temozolomide-induced aplastic anaemia: Case report and review of the literature. J Oncol Pharm Pract . 2020; :1078155220967087. [PubMed 33086908]

15. Newton SL, Kalamaha K, Fernandes HD. Temozolomide-induced Aplastic Anemia Treated with Eltrombopag and Granulocyte Colony Stimulating Factor: A Report of a Rare Complication. Cureus . 2018; 10:e3329. [PubMed 30473962]

16. Oh J, Kutas GJ, Davey P et al. Aplastic anemia with concurrent temozolomide treatment in a patient with glioblastoma multiforme. Curr Oncol . 2010; 17:124-6. [PubMed 20697524]

17. Alnahhas I, Alsawas M, Rayi A et al. Characterizing benefit from temozolomide in MGMT promoter unmethylated and methylated glioblastoma: a systematic review and meta-analysis. Neurooncol Adv . 2020; 2:vdaa082. [PubMed 33150334]

18. Jiapaer S, Furuta T, Tanaka S et al. Potential Strategies Overcoming the Temozolomide Resistance for Glioblastoma. Neurol Med Chir (Tokyo) . 2018; 58:405-421. [PubMed 30249919]

19. Brandes AA, Tosoni A, Franceschi E et al. Recurrence pattern after temozolomide concomitant with and adjuvant to radiotherapy in newly diagnosed patients with glioblastoma: correlation With MGMT promoter methylation status. J Clin Oncol . 2009; 27:1275-9. [PubMed 19188675]

20. Stupp R, Hegi ME, Mason WP et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol . 2009; 10:459-66. [PubMed 19269895]

21. Taylor JW, Schiff D. Treatment considerations for MGMT-unmethylated glioblastoma. Curr Neurol Neurosci Rep . 2015; 15:507. [PubMed 25394859]

22. Zhao YH, Wang ZF, Cao CJ et al. The Clinical Significance of O6-Methylguanine-DNA Methyltransferase Promoter Methylation Status in Adult Patients With Glioblastoma: A Meta-analysis. Front Neurol . 2018; 9:127. [PubMed 29619003]

23. McAleenan A, Kelly C, Spiga F et al. Prognostic value of test(s) for O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation for predicting overall survival in people with glioblastoma treated with temozolomide. Cochrane Database Syst Rev . 2021; 3:CD013316. [PubMed 33710615]

24. Wick W, Platten M, Meisner C et al. Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol . 2012; 13:707-15. [PubMed 22578793]

25. Malmström A, Grønberg BH, Marosi C et al. Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: the Nordic randomised, phase 3 trial. Lancet Oncol . 2012; 13:916-26. [PubMed 22877848]

26. Möllemann M, Wolter M, Felsberg J et al. Frequent promoter hypermethylation and low expression of the MGMT gene in oligodendroglial tumors. Int J Cancer . 2005; 113:379-85. [PubMed 15455350]

27. Wick W, Weller M, van den Bent M et al. MGMT testing--the challenges for biomarker-based glioma treatment. Nat Rev Neurol . 2014; 10:372-85. [PubMed 24912512]

28. Cabrera AR, Kirkpatrick JP, Fiveash JB et al. Radiation therapy for glioblastoma: Executive summary of an American Society for Radiation Oncology Evidence-Based Clinical Practice Guideline. Pract Radiat Oncol . 2016; 6:217-225. [PubMed 27211230]

29. Perry JR, Laperriere N, O'Callaghan CJ et al. Short-Course Radiation plus Temozolomide in Elderly Patients with Glioblastoma. N Engl J Med . 2017; 376:1027-1037. [PubMed 28296618]

30. Sulman EP, Ismaila N, Armstrong TS et al. Radiation Therapy for Glioblastoma: American Society of Clinical Oncology Clinical Practice Guideline Endorsement of the American Society for Radiation Oncology Guideline. J Clin Oncol . 2017; 35:361-369. [PubMed 27893327]

31. Kotecha R, Odia Y, Khosla AA, Ahluwalia MS. Key clinical principles in the management of glioblastoma. JCO Oncol Pract . 2023;19:180-189.

32. van den Bent MJ, Baumert B, Erridge SC, et al. Interim results from the CATNON trial (EORTC study 26053-22054) of treatment with concurrent and adjuvant temozolomide for 1p/19q non-co-deleted anaplastic glioma: a phase 3, randomized, open-label intergroup study. Lancet . 2017;390(10103):1645-53.

33. 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 randomized, open-label, phase 3 study. Lancet Oncol . 2021;22(6):813-23.