Abstract
Optimizing the management of elderly patients with glioblastoma is an ongoing task in neuro-oncology. The number of patients with this tumor type is gradually increasing with the aging of the population. Although available data and practice recommendations remain limited, the current strategy is maximal safe surgical resection followed by radiotherapy in combination with temozolomide. However, survival is significantly worse than that in the younger population. Surgical resection provides survival benefit in patients with good performance status. Hypofractionated radiotherapy decreases toxicities while maintaining therapeutic efficacy, thus improving treatment adherence and subsequently leading to better quality of life. The intensity of these treatments should be balanced with patient-specific factors and consideration of quality of life. This review discusses the current optimal management in terms of efficacy and safety, as well as future perspectives.
Keywords: glioblastoma, elderly, management, surgery, radiotherapy, chemotherapy
The optimal management is maximal safe surgical resection followed by hypofractionated radiotherapy in combination with temozolomide in elderly patients with glioblastoma, in considering patient-specific factors and consideration of quality of life.
Introduction
Glioblastoma is an aggressive primary brain tumor with a median overall survival (OS) of <18 months despite intensive treatment (1,2), falling to <12 months in elderly populations. The peak incidence of glioblastoma is in the 60s to 70s (1–3). The Japanese brain tumor registry (2005–2008) reported that glioblastomas comprised 2006 (12.0%) of 16 683 primary brain tumors and 44.6% of patients were over 65 years old (1). Initial symptoms are focal symptoms (57%), disturbance of consciousness (15%), seizure (14%) and intracranial hypertension (10%) (1). The number of elderly patients with glioblastoma is gradually increasing as the population ages (1–3), although the overall incidence of glioblastoma in the elderly remains stable (4). Elderly populations have been excluded from most clinical trials for glioblastoma because of their poor prognosis, comorbidities and the sensitivity of the aged brain to radiation (5). The cut-off age to define an elderly population differs among clinical studies, ranging between 60 and 75 years. The progression of glioblastoma impairs performance status by reducing functional and cognitive abilities, particularly among the elderly (6).
Current management for glioblastoma comprises surgery, radiotherapy and chemotherapy. For patients younger than 70 years old, the standard approach is maximal safe surgical resection followed by radiotherapy comprising 60 Gy in 30 fractions with concomitant and adjuvant temozolomide (Stupp regimen) (7,8). Some studies have shown that the Stupp regimen prolonged OS in elderly patients with good performance status (5,9–11). In a review of the United States National Cancer Database, prognostic factors were analyzed in elderly patients with glioblastoma (12). Gross total resection (GTR) was associated with the greatest survival benefit, and chemotherapy and radiotherapy also provided survival benefits. These treatment options improved outcomes regardless of performance status (12). However, all these therapies were less frequently used in elderly patients, probably due to the risk of side effects from the treatments. Treatment-related toxicities such as neurocognitive deterioration have thus remained as critical issues. However, temozolomide was found to significantly improve outcomes among elderly patients with glioblastoma showing hypermethylation of the O6-methylguanine-DNA methyltransferase (MGMT) promoter (13). The National Comprehensive Cancer Network (NCCN) guidelines (Version 2.2021) define elderly patients as age > 70 years and encourage active treatments in patients with Karnofsky Performance Status (KPS) scores ≥60 (14). In elderly patients with poor performance status, KPS < 60, NCCN guidelines recommend hypofractionated radiotherapy alone, temozolomide alone or palliative/best supportive care (14).
Conventionally fractionated radiotherapy prolonged survival in elderly populations with glioblastoma, but low completion rates and declines in activities of daily living presented a dilemma (15). Hypofractionated radiotherapy has thus been developed as a means of preserving efficacy while decreasing the toxicities encountered during treatment. Recent studies have demonstrated that 40 Gy administered in 15 fractions and 34 Gy in 10 fractions were both non-inferior to 60 Gy in 30 fractions in terms of OS (16,17). In addition, 25 Gy in five fractions was shown to be non-inferior to 40 Gy in 15 fractions. Concomitant and adjuvant temozolomide increased the survival benefit of radiotherapy with 40 Gy in 15 fractions (13). The Japan Society for Neuro-Oncology guidelines encourage the consideration of hypofractionated radiotherapy for elderly patients with glioblastoma, adding chemotherapy for patient with good performance (18). However, the optimal dose and fractionation with concomitant temozolomide remain unresolved. This review discusses better management of elderly patients with glioblastoma and highlights future perspectives in caring for this disease population.
Surgical resection
Glioblastoma is characterized by extensive invasion into the surrounding brain parenchyma (19). Such tumor invasion means that surgical resection is unable to provide a cure. Magnetic resonance imaging often demonstrates heterogeneous gadolinium-enhanced lesions with edematous changes to the brain parenchyma. Surgical resection of glioblastoma has been evaluated as the resection rate of gadolinium-enhanced lesions in most clinical studies. Biopsy is adopted in patients with tumors impossible to sufficiently resect due to tumor location or general condition. In retrospective studies of adult glioblastoma, a significant survival advantage was associated with extent of resection (EOR) ≥ 78–98% (20,21). Median OS was 13 months in patients with EOR ≥ 98% and 8.8 months in those with <98% in a study conducted at the University of Texas MD Anderson Cancer Center (20). Median OS was 12.8 months, 13.8 months and 16 months with EOR ≥ 80, ≥ 90 and 100%, respectively, in a study at the University of California (21). In a clinical study assessing the effect of fluorescence-guided resection with 5-aminolevulinic acid, complete resection (CR) of enhanced tumor was higher in patients assigned to receive 5-aminolevulinic acid (65%) compared with those assigned white light (36%) (22). Patients allocated 5-aminolevulinic acid displayed a significantly higher 6-month progression-free survival (PFS) rate (41.0%) than those allocated white light (21.1%) (22). However, no prospective trials have evaluated the contribution of EOR to survival.
In the case of elderly patients with glioblastoma, previous studies have indicated that a safe maximal resection may confer modest survival benefits even for patients >65 years old with glioblastoma (23–28) (Table 1). A randomized study at Helsinki University Hospital indicated that median OS was 5.6 months in 10 patients who underwent craniotomy, longer than the 2.8 months in 13 patients who underwent biopsy (27). The procedure-related complication rate was 10% with craniotomy and 0% with biopsy. In a retrospective study of 342 elderly patients ≥65 years old at the University of Freiburg, surgical resection and stereotactic biopsy were performed in 216 patients (63.2%) and 125 patients (36.5%), respectively (24). Median OS was significantly longer in patients with GTR (10.8 months) compared with partial resection (PR) (8.1 months) or biopsy (3.0 months). A systematic review between 2005 and 2018 demonstrated that total resection was associated with longer postoperative OS (13.13 months) when compared with PR (7.52 months) or biopsy (2.56 months) in seven clinical studies (28). In an Italian cohort of 178 elderly patients, CR was seen in 8 patients (4.5%), GTR in 63 (35.4%), subtotal resection (STR) in 46 (25.8%), PR in 16 (9.0%) and biopsy in 45 (25.3%). Postoperative neurological deficits were found in 11 patients (6.2%), and a worsening of preoperative neurological symptoms was recorded in four patients (2.2%). Tumor location, EOR and postoperative neurological status significantly affected survival (26). The analysis of 18 registries in the Surveillance, Epidemiology and End Results (SEER) program revealed that elderly patients with cerebellar glioblastoma and supratentorial glioblastoma have similar outcomes in OS, and those undergoing maximal resection with adjuvant therapies, independent of tumor location, show improved outcomes (25).
Table 1.
Authors, publication year, study design | Diagnosis | Age cut-off, years | Intervention | Number | Adjuvant therapy | mOS, months |
---|---|---|---|---|---|---|
Vuorinen et al. 2003 (27) | Grade IV glioma (83%) | ≥65 | Stereotactic biopsy | 13 | Radiotherapy (16–60 Gy) | 2.8 |
Prospective | Grade III glioma (17%) | Open craniotomy and resection | 10 | 5.6 | ||
Heiland et al. 2018 (24) | GBM | ≥65 | Gross total resection | 142 | Stupp 44, RTa 27, TMZa 58 | 10.8 |
Retrospective | Partial resection | 75 | Stupp 26, RTa 15, TMZa 20 | 8.1 | ||
Biopsy | 125 | Stupp 29, RTa 18, TMZa 45 | 3 | |||
Cunha et al. 2019 (28) | GBM | ≥65 | Total resection | 473 | N/A | 13.13 |
Systematic review | Partial resection | 513 | N/A | 7.52 | ||
Biopsy | 90 | N/A | 2.56 | |||
Pessina et al. 2018 (26) | GBM | ≥65 | Complete resection | 8 | RT 178, concomitant TMZ 149, adjuvant TMZ 132 | 24.5 |
Retrospective | Gross total resection | 63 | 15.1 | |||
Subtotal resection | 46 | 11.9 | ||||
Partial resection | 16 | 8 | ||||
Biopsy | 45 | 8.1 | ||||
Karsy et al. 2018 (30) | GBM | >75 | Gross total resection | 19 | None 17, TMZ 22, RT 32, bevacizumab 7, other 4 | 12.1 |
Retrospective | Subtotal resection | 33 | 5 | |||
Biopsy | 18 | 3.7 | ||||
No surgery | 8 | 0.8 |
mOS: median overall survival; fr: fractions; GBM: glioblatoma; N/A: not available, Stupp; Stupp regimen, RTa; radiotherapy alone, TMZa; temozolomide alone, RT; radiotherapy, TMZ; temozolomide
In patients ≥75 years old, analysis of the SEER database demonstrated a similar result that GTR was an independent predictor of OS, cancer-specific survival and early mortality (29). A retrospective review of 82 patients ≥75 years old at the University of Utah found that survival was associated with EOR only for patients without postoperative complications in EOR including no surgery (9.8%), biopsy (22.0%), STR (40.2%) and gross-total resection (23.2%) (30) (Table 1). Twenty-three patients showed 34 postoperative complications and postoperative complications were identified as an independent risk factor for worsened OS (30). Long-term survivors (≥ 12 months) and short-term survivors (< 12 months) had similar median preoperative KPS, but long-term survivors showed no deterioration in postoperative KPS and no postoperative complications (30). Although no randomized studies have provided strong evidence to evaluate the benefits of surgical resection in elderly patients with newly diagnosed glioblastoma, the existing literature recommends maximal safe resection to prolong OS in elderly patients with good preoperative condition if postoperative complications can be avoided. Surgical indications should be defined based on preoperative performance status instead of biological age.
Radiotherapy
Radiotherapy shows potential as the main postoperative treatment for patients with glioblastoma. Early studies reported that radiotherapy had a significant influence on the survival of patients with malignant glioma in a dose-effect manner (31–33). In a retrospective study conducted at the University of Tokyo, median OS in patients with glioblastoma was 16.2 months with 80–90 Gy and 12.4 months with 60 Gy, but OS did not differ between 80 Gy and 90 Gy. A higher frequency of radiation-induced white matter abnormalities was identified with 80–90 Gy, without incurring increased disability (33). A randomized controlled study by the Medical Research Council Brain Tumor Working Party revealed that 60 Gy in 30 fractions prolonged OS (12 months) compared with 45 Gy in 20 fractions (9 months) (34). As doses exceeding 60 Gy increased morbidity in some studies, 60 Gy in 30 fractions has been selected in most trials for glioblastoma (35).
The disadvantages of 60 Gy in 30 fractions are a prolonged initial treatment period, which is often associated with longer hospitalization, and deterioration of quality of life (QOL) due to radiation-induced brain damage (15). Reducing the burden of treatment while maintaining efficacy is important for elderly patients with glioblastoma, which shows poor prognosis (15). Hypofractionated radiotherapy has thus been evaluated in the elderly population. Three other phase III trials were conducted for further hypofractionated radiotherapy in elderly patients with newly diagnosed glioblastoma. Those studies compared 40 Gy in 15 fractions versus 60 Gy in 30 fractions (16), 34 Gy in 10 fractions versus 60 Gy in 30 fractions (17) and 40 Gy in 15 fractions versus 25 Gy in 5 fractions (36,37), finding non-inferior results in terms of safety or efficacy for elderly patients with newly diagnosed glioblastoma (Table 2) (13,16,17,36,38,39). According to these results, radiotherapeutic regimens of 45 Gy in 20 fractions, 40 Gy in 15 fractions, 34 Gy in 10 fractions and 25 Gy in 5 fractions have been considered to offer similar efficacy and safety for elderly patients with glioblastoma (16,17,36,37). However, all those studies comprised small sample sizes and the results have not been confirmed by other investigations, so the optimal dose and number of fractions remain unclear (15).
Table 2.
Authors, publication year, study name | Diagnosis | Age cut-off, years | Number | Intervention | mPFS, months | mOS, months |
---|---|---|---|---|---|---|
Bleehen et al. 1991 (34) | Astrocytoma | 156 (49 ≥ 60 years) | 45 Gy/20 fr | N/A | 9 (N/A ≥ 60 years) | |
Medical Research Council trial | WHO Grade 3 (33%), 3 + 4 (5%), 4 (61%) | 318 (91 ≥ 60 years) | 60 Gy/30 fr | N/A | 12 (N/A ≥ 60 years) | |
Roa et al. 2004 (16) | GBM | ≥60 | 47 | 60 Gy/30 fr | N/A | 5.1 |
48 | 40 Gy/15 fr | N/A | 5.6 | |||
Keime-Guibert et al. 2007 (38) | AA (2%), GBM (96%) | ≥70 | 42 | Best supportive care | 1.2 | 3.9 |
ANOCEF | 39 | 50 Gy/28 fr | 3.4 | 6.7 | ||
Wick et al. 2012 (39) | AA (11%), GBM (89%) | >65 | 178 | 60 Gy/30 fr | 4.7 | 9.6 |
NOA-08 | 195 | Dose-dense temozolomide | 3.3 | 8.6 | ||
Malmström et al. 2012 (17) | GBM | >70 | 100 | 60 Gy/30 fr | N/A | 6 |
Nordic study | 98 | 34 Gy/10 fr | N/A | 7.5 | ||
93 | Temozolomide | N/A | 8.3 | |||
Roa et al. 2015 (36) | GBM | ≥65 | 50 | 40 Gy/15 fr | 4.2 | 6.4 |
48 | 25 Gy/5 fr | 4.2 | 7.9 | |||
Perry et al. 2017 (13) | GBM | ≥65 | 281 | 40 Gy/15 fr | 3.9 | 7.6 |
CE.6 | 281 | 40 Gy/15 fr + temozolomide | 5.3 | 9.3 | ||
Wirsching et al. 2018 (61) | GBM | ≥65 | 50 | 40 Gy/15 fr + bevacizumab | 7.6 | 12.1 |
ARTE trial | 25 | 40 Gy/15 fr | 4.8 | 12.2 |
mPFS: median progression-free survival; mOS: median overall survival; fr: fractions; N/A: not available; GBM: glioblatoma
An analysis of the United States National Cancer Database between 2005 and 2012 showed that utilization of hypofractionated radiotherapy increased from 7% to 19% during this period in 9556 patients ≥65 years old with glioblastoma, suggesting that hypofractionated radiotherapy may offer better treatment than best supportive care alone and as good as conventional radiotherapy alone (40). In a review of patients ≥65 years old with glioblastoma in the National Cancer Data Base between 2006 and 2012, 126 patients (2.5%) underwent hypofractionated radiotherapy, whereas 5000 (97.5%) received conventional radiotherapy. Patients who underwent hypofractionated radiotherapy were older, showed worse performance status, underwent biopsy only and were more likely to receive treatment at an academic facility. Conventional radiotherapy was associated with improved median OS (10.7 vs. 6.2 months) (41). According to a UK investigation into the management of elderly patients with glioblastoma between 2016 and 2017, median OS was 5.0 months. Approximately 31.9% of patients received combined chemoradiation (42).
Retrospective studies have also demonstrated survival benefits of hypofractionated radiotherapy combined with temozolomide or temozolomide plus bevacizumab (43,44). A phase II study of 52.5 Gy in 15 fractions for elderly patients with poor conditions including age ≥ 70 years and KPS score ≤ 60 showed that median PFS and OS were 5.0 months and 8.0 months, respectively. KPS = 60, recursive partitioning analysis class V, methylated hypermethylation of MGMT promoter, stable neurological status or improvement after surgery, and hypofractionated radiotherapy with concurrent and adjuvant temozolomide were associated with better outcomes (45). In the interim results of a phase II study to evaluate 34 Gy in 10 fractions with temozolomide in patients ≥70 years old with glioblastoma, median PFS and OS were 6 months and not reached on a median follow-up of 9 months, respectively (46). In 2017, the CE.6 trial demonstrated that median OS was longer with radiotherapy comprising 40 Gy in 15 fractions plus concomitant and adjuvant temozolomide (9.3 months) than with 40 Gy in 15 fractions alone in elderly patients ≥65 years old with newly diagnosed glioblastoma (13). However, a select group of elderly patients with excellent performance status and hypermethylation of MGMT promoter or GTR may experience favorable survival with the Stupp regimen (47). The optimal dose and number of fractions thus remain unclear when administered in combination with temozolomide (15).
Chemotherapy
Temozolomide, an oral alkylating agent, exerts antitumor activity as a single agent against malignant glioma (48). The CE.3 trial demonstrated that a regimen of concomitant and adjuvant temozolomide with radiotherapy (Stupp regimen) significantly improved median OS from 12.1 to 14.6 months compared with radiotherapy alone in patients with glioblastoma younger than 70 years old (7,8). However, subgroup analysis of the CE.3 trial revealed a diminishing benefit from the addition of temozolomide with increasing age (hazard ratio 0.80 for 66–70 years) (49). Studies exploring the Stupp regimen in elderly patients with glioblastoma have demonstrated benefits compared with radiotherapy alone, although high frequencies of toxicities such as mental status deterioration and leukoencephalopathy were identified (5,10,50). Good performance status and hypermethylation of MGMT promoter were associated with favorable prognosis from the Stupp regimen in the elderly population. Temozolomide seemed to have minimal impact on seizure control in elderly patients with glioblastoma (51).
The Nordic study demonstrated that temozolomide (200 mg/m2 on Days 1–5 of every 28 days for up to 6 cycles) prolonged median OS from 6.0 months to 8.3 months compared with conventional radiotherapy 60 Gy in 30 fractions (17). In the temozolomide group, patients with hypermethylation of the MGMT promoter displayed significantly longer survival (9.7 months) than those with hypomethylation of the MGMT promoter (6.8 months). The most common grade 3–4 adverse events in the temozolomide group were neutropenia (12%) and thrombocytopenia (21%). The NOA-08 trial demonstrated that the regimen of dose-dense temozolomide (100 mg/m2, given on days 1–7 of a 1 week on, 1 week off cycle) alone is non-inferior to radiotherapy at 54–60 Gy in 30 fractions among elderly patients with anaplastic astrocytoma or glioblastoma and age > 65 years (39). Grade 3–4 adverse events in the temozolomide group were neutropenia (8.2%), lymphocytopenia (23.6%), thrombocytopenia (7.2%), increased liver-enzyme concentrations (15.4%) and thromboembolic events (12.3%). A long-term update of NOA-08 revealed that median OS was 8.2 months in the temozolomide group versus 9.4 months in the radiotherapy group (52). In patients with MGMT methylated tumors, dose-dense temozolomide improved OS from 9.6 to 18.4 months compared with radiotherapy. Pooled analysis of trials controlling for MGMT promoter methylation status demonstrated that temozolomide monotherapy confers similar survival benefits to radiotherapy in combination with temozolomide (53).
The CE.6 trial demonstrated that a regimen of concomitant and adjuvant temozolomide significantly improved median OS from 7.6 to 9.3 months compared with radiotherapy alone in patients ≥65 years old with glioblastoma (13). Addition of temozolomide did not decrease QOL and temozolomide-related adverse events were manageable. Subgroup analysis of the CE.6 trial revealed that patients 65–70 years old displayed less benefit from temozolomide than those 71–75 years old or ≥ 76 years old. As presumptive reasons, fewer patients 65–70 years old than 71–75 years old are enrolled and patients 65–70 years old with good performance status are generally treated with the Stupp regimen. Based on these considerations, a regimen of radiotherapy as 40 Gy in 15 fractions plus temozolomide could represent an appropriate standard treatment for patients ≥71 years old with newly diagnosed glioblastoma (15). Using 14 studies of 4561 patients, a network-based analysis demonstrated that the Stupp regimen provided similar survival benefit to hypofractionated radiotherapy in combination with temozolomide or hypofractionated radiotherapy alone. Recent meta-analyses and systematic reviews have demonstrated that hypofractionated radiotherapy in combination with temozolomide achieved the highest probability of improving survival in older patients with glioblastoma followed by the Stupp regimen (53–57).
Bevacizumab is a humanized monoclonal antibody targeting the vascular endothelial growth factor (VEGF)-A ligand, which inhibits vascular endothelial cell proliferation and angiogenesis (58). Bevacizumab improved PFS, but not OS in patients with newly diagnosed glioblastoma (59,60). The Avastin Plus Radiotherapy in Elderly Patients With Glioblastoma (ARTE) trial demonstrated that the addition of bevacizumab to hypofractionated radiotherapy of 40 Gy in 15 fractions did not prolong OS in 75 elderly patients ≥65 years old (bevacizumab + radiotherapy: 12.1 months; radiotherapy alone: 12.2 months) (61). Adding molecular subtypes into that model identified an association of the RTK II gene methylation subtype with inferior OS. The ANOCEF Phase II Trial suggested that the addition of bevacizumab to temozolomide is active in 66 elderly patients ≥70 years old with glioblastoma with KPS score < 70 with acceptable tolerance (62). Median PFS and OS were 15.3 weeks and 23.9 weeks, respectively. A retrospective study of SEER-Medicare data for patients ≥66 years old between 2006 and 2011 demonstrated that bevacizumab treatment was associated with lower risk of death, suggesting potential benefits of bevacizumab among elderly patients with glioblastoma (63).
Managements of recurrence
No standard treatment has been defined for glioblastoma recurrence, but re-resection, re-irradiation and systemic chemotherapy with bevacizumab or other drugs are selectable options according to patient status, particularly in elderly patients. The majority of glioblastomas in elderly patients recur within 6 months after the initial multimodal therapy (13,17,39), which is a shorter period than in younger patients. An analysis of the SEER-Medicare database reported low re-resection rates, with no survival advantage for those elderly patients who did undergo re-resection (64). Some retrospective studies have demonstrated that elderly patients with recurrent glioblastoma showed a survival benefit from re-resection (65,66). Re-irradiation in elderly patients with glioblastoma was feasible with acceptable safety, offering a possible treatment option. Patients with longer intervals from first-line treatment and patients who received systemic treatments in addition to re-irradiation showed better prognosis (67). In a retrospective study, the efficacy and safety of bevacizumab in recurrent glioblastoma appear similar in elderly and non-elderly patients. However, the clinical benefit seemed less evident in younger patients (68).
Prognostic markers
Performance status is a key prognostic factor to be considered for management decisions in elderly patients with newly diagnosed glioblastoma (69). Retrospective studies identified predictors for poor OS of older age, lower KPS, white people, higher comorbidity score, worse socioeconomic status, community treatment, tumor multifocality, STR, aphasia after surgery, motor dysfunction after surgery and no adjuvant treatment (30,69–74). A Fine-Gray competing risk model of 4975 elderly patients ≥65 years old with glioblastoma from the SEER database demonstrated age ≥ 75 years old, white people, size >5.4 cm, frontal lobe tumor and overlapping lesion were independently associated with more glioblastoma-related death, whereas GTR, chemotherapy and chemoradiation were identified as independently protective factors for glioblastoma-related death (75). In a secondary analysis of the CCTG CE.6 trial evaluating the impact of lymphopenia, development of lymphopenia was not associated with radiotherapy alone, but baseline lymphopenia was associated with worsened OS (76). Multiple socioeconomic parameters can influence access to treatment modalities for elderly patients compared with younger patients in different geographic regions of the United States (77).
The Cumulative Illness Rating Scale (CIRS) is a comprehensive and reliable instrument for assessing physical impairment (78). As high CIRS plays a predictive role for OS in elderly patients with glioblastoma (79), if the prognostic role of comorbidity measured by CIRS on outcome can be confirmed, CIRS offers an interesting scale for optimal treatment according to personal comorbidities. The predictive value of the comprehensive geriatric assessment (CGA) regarding tolerance of chemotherapy and prediction of early mortality was validated for elderly patients with glioblastoma in a retrospective trial (80,81). CGA score offered a good predictor of OS in elderly patients with glioblastoma, which may prove useful in making treatment decisions.
Analyses of the CGGA and TCGA databases detected no age-related hallmarks of glioblastoma including pathological characteristics or mutations (82). However, isocitrate dehydrogenase 1 (IDH1) mutation is rare in the elderly, and more often identified for glioma in young populations (17,83). Glioblastomas in elderly patients possess unique molecular signatures such as telomerase reverse transcriptase promoter mutation, PTEN mutation/deletion, MGMT methylation, high expression of VEGF-A, hypermethylation in polycomb group protein target genes, upregulation of angiogenesis-related genes, somatic copy number alterations and CDK4/MDM2 coamplification (84–89) (Table 3). The genetic analysis in the NOA-08 trial demonstrated an age-independent and stable frequency of MGMT promoter hypermethylation (83). DNA methylation-based classification has recently become a useful tool to classify brain tumors (90). The characterization of molecular subgroups revealed three types of IDH wildtype glioblastoma, indicating subgroups of receptor tyrosine kinase I (RTK I), receptor tyrosine kinase II (RTK II) and mesenchymal. In the NOA-08 trial, MGMT promoter methylation is a strongly predictive biomarker for the choice between radiotherapy and temozolomide. This indicates favorable long-term outcomes with initial temozolomide monotherapy in patients with MGMT promoter-methylated tumors, primarily in the RTK II subgroup (52). Investigation of the DNA methylome (Human Methylation 450 K BeadChip) for age-related associations revealed that acceleration of DNA methylation with age was significantly associated with better outcomes (91). These molecular characteristics provide the possibility of developing age-specific adjuvant treatments.
Table 3.
Authors, publication year | Genetic signature | Possible impact on prognosis |
---|---|---|
Bozdag et al. 2013 (85) | Hypermethylation in polycomb group protein target genes | Poor |
Nghiemphu et al. 2009 (84) Bozdag et al. 2013 (85) | Upregulation of VEGF-A, angiogenesis-related genes | Poor |
Ferguson et al. 2016 (87) Fukai et al., 2020 (89) | PTEN mutation/deletion | Poor |
Wiestler et al. 2013 (83) Ferguson et al. 2016 (87) | IDH1/2 mutation | Good |
Wiestler et al. 2013 (83) | MGMT promoter hypomethylation | Poor |
Eckel-Passow et al. 2015 (86) | TERT promoter mutation | Poor |
Cimino et al. 2018 (88) Fukai et al. 2020 (89) | CDK4/MDM2 coamplification CDK4 amplification/gain | Poor |
Bady et al. 2022 (91) | DNA methylation age acceleration | Good |
Future perspectives
No strong evidence is yet available to establish a ‘best’ regimen for elderly populations with glioblastoma. The goal of treatment in elderly patients with glioblastoma and good performance status is to extend OS preserving KPS, QOL and cognitive function, which is the same in younger patients. In elderly patients with poor performance status, however, the optimal endpoint is considered to improve or preserve their condition. A web-based survey showed that treatment recommendations for elderly patients with newly diagnosed glioblastoma vary widely (92). Statistical comparisons have demonstrated that the common treatment regimens for elderly patients with glioblastoma in previous randomized controlled trials conferred similar survival benefits. Adjustments for the methylation status of MGMT promoter demonstrated that radiotherapy alone was inferior to temozolomide-based treatments (53). A randomized study comparing temozolomide monotherapy with radiotherapy combined with temozolomide is warranted. As a framework for modeling COVID-19 risk on the analysis of five randomized trials, hypofractionated radiotherapy with concurrent and adjuvant temozolomide demonstrated the best outcomes in low- and medium-risk scenarios during the COVID-19 pandemic (93).
Tumor-treating fields represent a locoregional, noninvasive, antimitotic therapy delivering low-intensity, intermediate-frequency alternating electric fields to the tumor. Combining tumor-treating fields (200 kHz) with maintenance temozolomide significantly improved PFS and OS in elderly patients with newly diagnosed glioblastoma in the EF-14 trial, without significantly increasing systemic toxicity or negatively affecting QOL. Tumor-treating fields correlated with low-grade, manageable skin adverse events (94–98). Tumor-treating fields offer an option for elderly patients with good performance status.
A systematic review and network meta-analysis (NMA) also showed that OS estimates from NMA did not provide strong evidence of a difference between different hypofractionated radiotherapies of 40 Gy versus 45 Gy; 34 Gy versus 45 Gy; 25 Gy versus 45 Gy; 34 Gy versus 40 Gy and 25 Gy versus 34 Gy (99). A multi-institutional retrospective study in Korea reported that conventional radiotherapy significantly improved OS compared with short-course radiotherapy in selected elderly patients amenable to chemoradiation (100).
Although concomitant and adjuvant temozolomide is effective in addition to hypofractionated radiotherapy comprising 40 Gy in 15 fractions, one clinical question is which radiation dose fractionation can provide the most survival benefit, particularly in combination with temozolomide, for elderly patients with newly diagnosed glioblastoma.
In August 2020, the Brain Tumor Study Group and Radiation Therapy Study Group of the Japan Clinical Oncology Group (JCOG) started a multi-institutional randomized phase III trial to confirm the non-inferiority of radiotherapy at 25 Gy in 5 fractions with concomitant (150 mg/m2/day, 5 days) and adjuvant temozolomide >40 Gy in 15 fractions with concomitant (75 mg/m2/day, every day from first to last day of radiation) and adjuvant temozolomide in terms of OS for elderly patients with newly diagnosed glioblastoma (JCOG1910, AgedGlio-PIII) (15). The results of JCOG1910 will confirm whether hypofractionated radiotherapy requiring a shorter treatment period with temozolomide can overcome the disadvantages of standard treatment without compromising efficacy (15). If the primary endpoint in JCOG1910 is met, radiotherapy at 25 Gy in five fractions with concomitant and adjuvant temozolomide will be established as a standard of care for elderly patients with newly diagnosed glioblastoma.
On the linear-quadratic (LQ) model, equivalent-dose fractionation can be estimated by calculating equivalent doses in 2-Gy fractions (101). Previous analyses have indicated that the α/β values of glioma and normal brain tissue were 5–10 Gy and 2–3 Gy, respectively (102–104). However, the results of phase III trials have suggested that the α/β value of glioblastoma in elderly patients is estimated as <1.2 Gy (13,17,36) (Table 4). Retrospective analysis of elderly patients ≥65 years old who received resection and hypofractionated radiation with temozolomide demonstrated that 52.5 Gy in 15 fractions was associated with superior OS compared with 40 Gy in 15 fractions (105). If JCOG1910 demonstrates the non-inferiority of radiotherapy of 25 Gy in 5 fractions over 40 Gy in 15 fractions on addition of temozolomide, the α/β value of glioblastoma in elderly patients will be estimated as <1.2 Gy.
Table 4.
α/β value | |||||
---|---|---|---|---|---|
Authors, publication year | Radiation dose/fractions | 1.2 | 3.0 | 5.6 | 10.0 |
Conventional radiotherapy | 60 Gy/30 fr | 60.0 | 60.0 | 60.0 | 60.0 |
Bleehen et al. 1991 (34) | 45 Gy/20 fr | 48.5 | 47.3 | 46.5 | 45.9 |
Perry et al. 2017 (13) | 40 Gy/15 fr | 48.4 | 45.4 | 43.6 | 42.3 |
Roa et al. 2015 (36) | 25 Gy/5 fr | 48.4 | 40.0 | 34.9 | 31.3 |
Malmström et al. 2012 (17) | 34 Gy/10 fr | 48.9 | 43.5 | 40.3 | 38.0 |
Perlow et al. 2022a (105) | 52.5 Gy/15 fr | 77.1 | 68.3 | 62.9 | 59.1 |
A retrospective study of 66 patients
Conclusions
The survival impacts of multimodal treatment differ among elderly patients with impaired performance status, advanced age, severe comorbidities and hypomethylation status of MGMT promoter. Biological age is more important than chronological age. Chronological age should not prohibit multimodal treatment. Instead, optimal management should be considered for the condition of each individual patient to reduce complications and achieve satisfactory QOL. Although hypofractionated radiotherapy in combination with temozolomide following surgery has become the recommended treatment for elderly patients with glioblastoma, several issues remain unresolved. The optimal dose and number of fractions in radiotherapy, particularly in combination with temozolomide, should be explored for better survival and QOL.
Acknowledgements
We thank all members of the JCOG Data Center/Operation office, Brain Tumor Study Group and Radiation Therapy Study Group of the Japan Clinical Oncology Group, and Ms. Keiko Furukawa from the Kyoto University Hospital Cancer Center.
Contributor Information
Yoshiki Arakawa, Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Yohei Mineharu, Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Megumi Uto, Department of Radiation Oncology and Image-Applied Therapy, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Takashi Mizowaki, Department of Radiation Oncology and Image-Applied Therapy, Kyoto University Graduate School of Medicine, Kyoto, Japan.
Availability of data and materials
Data sharing is not applicable for this article, as no datasets have been generated or analyzed at the time of submission.
Funding
This study is supported by the National Cancer Center Research and Development Fund (2020-J-3) and the Japan Agency for Medical Research and Development (AMED 21ck0106619, 22ck0106619).
Contributions
Y.A. wrote the manuscript. All authors critically revised the manuscript for intellectual content and approved the final manuscript.
Consent for publication
Not applicable.
Competing interests
Y.A. received grants from Philips, Otsuka, Chugai, Nihon Medi-Physic, Daiichi Sankyo, Stryker, Eisai, Japan Blood Products Organization, Ono Pharmaceutical, Taiho Pharma, Sumitomo Dainippon Pharma, Astellas Pharma, and Sanofi, honoraria from Nippon Kayaku, Novocure, UCB Japan, Ono Pharmaceutical, Brainlab, Merck, Chugai, Eisai, Daiichi Sankyo, Carl Zeiss, Nihon Medi-Physic. Other authors declare no competing interests.
References
- 1. Brain Tumor Registry of Japan (2005-2008). Neurol Med Chir (Tokyo) 2017;57(Suppl 1):9–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C, Barnholtz-Sloan JS. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2013-2017. Neuro Oncol. 2020;22(12 Suppl 2):iv1-iv96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Brain Tumor Registry of Japan (2001-2004). Neurol Med Chir (Tokyo) 2014;54(Supplement-1):9–102. [Google Scholar]
- 4. Chen B, Chen C, Zhang Y, Xu J. Recent incidence trend of elderly patients with glioblastoma in the United States, 2000-2017. BMC Cancer 2021;21(1):54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Brandes AA, Franceschi E, Tosoni A et al. Temozolomide concomitant and adjuvant to radiotherapy in elderly patients with glioblastoma: correlation with MGMT promoter methylation status. Cancer 2009;115(15):3512–8. [DOI] [PubMed] [Google Scholar]
- 6. Yamawaki R, Nankaku M, Umaba C, et al. Assessment of neurocognitive function in association with WHO grades in gliomas. Clin Neurol Neurosurg 2021;208:106824. [DOI] [PubMed] [Google Scholar]
- 7. 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(5):459–66. [DOI] [PubMed] [Google Scholar]
- 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(10):987–96. [DOI] [PubMed] [Google Scholar]
- 9. Brandes AA, Vastola F, Basso U et al. A prospective study on glioblastoma in the elderly. Cancer 2003;97(3):657–62. [DOI] [PubMed] [Google Scholar]
- 10. Minniti G, De Sanctis V, Muni R et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma in elderly patients. J Neurooncol 2008;88(1):97–103. [DOI] [PubMed] [Google Scholar]
- 11. Fiorica F, Berretta M, Colosimo C et al. Glioblastoma in elderly patients: safety and efficacy of adjuvant radiotherapy with concomitant temozolomide. Arch Gerontol Geriatr 2010;51(1):31–5. [DOI] [PubMed] [Google Scholar]
- 12. Nunna RS, Khalid SI, Patel S, et al. Outcomes and patterns of care in elderly patients with glioblastoma multiforme. World Neurosurg 2021;149:e1026–e37. [DOI] [PubMed] [Google Scholar]
- 13. Perry JR, Laperriere N, O'Callaghan CJ et al. Short-course radiation plus temozolomide in elderly patients with glioblastoma. New Engl J Med 2017;376(11):1027–37. [DOI] [PubMed] [Google Scholar]
- 14. Central Nervous System Cancers Version: 2.2021 . National Comprehensive Cancer Network (NCCN) NCCN clinical practice guidelines in oncology. 2021.
- 15. Arakawa Y, Sasaki K, Mineharu Y et al. A randomized phase III study of short-course radiotherapy combined with Temozolomide in elderly patients with newly diagnosed glioblastoma; Japan clinical oncology group study JCOG1910 (AgedGlio-PIII). BMC Cancer 2021;21(1):1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Roa W, Brasher PM, Bauman G et al. Abbreviated course of radiation therapy in older patients with glioblastoma multiforme: a prospective randomized clinical trial. J Clin Oncol 2004;22(9):1583–8. [DOI] [PubMed] [Google Scholar]
- 17. Malmstrom A, Gronberg 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(9):916–26. [DOI] [PubMed] [Google Scholar]
- 18. Glioblastoma A. Practical Guidelines for Neuro-Oncology 2019. Tokyo, Japan: Kanehara-Shuppan, 2019. [Google Scholar]
- 19. Hirata E, Arakawa Y, Shirahata M et al. Endogenous tenascin-C enhances glioblastoma invasion with reactive change of surrounding brain tissue. Cancer Sci 2009;100(8):1451–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. 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. [DOI] [PubMed] [Google Scholar]
- 21. Sanai N, Polley MY, McDermott MW, Parsa AT, Berger MS. An extent of resection threshold for newly diagnosed glioblastomas. J Neurosurg 2011;115(1):3–8. [DOI] [PubMed] [Google Scholar]
- 22. Stummer W, Pichlmeier U, Meinel T et al. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol 2006;7(5):392–401. [DOI] [PubMed] [Google Scholar]
- 23. Babu R, Komisarow JM, Agarwal VJ et al. Glioblastoma in the elderly: the effect of aggressive and modern therapies on survival. J Neurosurg 2016;124(4):998–1007. [DOI] [PubMed] [Google Scholar]
- 24. Heiland DH, Haaker G, Watzlawick R et al. One decade of glioblastoma multiforme surgery in 342 elderly patients: what have we learned? J Neurooncol 2018;140(2):385–91. [DOI] [PubMed] [Google Scholar]
- 25. Chandra A, Lopez-Rivera V, Dono A, et al. Comparative analysis of survival outcomes and prognostic factors of supratentorial versus cerebellar glioblastoma in the elderly: does location really matter? World Neurosurg 2021;146:e755–e67. [DOI] [PubMed] [Google Scholar]
- 26. Pessina F, Navarria P, Cozzi L et al. Is surgical resection useful in elderly newly diagnosed glioblastoma patients? Outcome evaluation and prognostic factors assessment. Acta Neurochir 2018;160(9):1779–87. [DOI] [PubMed] [Google Scholar]
- 27. Vuorinen V, Hinkka S, Farkkila M, Jaaskelainen J. Debulking or biopsy of malignant glioma in elderly people – a randomised study. Acta Neurochir 2003;145(1):5–10. [DOI] [PubMed] [Google Scholar]
- 28. Cunha M, Esmeraldo ACS, Henriques LAW, Santos M, Jr., Medeiros RTR, Botelho RV. Elderly patients with glioblastoma: the impact of surgical resection extent on survival. Rev Assoc Med Bras (1992). 2019;65(7):937–45. [DOI] [PubMed] [Google Scholar]
- 29. Li T, Liu Y, Li J, Zuo M, Cheng Y. Do elderly patients (>/= 75 years old) with glioblastoma benefit from more radical surgeries in the era of temozolomide? Neurosurg Rev 2022;45(1):741–50. [DOI] [PubMed] [Google Scholar]
- 30. Karsy M, Yoon N, Boettcher L et al. Surgical treatment of glioblastoma in the elderly: the impact of complications. J Neurooncol 2018;138(1):123–32. [DOI] [PubMed] [Google Scholar]
- 31. Walker MD, Strike TA, Sheline GE. An analysis of dose-effect relationship in the radiotherapy of malignant gliomas. Int J Radiat Oncol Biol Phys 1979;5(10):1725–31. [DOI] [PubMed] [Google Scholar]
- 32. Salazar OM, Rubin P, Feldstein ML, Pizzutiello R. High dose radiation therapy in the treatment of malignant gliomas: final report. Int J Radiat Oncol Biol Phys 1979;5(10):1733–40. [DOI] [PubMed] [Google Scholar]
- 33. Tanaka M, Ino Y, Nakagawa K, Tago M, Todo T. High-dose conformal radiotherapy for supratentorial malignant glioma: a historical comparison. Lancet Oncol 2005;6(12):953–60. [DOI] [PubMed] [Google Scholar]
- 34. Bleehen NM, Stenning SP. A Medical Research Council trial of two radiotherapy doses in the treatment of grades 3 and 4 astrocytoma. The Medical Research Council Brain Tumour Working Party. Br J Cancer 1991;64(4):769–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Mornex F, Nayel H, Taillandier L. Radiation therapy for malignant astrocytomas in adults. Radiother Oncol 1993;27(3):181–92. [DOI] [PubMed] [Google Scholar]
- 36. Roa W, Kepka L, Kumar N et al. International atomic energy agency randomized phase iii study of radiation therapy in elderly and/or frail patients with newly diagnosed glioblastoma multiforme. J Clin Oncol 2015;33(35):4145−+. [DOI] [PubMed] [Google Scholar]
- 37. Guedes de Castro D, Matiello J, Roa W et al. Survival outcomes with short-course radiation therapy in elderly patients with glioblastoma: data from a randomized phase 3 trial. Int J Radiat Oncol Biol Phys 2017;98(4):931–8. [DOI] [PubMed] [Google Scholar]
- 38. Keime-Guibert F, Chinot O, Taillandier L et al. Radiotherapy for glioblastoma in the elderly. New Engl J Med 2007;356(15):1527–35. [DOI] [PubMed] [Google Scholar]
- 39. 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(7):707–15. [DOI] [PubMed] [Google Scholar]
- 40. Bingham B, Patel CG, Shinohara ET, Attia A. Utilization of hypofractionated radiotherapy in treatment of glioblastoma multiforme in elderly patients not receiving adjuvant chemoradiotherapy: a National Cancer Database Analysis. J Neurooncol 2018;136(2):385–94. [DOI] [PubMed] [Google Scholar]
- 41. Haque W, Verma V, Butler EB, Teh BS. Patterns of care and outcomes of hypofractionated chemoradiation versus conventionally fractionated chemoradiation for glioblastoma in the elderly population. Am J Clin Oncol 2018;41(2):167–72. [DOI] [PubMed] [Google Scholar]
- 42. Chong MY, Lorimer CF, Mehta S et al. An audit of the management of elderly patients with glioblastoma in the UK: have recent trial results changed treatment? CNS Oncologia 2019;8(4):CNS47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Ohno M, Miyakita Y, Takahashi M et al. Survival benefits of hypofractionated radiotherapy combined with temozolomide or temozolomide plus bevacizumab in elderly patients with glioblastoma aged >/= 75 years. Radiat Oncol 2019;14(1):200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Uto M, Mizowaki T, Ogura K et al. Feasibility evaluation of hypofractionated radiotherapy with concurrent temozolomide in elderly patients with glioblastoma. Int J Clin Oncol 2016;21(6):1023–9. [DOI] [PubMed] [Google Scholar]
- 45. Navarria P, Pessina F, Cozzi L et al. Phase II study of hypofractionated radiation therapy in elderly patients with newly diagnosed glioblastoma with poor prognosis. Tumori 2019;105(1):47–54. [DOI] [PubMed] [Google Scholar]
- 46. Yusuf M, Ugiliweneza B, Amsbaugh M et al. Interim results of a phase II study of hypofractionated radiotherapy with concurrent temozolomide followed by adjuvant temozolomide in patients over 70 years old with newly diagnosed glioblastoma. Oncology 2018;95(1):39–42. [DOI] [PubMed] [Google Scholar]
- 47. Jackson WC, Tsien CI, Junck L et al. Standard dose and dose-escalated radiation therapy are associated with favorable survival in select elderly patients with newly diagnosed glioblastoma. J Neurooncol 2018;138(1):155–62. [DOI] [PubMed] [Google Scholar]
- 48. Newlands ES, Stevens MF, Wedge SR, Wheelhouse RT, Brock C. Temozolomide: a review of its discovery, chemical properties, pre-clinical development and clinical trials. Cancer Treat Rev 1997;23(1):35–61. [DOI] [PubMed] [Google Scholar]
- 49. Laperriere N, Weller M, Stupp R et al. Optimal management of elderly patients with glioblastoma. Cancer Treat Rev 2013;39(4):350–7. [DOI] [PubMed] [Google Scholar]
- 50. Sijben AE, McIntyre JB, Roldan GB et al. Toxicity from chemoradiotherapy in older patients with glioblastoma multiforme. J Neurooncol 2008;89(1):97–103. [DOI] [PubMed] [Google Scholar]
- 51. Climans SA, Brandes AA, Cairncross JG et al. Temozolomide and seizure outcomes in a randomized clinical trial of elderly glioblastoma patients. J Neurooncol 2020;149(1):65–71. [DOI] [PubMed] [Google Scholar]
- 52. Wick A, Kessler T, Platten M et al. Superiority of temozolomide over radiotherapy for elderly patients with RTK II methylation class, MGMT promoter methylated malignant astrocytoma. Neurooncology 2020;22(8):1162–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Nassiri F, Taslimi S, Wang JZ et al. Determining the optimal adjuvant therapy for improving survival in elderly patients with glioblastoma: a systematic review and network meta-analysis. Clin Cancer Res 2020;26(11):2664–72. [DOI] [PubMed] [Google Scholar]
- 54. Kalra B, Kannan S, Gupta T. Optimal adjuvant therapy in elderly glioblastoma: results from a systematic review and network meta-analysis. J Neurooncol 2020;146(2):311–20. [DOI] [PubMed] [Google Scholar]
- 55. Hanna C, Lawrie TA, Rogozinska E, et al. Treatment of newly diagnosed glioblastoma in the elderly: a network meta-analysis. Cochrane Database Syst Rev 2020;3:CD013261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. de Melo SM, Marta GN, Yan M, Cruz C, Moraes FY, Riera R. Management of elderly patients with glioblastoma: current status with a focus on the post-operative radiation therapy. Ann Palliat Med 2020;9(5):3553–61. [DOI] [PubMed] [Google Scholar]
- 57. Lu VM, Kerezoudis P, Brown DA, Burns TC, Quinones-Hinojosa A, Chaichana KL. Hypofractionated versus standard radiation therapy in combination with temozolomide for glioblastoma in the elderly: a meta-analysis. J Neurooncol 2019;143(2):177–85. [DOI] [PubMed] [Google Scholar]
- 58. Ferrara N, Hillan KJ, Novotny W. Bevacizumab (Avastin), a humanized anti-VEGF monoclonal antibody for cancer therapy. Biochem Biophys Res Commun 2005;333(2):328–35. [DOI] [PubMed] [Google Scholar]
- 59. Chinot OL, Wick W, Mason W et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med 2014;370(8):709–22. [DOI] [PubMed] [Google Scholar]
- 60. Gilbert MR, Dignam JJ, Armstrong TS et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med 2014;370(8):699–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Wirsching HG, Tabatabai G, Roelcke U et al. Bevacizumab plus hypofractionated radiotherapy versus radiotherapy alone in elderly patients with glioblastoma: the randomized, open-label, phase II ARTE trial. Ann Oncol 2018;29(6):1423–30. [DOI] [PubMed] [Google Scholar]
- 62. Reyes-Botero G, Cartalat-Carel S, Chinot OL et al. Temozolomide plus bevacizumab in elderly patients with newly diagnosed glioblastoma and poor performance status: an ANOCEF phase II trial (ATAG). Oncologist 2018;23(5):524-e44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Davies J, Reyes-Rivera I, Pattipaka T et al. Survival in elderly glioblastoma patients treated with bevacizumab-based regimens in the United States. Neurooncol Pract. 2018;5(4):251–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Goldman DA, Reiner AS, Diamond EL, DeAngelis LM, Tabar V, Panageas KS. Lack of survival advantage among re-resected elderly glioblastoma patients: a SEER-Medicare study. Neurooncol Adv. 2021;3(1):vdaa159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Farina Nunez MT, Franco P, Cipriani D et al. Resection of recurrent glioblastoma multiforme in elderly patients: a pseudo-randomized analysis revealed clinical benefit. J Neurooncol 2020;146(2):381–7. [DOI] [PubMed] [Google Scholar]
- 66. Hager J, Herrmann E, Kammerer S, et al. Impact of resection on overall survival of recurrent glioblastoma in elderly patients. Clin Neurol Neurosurg 2018;174:21–5. [DOI] [PubMed] [Google Scholar]
- 67. Straube C, Antoni S, Gempt J et al. Re-irradiation in elderly patients with glioblastoma: a single institution experience. J Neurooncol 2019;142(2):327–35. [DOI] [PubMed] [Google Scholar]
- 68. Barrascout E, Lamuraglia M. Glioblastoma and bevacizumab in elderly patients: monocentric study. J Oncol Pharm Pract 2021;27(4):842–6. [DOI] [PubMed] [Google Scholar]
- 69. Al Feghali KA, Buszek SM, Elhalawani H, Chevli N, Allen PK, Chung C. Real-world evaluation of the impact of radiotherapy and chemotherapy in elderly patients with glioblastoma based on age and performance status. Neurooncol Pract 2021;8(2):199–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Mir T, Pond G, Greenspoon JN. Outcomes in elderly patients with glioblastoma multiforme treated with short-course radiation alone compared to short-course radiation and concurrent and adjuvant temozolomide based on performance status and extent of resection. Curr Oncol 2021;28(4):2399–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Dobran M, Nasi D, Della Costanza M et al. Characteristics of treatment and outcome in elderly patients with brain glioblastoma: a retrospective analysis of case series. Acta Clin Croat 2019;58(2):221–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Straube C, Kessel KA, Antoni S et al. A balanced score to predict survival of elderly patients newly diagnosed with glioblastoma. Radiat Oncol 2020;15(1):97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Ius T, Somma T, Altieri R et al. Is age an additional factor in the treatment of elderly patients with glioblastoma? A new stratification model: an Italian Multicenter Study. Neurosurg Focus 2020;49(4):E13. [DOI] [PubMed] [Google Scholar]
- 74. Voisin MR, Sasikumar S, Zadeh G. Predictors of survival in elderly patients undergoing surgery for glioblastoma. Neurooncol Adv 2021;3(1):vdab083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Liu ZY, Feng SS, Zhang YH et al. Competing risk model to determine the prognostic factors and treatment strategies for elderly patients with glioblastoma. Sci Rep 2021;11(1):9321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Song AJ, Ding K, Alnahhas I et al. Impact of lymphopenia on survival for elderly patients with glioblastoma: a secondary analysis of the CCTG CE.6 (EORTC 26062-22061, TROG03.01) randomized clinical trial. Neurooncol Adv 2021;3(1):vdab153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Lu VM, Lewis CT, Esquenazi Y. Geographic and socioeconomic considerations for glioblastoma treatment in the elderly at a national level: a US perspective. Neurooncol Pract 2020;7(5):522–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Linn BS, Linn MW, Gurel L. Cumulative illness rating scale. J Am Geriatr Soc 1968;16(5):622–6. [DOI] [PubMed] [Google Scholar]
- 79. Villani V, Tanzilli A, Telera SM et al. Comorbidities in elderly patients with glioblastoma: a field-practice study. Future Oncol 2019;15(8):841–50. [DOI] [PubMed] [Google Scholar]
- 80. Lutgendorf-Caucig C, Freyschlag C, Masel EK, Marosi C. Guiding treatment choices for elderly patients with glioblastoma by a comprehensive geriatric assessment. Curr Oncol Rep 2020;22(9):93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Lombardi G, Bergo E, Caccese M et al. Validation of the comprehensive geriatric assessment as a predictor of mortality in elderly glioblastoma patients. Cancer 2019;11(10):1509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Li G, Zhai Y, Wang Z et al. Postoperative standard chemoradiotherapy benefits primary glioblastoma patients of all ages. Cancer Med 2020;9(6):1955–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Wiestler B, Claus R, Hartlieb SA et al. Malignant astrocytomas of elderly patients lack favorable molecular markers: an analysis of the NOA-08 study collective. Neuro Oncol 2013;15(8):1017–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Nghiemphu PL, Liu W, Lee Y et al. Bevacizumab and chemotherapy for recurrent glioblastoma: a single-institution experience. Neurology 2009;72(14):1217–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Bozdag S, Li AG, Riddick G et al. Age-specific signatures of glioblastoma at the genomic, genetic, and epigenetic levels. PLoS One 2013;8(4):e62982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Eckel-Passow JE, Lachance DH, Molinaro AM et al. Glioma groups based on 1p/19q, IDH, and TERT promoter mutations in tumors. N Engl J Med 2015;372(26):2499–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Ferguson SD, Xiu J, Weathers SP et al. GBM-associated mutations and altered protein expression are more common in young patients. Oncotarget 2016;7(43):69466–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Cimino PJ, McFerrin L, Wirsching HG et al. Copy number profiling across glioblastoma populations has implications for clinical trial design. Neuro Oncol 2018;20(10):1368–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Fukai J, Arita H, Umehara T et al. Molecular characteristics and clinical outcomes of elderly patients with IDH-wildtype glioblastomas: comparative study of older and younger cases in Kansai Network cohort. Brain Tumor Pathol 2020;37(2):50–9. [DOI] [PubMed] [Google Scholar]
- 90. Capper D, Jones DTW, Sill M et al. DNA methylation-based classification of central nervous system tumours. Nature 2018;555(7697):469–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Bady P, Marosi C, Weller M, et al. DNA methylation-based age acceleration observed in IDH wild-type glioblastoma is associated with better outcome - including in elderly patients. medRxiv 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Palmer JD, Bhamidipati D, Mehta M et al. Treatment recommendations for elderly patients with newly diagnosed glioblastoma lack worldwide consensus. J Neurooncol 2018;140(2):421–6. [DOI] [PubMed] [Google Scholar]
- 93. Tabrizi S, Trippa L, Cagney D, et al. A quantitative framework for modeling COVID-19 risk during adjuvant therapy using published randomized trials of glioblastoma in the elderly. Neuro Oncol 2020;22(7):918–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Ram Z, Kim CY, Hottinger AF, Idbaih A, Nicholas G, Zhu JJ. Efficacy and safety of tumor treating fields (TTFields) in elderly patients with newly diagnosed glioblastoma: subgroup analysis of the phase 3 EF-14 clinical trial. Front Oncol 2021;11:671972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Stupp R, Taillibert S, Kanner AA et al. Maintenance therapy with tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma: a randomized clinical trial. JAMA 2015;314(23):2535–43. [DOI] [PubMed] [Google Scholar]
- 96. Stupp R, Taillibert S, Kanner A et al. Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. JAMA 2017;318(23):2306–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Taphoorn MJB, Dirven L, Kanner AA et al. Influence of treatment with tumor-treating fields on health-related quality of life of patients with newly diagnosed glioblastoma: a secondary analysis of a randomized clinical trial. JAMA Oncol 2018;4(4):495–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Zhu JJ, Demireva P, Kanner AA et al. Health-related quality of life, cognitive screening, and functional status in a randomized phase III trial (EF-14) of tumor treating fields with temozolomide compared to temozolomide alone in newly diagnosed glioblastoma. J Neurooncol 2017;135(3):545–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Melo SM, Marta GN, Latorraca COC, Martins CB, Efthimiou O, Riera R. Hypofractionated radiotherapy for newly diagnosed elderly glioblastoma patients: a systematic review and network meta-analysis. PLoS One 2021;16(11):e0257384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Wee CW, Kim IH, Park CK et al. Chemoradiation in elderly patients with glioblastoma from the multi-institutional GBM-molRPA cohort: is short-course radiotherapy enough or is it a matter of selection? J Neurooncol 2020;148(1):57–65. [DOI] [PubMed] [Google Scholar]
- 101. van Leeuwen CM, Oei AL, Crezee J et al. The alfa and beta of tumours: a review of parameters of the linear-quadratic model, derived from clinical radiotherapy studies. Radiat Oncol 2018;13(1):96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Barazzuol L, Burnet NG, Jena R, Jones B, Jefferies SJ, Kirkby NF. A mathematical model of brain tumour response to radiotherapy and chemotherapy considering radiobiological aspects. J Theor Biol 2010;262(3):553–65. [DOI] [PubMed] [Google Scholar]
- 103. Jones B, Sanghera P. Estimation of radiobiologic parameters and equivalent radiation dose of cytotoxic chemotherapy in malignant glioma. Int J Radiat Oncol Biol Phys 2007;68(2):441–8. [DOI] [PubMed] [Google Scholar]
- 104. Qi XS, Schultz CJ, Li XA. An estimation of radiobiologic parameters from clinical outcomes for radiation treatment planning of brain tumor. Int J Radiat Oncol Biol Phys 2006;64(5):1570–80. [DOI] [PubMed] [Google Scholar]
- 105. Perlow HK, Yaney A, Yang M, et al. Dose-escalated accelerated hypofractionation for elderly or frail patients with a newly diagnosed glioblastoma. J Neurooncol 2022;156(2):399–406. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable for this article, as no datasets have been generated or analyzed at the time of submission.