11

Current Standards of Care in Glioblastoma Therapy

Catarina Fernandes1 Andreia Costa1 Lígia Osório2 Rita Costa Lago2 Paulo Linhares3,4 Bruno Carvalho3,4 Cláudia Caeiro1

1Department of Medical Oncology, Centro Hospitalar de São João, Porto, Portugal; 2Department of Radiotherapy, Centro Hospitalar de São João, Porto, Portugal; 3Department of Neurosurgery, Centro Hospitalar de São João, Porto, Portugal; 4Faculty of Medicine of the University of Porto, Portugal

Abstract: Glioblastoma (GBM) is the most common primary malignant brain tumor in adults. Regardless of ideal multidisciplinary treatment, including maximal surgical resection, followed by radiotherapy plus concomitant and maintenance temozolomide (TMZ), almost all patients experience tumor progression with nearly universal mortality and a median survival of less than 15 months. The addition of bevacizumab to standard treatment with TMZ revealed no increase in overall survival (OS) but improved progression-free survival (PFS). In newly diagnosed GBM, methylation of the O6-methylguanine-DNA methyltransferase (MGMT) promoter has been shown to predict response to alkylating agents, as well as prognosis. Therefore, MGMT promoter status may have a crucial role in the choice of single modality treatment in fragile elderly population. No standard of care is established in recurrent or progressive GBM. Treatment alternatives may include supportive care, surgery, re-irradiation, systemic therapies, and combined modality therapy. Despite numerous clinical trials, the identification of effective therapies is complex because of the lack of appropriate control arms, selection bias, small sample sizes, and disease heterogeneity. Tumor-treating fields plus TMZ represent a major advance in the field of GBM therapy, and should be considered for patients with newly diagnosed GBM with no contraindications. As a disease with such a poor prognosis, treatment of GBM should go beyond improving survival and aim at preserving and even improving the quality of life of both the patient and the caregiver.

Keywords: Bevacizumab; Glioblastoma; MGMT; Radiotherapy; Temozolomide

Author for correspondence: Catarina Fernandes, Department of Medical Oncology, Centro Hospitalar de São João, Porto, Portugal. E-mail: ana.catarinaf@gmail.com

Doi: http://dx.doi.org/10.15586/codon.glioblastoma.2017.ch11

In: Glioblastoma. Steven De Vleeschouwer (Editor), Codon Publications, Brisbane, Australia ISBN: 978-0-9944381-2-6; Doi: http://dx.doi.org/10.15586/codon.glioblastoma.2017

Copyright: The Authors.

Licence: This open access article is licenced under Creative Commons Attribution-NonCommercial 4.0 International (CC BY 4.0). https://creativecommons.org/licenses/by-nc/4.0/

Introduction

Glioblastoma (GBM) is the most common and devastating primary malignant brain tumor in adults, encompassing 16% of all primary brain and central nervous system neoplasms (1). Regardless of advanced diagnostic modalities and ideal multidisciplinary treatment that includes maximal surgical resection, followed by radiotherapy (RT) plus concomitant and maintenance temozolomide (TMZ) chemotherapy, almost all patients experience tumor progression with nearly universal mortality. The median survival from initial diagnosis is less than 15 months, with a 2-year survival rate of 26–33% (2, 3). The addition of bevacizumab to standard treatment revealed no increase in overall survival (OS), but improved progression-free survival (PFS). That finding caused considerable debate regarding whether the combination is cost-effective in first-line treatment (4, 5). In – newly diagnosed GBM (nGBM), methylation of O6-methylguanine-DNA methyltransferase (MGMT) promoter has been shown to predict response to alkylating agents; its status may play a crucial role in the choice of single modality treatment in fragile elderly population (68).

Currently, no standard of care is established for recurrent or progressive GBM (rGBM) (9). Despite numerous clinical trials, the identification of effective therapies is complex due to the lack of appropriate control arms, selection bias, small sample size, and disease heterogeneity (10). Treatment alternatives may include supportive care, reoperation, re-irradiation, systemic therapies, and combined modality therapy. Therapeutic options need to be carefully weighted, taking into account tumor size and location, previous treatments, age, Karnofsky performance score (KPS), patterns of relapse, and prognostic factors. The association of tumor-treating fields (TTFields) with TMZ represents the first major advance in the field of GBM therapy in approximately a decade and should be considered for newly diagnosed patients with no contraindications (11).As a disease with such a poor prognosis, treatment of GBM should go beyond improving survival and aim at preserving and even improving the quality of life (QoL) of both the patient and the caregiver.

Newly Diagnosed Glioblastoma

SURGERY

Surgery is the initial therapeutic approach for GBM and remains a hallmark in the treatment of malignant brain tumors. Some preoperative issues such as medical conditions of the patient, appropriate imaging and functional studies, neuropsychological evaluation, and the use of corticosteroid and antiepileptic drugs should be taken into account. While steroids can control cerebral edema and symptoms/signs of intracranial hypertension, thus improving brain conditions for surgical resection, antiepileptic drugs should not be used prophylactically (12). In patients with brain tumors who have not had a seizure, tapering and discontinuing anticonvulsants after the first postoperative week is appropriate (12). Attention should be paid to patients who are going to be operated with cortical stimulation, in an asleep–awake–asleep manner, due to the potential development of stimulation-induced seizures. The goals of surgical treatment are: maximal safe resection; tissue specimen for pathological diagnosis; improving conditions for complementary treatments; delaying clinical worsening; and improving QoL.

While strong predictors of good outcome are essentially patient related, the most important treatment-related predictor is extent of resection (EOR) (13). A more extensive surgical resection is associated with longer life expectancy, achieving the longest survival in those patients who undergo gross total resection followed by RT and TMZ (1315). An important issue is the fine balance between the aggressive removal and the preservation of function; so the goal is to achieve maximal safe surgical resection. A postoperative magnetic resonance imaging (MRI) should assess the EOR within 72 h of surgery. MRI after 72 h of surgery cannot be relied upon because of inflammatory postoperative changes. It has been postulated that ≥98% EOR is necessary to improve survival significantly (16). However, Sanai and colleagues showed that, for oncological purposes, resections of 78% of the tumor volume, associated with chemoradiotherapy, already have prognostic advantages (17). More recently, some authors revealed that more important than the EOR is the amount of the residual volume (18, 19).

Tumors located within eloquent cortex pose a particular surgical challenge due to the high risk of postoperative neurological deficits (20). Muller and colleagues, using functional MRI to map the functional cortex, showed that postoperative neurological deficits occurred in 0% of cases in which the resection margins were beyond 2 cm of the eloquent cortex, in 33% of cases when resection margins were within 1 to 2 cm, and in 50% of cases when resection margins were less than 1 cm (21). Intraoperative electrical stimulation mapping with awake craniotomy decreases the risk of novel neurological deficits, while maximizing the EOR (17, 22). A large meta-analysis demonstrated that resections with the use of intraoperative functional mapping were associated with fewer late severe neurological deficits (3.4% vs. 8.2%) and more extensive resection (75% vs. 58%), although the tumors were more frequently in eloquent locations (100% vs. 96%) (23). Motor evoked potentials and somatosensory evoked potentials can also be recorded during surgery to continuously monitor the integrity of motor and somatosensory pathways.

To increase the EOR, enhancing the visualization of the tumor margins, some fluorescent agents have been used, namely the most widely employed 5-aminolevulinic acid (5-ALA). Panciani and colleagues showed that fluorescence-guided resection revealed a sensitivity of 91.4% and a specificity of 89.2% (24). The use of 5-ALA increases the rate of gross total resection, in randomized controlled trials (65% vs. 35%) and in observational studies (from 25 to 94.3%) (25, 26), and also increases PFS (8.6 vs. 4.8 months) and the 6-month PFS (PFS6) (46% vs. 28%) (27, 28). The increase in gross total resection rate and PFS was confirmed by three meta-analyses performed to evaluate the literature on 5-ALA. Fluorescein is another option but was not tested in randomized controlled trials (25). Fluorescence near eloquent areas should be managed carefully. A biopsy should be reserved for patients with multiple comorbidities, who would not be able to tolerate a large cranial surgery, or for those with unresectable tumors, and the only benefit is provision of tissue specimen for pathological diagnosis (29). The surgery also allows relieving of the mass effect with concurrent amelioration of symptoms, in patients with increased intracranial hypertension and brain edema, leading to an improvement in the QoL.

COMPLEMENTARY TREATMENT

The current standard of care for patients with nGBM is maximum safe surgical resection followed by concurrent TMZ (75 mg/m2/day for 6 weeks) and RT (60 Gy in 30 fractions) and then six maintenance cycles of TMZ (150–200 mg/m2/day for the first 5 days of a 28-day cycle—sdTMZ), according to the results of the phase III EORTC 26981 (2). Stupp et al. showed an OS and PFS improvement with the combination therapy relative to RT alone (median OS 14.6 vs. 12.1 months; P < 0.001) (3). These results were supported by other trials (3033). A recently published meta-analysis by Feng et al. revealed a median OS of 13.4–19.0 months in the combination treatment group, as opposed to 7.7–17.1 months in the RT-alone group (34).

Age, neurological status (assessed by KPS and Mini Mental State Examination), EOR, IDH (isocitrate dehydrogenase) mutations, and methylation of the MGMT promoter region are established prognostic factors in GBM patients (35, 36). The predictive role of MGMT promoter methylation in response to TMZ has also been established in several studies (3, 37). Nevertheless, the clinical utility of MGMT remains poor, primarily because of a lack of therapeutic options for patients with unmethylated MGMT promoter GBM. The only exception is in the management of elderly patients with GBM. TMZ is an oral chemotherapeutic drug that induces DNA methylation and tumor cytotoxicity through cell cycle arrest. The cytotoxic activity of TMZ and other alkylating agents is apparent by the formation of O6-methylguanine DNA adducts, which are repaired by the enzyme MGMT. Consequently, the primary mechanism of resistance to TMZ is dependent on the MGMT activity (38). It exhibits a linear pharmacokinetics with excellent bioavailability, readily enters the cerebrospinal fluid, and it does not require hepatic metabolism for activation (39). Although evidence suggests that TMZ chemotherapy is associated with few adverse events, risk of hematological complications, fatigue, and infections were increased with its use (40).

DOSE-DENSE TEMOZOLOMIDE

Dose-dense schedules of TMZ (ddTMZ) have been designed to deplete tumor MGMT levels and thereby improve activity of TMZ, particularly in the MGMT unmethylated GBM cohort (41). In the RTOG 0525 phase III trial, 833 patients were randomized to receive sdTMZ or ddTMZ (75–100 mg/m2 days 1 through 21 of a 28-day cycle), for 6–12 cycles, after completion of concomitant RT-TMZ. The median OS (16.6 vs. 14.9 months; P = 0.63) and the median PFS (5.5 vs. 6.7 months; P = 0.06) were not significantly different between the two treatment arms. There was increased grade ≥3 toxicity in ddTMZ arm (34% vs. 53%; P < 0.001), as well as a greater deterioration on function subscales and QoL (2).

DURATION OF TEMOZOLOMIDE MAINTENANCE THERAPY

RT with concomitant and adjuvant TMZ was initially introduced with six TMZ maintenance cycles (3). In clinical practice, however, many centers continue TMZ therapy beyond six cycles. The impact of this strategy is controversial and has not yet been confirmed in prospective randomized clinical trials. A phase II comparison of 6 versus 12 cycles of TMZ (NCT02209948) is currently underway. Some retrospective studies suggested a benefit in OS with extension of maintenance TMZ (4245). The major limitation of all these studies, beyond the retrospective nature, is the comparison of patients who were treated with at least seven cycles of TMZ to patients receiving ≤6 cycles, who, in most cases, stopped TMZ because of tumor progression. Other limitations are missing information on MGMT methylation and univariate Kaplan–Meier description of OS, but no investigation of significance by multivariate Cox regression (4245). Data from a large pooled analysis of four clinical trials for nGBM indicates that extended treatment with TMZ beyond six cycles is not associated with improved OS, but prolongs PFS (2, 3, 4648). A similar analysis was performed in patients enrolled in the German Glioma Network. A total of 61 of the 142 identified patients received at least seven maintenance TMZ cycles (median 11, range 7–20). Patients with extended maintenance TMZ treatment had better PFS (20.5 months vs. 17.2 months; P = 0.035) but not OS (32.6 months vs. 33.2 months; P = 0.126). However, there was no significant association of prolonged TMZ chemotherapy with PFS or OS adjusted for age, EOR, KPS, presence of residual tumor, MGMT promoter methylation status, or IDH mutation status (49). This study provides Class III evidence that in patients with nGBM, prolonged TMZ chemotherapy does not significantly increase PFS or OS.

GLIADEL (CARMUSTINE) IMPLANTABLE WAFERS

Biodegradable carmustine wafers, implanted into the tumor bed, after near or complete tumor resection, has been approved by the FDA for first-line treatment of GBM and anaplastic glioma. Nevertheless, the use of carmustine wafers remains controversial due to the questionable survival benefit and potential adverse events (50).

OPTIMAL DOSE-FRACTIONATION SCHEDULE FOR EXTERNAL BEAM RADIATION THERAPY

For patients aged under 70 years with good PS (KPS ≥ 60), the optimal dose-fractionation schedule for external beam RT, following resection or biopsy, is 60 Gy in 2 Gy fractions delivered over 6 weeks. Numerous other dose schedules have been explored without clear benefits. Attention must be paid to ensure that dose to critical structures (such as brainstem, optic chiasm, optic nerves) is kept within acceptable limits. Risk of radiation necrosis increases with concurrent chemotherapy and larger volume of irradiated brain. The QUANTEC authors emphasize that for most brain tumors, there is no clinical indication to give fractionated RT > 60 Gy (51).

TARGETED THERAPY—IS THERE A PLACE IN NEWLY DIAGNOSED GLIOBLASTOMA?

Since GBM is one of the most vascularized tumors, antiangiogenic therapeutic strategies are very attractive. Bevacizumab is a monoclonal antibody that binds to circulating VEGF-A and inhibits its biological activity by preventing the interaction with the VEGF receptor. This leads to a reduction in endothelial proliferation and vascular growth within the tumor (52). Bevacizumab was approved by the FDA, based on unprecedented response rates (RRs) in rGBM, which led to its evaluation in the postoperative setting of nGBM (Table 1) (64, 65). Two large phase III, randomized, placebo-controlled trials, adding bevacizumab to standard treatment were conducted (4, 5). In the RTOG 0825 trial, median PFS was increased (10.7 vs. 7.3 months), although it did not reach the predefined significance level, and there was no difference in OS between the two treatment groups. The MGMT methylation status and recursive partitioning analysis (RPA) class were prognostic regardless of the study treatment. A decline in QoL and neurocognitive function (NCF) was more frequently observed with bevacizumab (5). In the AVAglio trial, there was a significant increase in PFS (10.6 vs. 6.2 months; P < 0.001), but not in OS. Baseline health-related QoL and PS were maintained longer in the bevacizumab group (4). Grade 3 or 4 toxicities occurred more often in the bevacizumab arms of both studies. In RTOG 0825, the crossover from placebo to bevacizumab, at disease progression, was planned and occurred in 48.3% of patients, and in AVAglio, the crossover was about 30%. This may have eliminated a potential survival benefit (4, 5). In both RTOG 0825 and AVAglio, efforts have been made to identify a subset of patients who could benefit from upfront treatment with bevacizumab, but no marker proved consistently effective in predicting either response or resistance to bevacizumab (4, 5).

TABLE 1 Clinical trials with bevacizumab in the newly diagnosed glioblastoma

Reference Arm 1 Arm 2 Phase n RR (%) Arm 1 versus Arm 2 PFS (months) Arm 1 versus Arm 2 PFS6 (%) Arm 1 versus Arm 2 OS (months) Arm 1 versus Arm 2
RTOG 0825 trial (5) BVZ/TMZ + RT→TMZ/BVZ Placebo/TMZ + RT→TMZ/placebo III 637 10.7 vs. 7.3
(P < 0.007)
15.7 vs. 16.1
(P = 0.21)
AVAglio trial (4) BVZ/TMZ + RT→TMZ/BVZ Placebo/TMZ + RT→TMZ/placebo III 921 10.6 vs. 6.2
(P < 0.001)
16.8 vs. 16.7
(P = 0.10)
(53, 54) BVZ + RT →BVZ/IRI TMZ + RT ® TMZ III
(MGMT-nonmethylated)
171 9.7 vs. 5.9
(P = 0.0004)
80 vs. 41
(P < 0.0001)
16.6 vs. 17.3
(P = 0.9)
(55) BVZ/TMZ + hypo-IMRT→TMZ/BVZ TMZ+ hypo-IMRT→TMZ II   56 12.8 vs. 9.4
(P = 0.58)
84 vs. 83
(P = 0.702)
16.3 in both
(56) BVZ/TMZ + hypo-RT→TMZ/BVZ II   40 57 10 93 (1 year) 19
(57) TMZ + RT →TMZ/BVZ II   62 8.8 16.5
(58) TMZ + RT →BVZ/EVE II
(MGMT-nonmethylated)
  48 7.3 14.2
(59) BVZ/TMZ + RT→TMZ/BVZ II   51 13.0 85.1 23.0
(60) BVZ/TMZ + RT→BVZ/EVE II   68 61 11.3 13.9
(61) BVZ/TMZ + RT→TMZ/BVZ/TOP II   80 11.1 17.2
(62) BVZ/TMZ + RT→TMZ/BVZ/IRI II   75 14.2 21.2
(63) BVZ/TMZ + RT→TMZ/BVZ II   70 13.6 88 19.6

BVZ, bevacizumab; EVE, everolimus; hypo-IMRT, hypofractionated-intensity-modulated RT; IRI, irinotecan; OS, overall survival; PFS, progression-free survival; PFS6, 6-month PFS; RR, Response Rate; RT, radiotherapy; TMZ, temozolomide; TOP, topotecan.

In summary, these trials have shown that the combination of bevacizumab with standard RT–TMZ for the treatment of nGBM resulted in improved median PFS, without gain in OS. Data regarding QoL and functional status are contradictory. Not surprisingly, there was an increase in adverse events associated with bevacizumab therapy. Cilengitide, a selective αvβ3-αvβ5-integrin inhibitor, had shown promising results in phase II trials, with more pronounced benefits in GBM with methylated MGMT promoter (6668). Two prospective randomized trials evaluated the role of cilengitide in combination with standard treatment, in patients with a methylated MGMT gene promoter (CENTRIC) and in those with an unmethylated MGMT status (CORE). They both failed in demonstrating an OS gain (47, 48). Other agents, namely enzastaurin and temsirolimus, have been studied in phase II trials, without any improvement in OS or PFS (41).

Recurrent Glioblastoma

SURGERY AT RECURRENCE

When tumor recurs, treatment options include supportive care, reoperation, re-irradiation, systemic therapies, and combined modality therapy. In this setting, the role of reoperation remains unclear. A recent review of the literature, including 28 studies and 2279 patients, who underwent second surgery, showed a median survival from reoperation of 9.7 months and concluded that EOR at reoperation improves OS, even in patients with subtotal resection at initial surgery (69). Nonetheless, clinical and survival benefit is dependent on patient and tumor characteristics, which need to be considered before pursuing a second surgery. The most consistently demonstrated prognostic factor is favorable PS (KPS ≥ 70), which associates with significantly improved PFS and OS, following salvage therapy (7076). Younger age is the second most frequently reported prognostic factor associated with improved survival (70, 72, 77, 78). Park et al. have devised a scale to predict survival after reoperation based on tumor involvement of pre-specified eloquent/critical brain regions (MSM, motor–speech–middle cerebral artery score), KPS score of 80, and tumor volume (50 cm3). The scale identified three statistically distinct groups within the validation cohort as well (median survival of 9.2, 6.3, and 1.9 months, respectively) (76). Recently, a new 3-tier scale was developed, including KPS score of 70 and ependymal involvement, allowing identification of groups of patients with significant differences in median OS after reoperation (79). Maximal tumor volume resection should be the surgical goal even in candidates for a second surgery. In this perspective, involvement of eloquent brain usually precludes this objective and is associated with shorter OS (15, 80). Molecular markers’ impact in rGBM is still a matter of debate. Brandes et al. reported that MGMT methylation status determined at first surgery seems to be of prognostic value, although it is not predictive of outcome after the second surgery (81).

The DIRECTOR trial, although not aimed at addressing the reoperation issue, allowed the retrospective analysis of EOR and residual tumor volume in approximately two-thirds of the patients, who underwent surgery prior to study entry. Complete resection of enhancing tumor was achieved in 68% of the patients, and in multivariate analysis it was found to be an independent predictor for post-resection survival (82). A multicenter retrospective study, including 503 patients with rGBM submitted to reoperation, concluded that preoperative and postoperative KPS, EOR of first re-resection, and chemotherapy after first re-resection significantly influenced survival after reoperation. Importantly, this study reported a rate of permanent new deficits after first re-resection of 8% (83). In conclusion, evidence suggests higher OS in selected patients who undergo reoperation at the time of GBM recurrence. It should be considered in patients with a good KPS and a favorable preoperative clinical and radiological characteristics. Age <60 years and KPS ≥70 are particularly associated with better outcome. Of paramount importance are the preservation of eloquent brain areas and the avoidance of neurological deterioration after second surgery, since that might mitigate the expected survival benefit.

RE-IRRADIATION AND SPECIAL TECHNIQUES

The majority of studies on re-irradiation of gliomas are retrospective and they use a variety of techniques, including brachytherapy, fractionated stereotactic RT (FSRT), radiosurgery, and conformal or intensity-modulated RT, with or without new systemic agents. Furthermore, the published data include a wide range of doses, emphasizing the fact that no standard approach exists (84). Inter-study comparison is difficult because studies have heterogeneous samples, different endpoints, and some patients were treated at first and others at second or third progression. Although the biology of re-irradiation remains to be fully understood, there is now a large body of clinical and animal data that can guide recommendations. Mayer and Sminia identified and analyzed 21 studies on re-irradiation of gliomas (85). They opined that the incidence of toxicity, including radionecrosis, may be underreported, since only symptomatic necrosis is likely to be recorded. The major factor contributing to necrosis was the total dose received. There was no correlation between time to re-irradiation and its development, although the minimum time interval between treatments was 3 months. They concluded that the incidence of necrosis did not increase significantly until the total cumulative dose was 100 Gy. In younger patients with good PS, focal reirradiation (stereotactic radiosurgery, SRS; hypofractionated stereotactic radiotherapy, HFSRT) for rGBM may improve outcomes compared to supportive care or systemic therapy alone. Tumor size and location should be taken in to account, when evaluating safety of re-irradiation.

STEREOTACTIC RADIOSURGERY AND HYPOFRACTIONATED STEREOTACTIC RADIOTHERAPY

Since most recurrences occur within brains previously irradiated with a high dose, re-irradiation with doses and margins used in the primary treatment of GBM could confer high toxicity risks. Thus, limited volume re-irradiation using SRS or HFSRT is often employed. Stereotactic methods offer optimal precision of target definition while sparing dose to the surrounding tissues. Both SRS and HFSRT deliver more than 2 Gy per fraction and typically have smaller margins and much shorter durations than conventionally fractionated radiotherapy (cfRT). RTOG 90-05, a phase I dose escalation study, established maximum tolerated doses and demonstrated that single-fraction SRS could be performed, in this setting, with acceptable morbidity (86). In the rare event that disease recurs in a portion of brain not previously irradiated, cfRT with chemotherapy should be considered, after surgery.

SRS and HFSRT appear to provide promising outcomes compared to chemotherapy alone for the treatment of rGBM. Shepherd et al. described 29 recurrent high-grade glioma patients treated with a diversity of HFSRT doses with a median OS of 11 months (87). This compared favorably to a matched cohort of patients treated with nitrosourea chemotherapy, with a median OS of 7 months. The studies were nearly all retrospective, however, lacking randomized control groups and with inherent selection bias limiting conclusions. Several of the early studies involving single-fraction SRS reported high rates of radiation necrosis requiring reoperation (20–40%) (86, 8890). Compared to SRS, the use of HFSRT may help to mitigate the risk of adverse radiation events. A series of 105 GBM patients treated with 35 Gy in 10 fractions had a median survival, from salvage HFSRT, of 11 months, without clinically significant acute morbidity and only one case of late grade 3CNS toxicity (78). However, no direct comparison between salvage SRS and HFSRT is available. Defining target volumes for SRS and HFSRT is controversial and variable. A variety of dose-fractionation regimens, target volumes, and stereotactic systems have been used in the treatment of rGBM. These approaches have not been subjected to randomized comparison, so the optimum technique is yet to be established.

CONVENTIONALLY FRACTIONATED RADIATION

Despite most studies discussing re-irradiation with SRS or HFSRT focus, cfRT may theoretically allow more generous target volumes. A large retrospective series of 172 recurrent glioma patients included 59 patients with GBM, who attained a median survival of 8 months, with only one patient developing radiation necrosis (91). The median dose was 36 Gy (15–62 Gy; 2 Gy/day) and was delivered to the enhancing volume plus a 0,5–1 cm margin. There are not enough clinical data available to recommend cfRT for routine use in the recurrent setting. Practitioners using large-volume re-irradiation should take into account brain tolerance data to reduce the risk of radionecrosis (51).

BRACHYTHERAPY

Brachytherapy has also been evaluated for use in rGBM. Typically performed after resection of recurrent disease, brachytherapy features a sharp dose gradient. Strategies include permanent iodine 125 (I-125) seeds and a silicone balloon catheter system containing I-125 solution. Retrospective studies on I-125 have demonstrated median survivals, from the time of brachytherapy, ranging from 11 to 15 months (92). A review by Combs et al. reported high reoperation rates and radionecrosis incidence (93). It should be noted that patients that are selected for brachytherapy are normally those with resectable tumors, good PS, and small volume of disease. As given in the literature on SRS, selection bias confuses interpretation. Also, the patients receiving brachytherapy need to be healthy enough to undergo surgery and, generally, have localized rather than diffuse recurrences.

COMBINATION TREATMENT

Several studies have addressed the combination of chemotherapy with re-irradiation. A few studies have explored TMZ, given its efficacy at radiosensitization in the upfront treatment of GBM. TMZ plus re-irradiation has been found to be safe and effective. Other studies have explored the addition of bevacizumab, which may block hypoxia factor-mediated angiogenesis, which is upregulated by RT (9496). Moreover, bevacizumab has been used to treat radionecrosis and may reduce its risk following re-irradiation (9799). A few small studies have investigated concurrent TMZ and SRS or FSRT. Median OS ranged between 8 and 9.7 months. Regarding toxicity, it was mild in one study, while neurologic toxicity was reported in two other studies (8–13%) (89, 100, 101). Several studies have investigated adding bevacizumab to SRS (72, 88, 102105). A prospective trial at Memorial-Sloan Kettering Cancer Center, investigating the safety of SRS (30 Gy in 5 fractions) with bevacizumab, reported no radionecrosis among 25 recurrent malignant glioma patients, but three patients discontinued treatment because of bevacizumab grade 3 related toxicity. They documented a 50% RR in the GBM population and a median OS of 12.5 months (102, 106). Another prospective study, in 15 patients with recurrent malignant gliomas, reported one grade 3 and no grade 4–5 toxicities, while QoL and neurocognition were well maintained (88). Median OS from SRS was 14.4 months. A retrospective study from Duke University, in 63 recurrent malignant glioma patients, found that median survival was longer for those who received bevacizumab around the time of SRS, than those who did not (11 vs. 4 months for GBM patients, P = 0.014) (72). Several studies have reported relatively low rates of adverse radiation events in patients treated with bevacizumab and SRS/HFSRT (72). Minniti et al. combined HFSRT (25 Gy in five fractions) with bevacizumab or fotemustine and described significantly better OS and PFS in the bevacizumab cohort (107). These studies are nonrandomized, so selection bias remains a serious concern and additional data are required.

SECOND-LINE CHEMOTHERAPY

Several chemotherapy options are available for second-line treatment, but no standard of care has been established. Comparing results between the various studies, particularly the older ones, is difficult, given the heterogeneity of inclusion criteria, patient characteristics, and choice of endpoints and response criteria. Many trials included patients with anaplastic gliomas (WHO grade 3) and GBM (WHO grade 4). Trials conducted prior to the establishment of standard first-line TMZ chemoradiotherapy often included patients not pretreated with TMZ. In addition, most studies were noncomparative, or did not include an adequate control arm. Most considered the PFS6 and the median OS since recurrence as the primary end points. Although PFS6 is considered a reliable measure of tumor control and a strong predictor of survival, this is influenced by other rescue therapies (108). Regarding radiological response assessments, they were often incompletely reported, with most using the Macdonald criteria. The following sections will describe the most relevant trials performed to date with respect to the medical treatment of rGBM.

NITROSOUREA MONOTHERAPY AND COMBINATION REGIMENS

Nitrosoureas are DNA alkylating agents, namely carmustine (BCNU), lomustine (CCNU), nimustine (ACNU), and fotemustine. They are characterized by high lipophilicity and thus can cross the blood–brain barrier, making them useful in the treatment of brain tumors such as GBM (109). Table 2 summarizes nitrosourea-based trials in rGBM. Nitrosoureas, particularly BCNU, were the chemotherapeutic agents of choice for first-line treatment of GBM in the 1970s and 1980s. Based on two phase II trials, TMZ was approved for recurrent high-grade gliomas, and nitrosoureas were relocated into second-line therapy (126, 127). Two phase II trials and a retrospective study assessed the efficacy of BCNU monotherapy regimens in rGBM (110, 118, 125). They reported a PFS6 and a median OS of 13.0–17.5% and 5.1–7.5 months, respectively. RRs were limited and no complete remission was observed. The predominant side effects were hematologic and long-lasting hepatic and pulmonary toxicity. Although BCNU regimens have shown similar efficacy to other cytotoxic therapies, toxicity can be substantial, and the patient recovers slowly, such that the administration of other drugs in the case of further tumor progression can be infeasible (110). BCNU was also evaluated in combination with other agents, such as irinotecan and TMZ, in two phase II studies, with a median OS of 7.8–11.7 months (115, 116). These data demonstrate that BCNU is an effective agent in the treatment of rGBM, but at present its use in clinical practice is limited.

TABLE 2 Retrospective studies and clinical trials with nitrosourea in recurrent GBM (nitrosourea + bevacizumab combination studies are described in Table 4)

Reference Arm 1 Arm 2 Arm 3 Previous TMZ Relapse n Phase RR (%) Arm 1 vs. 2 vs. 3 PFS (months) Arm 1 vs. 2 vs. 3 PFS6 (%) Arm 1 vs. 2 vs. 3 OS (months) Arm 1 vs. 2 vs. 3
Single-arm monotherapy trials
(110) BCNU No 1st   40 II 15   17.5   7.5
(111) FOT Yes 1st   27 II 30 5.7   48.2   9.1
(112) FOT Yes 1st   43 II   7 1.7   20.9   6.0
(113) FOT Yes 1st   50 II 18 6.1 52   8.1
(114) FOT Yes 1st   40 II 25 6.1 61 11.1
Single-arm combination therapy trials
(115) BCNU + IRI Yes 1st   42 II   21.4 3.9   30.3 11.7
(116) BCNU + TMZ No NA   38 II     5.6 2.5 21   7.8
(117) FOT + TMZ Yes NA   20 Prospect. NA 4.3 40 NA
Randomized trials
(118) BCNU or TMZ ERL Some (number of pts NA) 1st 106 II 10 vs. 4 2.4 vs. 1.8 24.1 vs. 11.4 7.3 vs. 7.7
(119) CCNU Enzastaurin NA 1st, 2nd 265 III (2:1) 4 vs. 3 1.6 vs. 1.5 19 vs. 11 7.1 vs. 6.6
(120) CCNU + P CED CCNU + CED Yes 1st 325 III (1:2:2) 8 vs. 14 vs. 16 2.7 vs. 3.1 vs. 4.2 24.5 vs. 16 vs. 34.5 9.8 vs. 8.0 vs. 9.4
(121) CCNU + Galun Galun CCNU + P NA NA 158 II (2:1:1) 1 vs. 5 vs. 0 1.8 vs. 1.8 vs. 1.9 6.7 vs. 8.0 vs. 7.5
Retrospective studies
(122) PCV 4 pts NA   63 Retrosp.   17.5 NA 29 7.7
(123) PCV 12 pts 1st   86 Retrosp.     3.5 4.0   38.4 7.8
(124) ACNU ± combinationa Yes ≥1st   32 Retrosp.     6.3 2.7 20 6.7
(125) BCNU 24 pts 1st, 2nd, 4th   35 Retrosp.     6.2 2.6 13 5.1

ACNU, nimustine; BCNU, carmustine; CCNU, lomustine; CED, cediranib; ERL, erlotinib; FOT, fotemustine; Galun, galunisertib; IRI, irinotecan; NA, not available; OS, median overall survival; P, placebo; PCV, procarbazine-lomustine-vincristine; PFS, median progression-free survival; PFS6, 6-month PFS; Pts, patients; Retrosp., retrospective; RR, response rate; TMZ, temozolomide.

aCombination with teniposide (n = 17) or cytarabine (n = 1).

In a small retrospective study, with 32 patients pretreated with TMZ, ACNU was given alone (n = 14) or in combination with teniposide (n = 17) or cytarabine (n = 1), yielding a PFS6 of 20% and a median OS of 6.7 months (124). Hematological toxicity was substantial (grade 3 or 4 in 50% of patients). Three phase II–III randomized trials compared lomustine as monotherapy with investigational agents, namely enzastaurin, cediranib, or galunisertib (119121). In all three trials, the results were comparable between arms, pointing toward relevant activity of the control arm or lack of efficacy of the investigational agent. PFS6 ranged from 11 to 34.5%, median OS from 6.6 to 9.8 months, and observed RRs were low (0–16%). Four prospective phase II trials, using different schedules of administration, evaluated fotemustine in TMZ pretreated patients at first recurrence/relapse of GBM (111114). These four studies, encompassing 160 patients, showed a PFS6 of 20.9–61% and a median OS of 6 to 11 months. The best efficacy and toxicity profile was obtained with a low-dose induction regimen (fotemustine 80 mg/m2 on days 1, 15, 30, 45, and 60, followed by a 4-week rest period) ensued by a maintenance therapy (80 mg/m2 every 4 weeks) in nonprogressive patients (114). However, these data were derived from phase II trials with a small sample of patients. Phase III studies are required to determine the efficacy and safety of fotemustine, in the treatment of rGBM, after TMZ. The efficacy of PCV (procarbazine–lomustine–vincristine) was described in two retrospective studies (122, 123). They included 149 patients, of whom 16 received previous TMZ treatment. Similar results were described, with PFS6 of 29–38.4% and a median OS of 7.7–7.8 months. As expected, grade 3/4 hematologic toxicity was the most common (26%); pulmonary fibrosis was not reported (123). There is also suggestion that MGMT promoter methylation may be predictive of responsiveness to this class of agents (7, 128). In summary, different nitrosoureas show comparable efficacy in monotherapy, remaining an option in the treatment of rGBM. However, their toxicity profile, particularly hematological, limits the combination with other agents, as well as a more widespread use.

TEMOZOLOMIDE MONOTHERAPY AND COMBINATION REGIMENS

In both trials leading to the approval of TMZ, the sdTMZ schedule was used (126, 127). Four other prospective single-arm trials, all without previous TMZ treatment, used the same schedule and reached similar results with a PFS6 rate of 21–32% and a median OS of 7.0–9.9 months (129132). Table 3 reviews TMZ-based trials in rGBM. Different schedules of TMZ were experimented to increase dose intensity, aimed at overcoming TMZ resistance by cumulative depletion of MGMT (165). The main alternative schedules were continuous low dose (40–50 mg/m2 daily), 3 weeks on/1 week off (75–100 mg/m2 for 21 days every 28 days), 1 week on/1 week off (150 mg/m2 for 7 days every 14 days), but other dose-dense schedules are described (133145). The toxicity profile did not vary between the different schemes. Besides the fact that the studies did not have a comparator arm, the heterogeneity in the inclusion criteria, regarding the number of recurrences and previous treatments, limits the comparison of efficacy data. The RESCUE phase II trial examined the best timing for TMZ rechallenge, by prospectively dividing the 91 GBM patients into three groups, according to the “TMZ-free interval”: early group (progression during the first six cycles of adjuvant TMZ); extended group (progression while receiving extended adjuvant TMZ, beyond the standard six cycles, but before completion of adjuvant treatment); and rechallenge group (progression after completion of adjuvant treatment and a treatment-free interval greater than 2 months). The “early” and “rechallenge” groups, respectively, showed comparable PFS6 rates of 27.3% and 35.7%, with median PFS of 3.6 and 3.7 months, experiencing most benefit than the “extended group” (PFS6 of 7.4%, median PFS of 1.8 months). The authors considered the possibility that the PFS6 results in the “early” group could be attributable to pseudoprogression (140).

TABLE 3 Retrospective studies and clinical trials with temozolomide in recurrent GBM (temozolomide + nitrosourea combination studies are described in Table 2)

Reference Arm 1 Arm 2 Arm 3 Previous TMZ Relapse n Phase RR (%) Arm 1 vs. 2 vs. 3 PFS (months) Arm 1 vs. 2 vs. 3 PFS6 (%) Arm 1 vs. 2 vs. 3 OS (months) Arm 1 vs. 2 vs. 3
Single-arm monotherapy trials
(126) TMZ 5/28 No 1st 126 II 8 2.1 18 5.4
(129) TMZ 5/28 No 2nd   22 II 23 NA 32 7.6
(130) TMZ 5/28 No 2nd   42 II 19 NA 24 7.0
(133) TMZ 42/70 No NA   28 II 0 2.3 19 7.7
(134) TMZ 7/14 No 1st, 2nd, 3rd   21 II 10 4.9 48 NA
(131) TMZ 5/28 No 1st   13 Prospect. NA NA 21 NA
(135) TMZ 21/28 No 1st   33 II 9 3.7 30 9.2
(136) TMZ cont. Yes NA   12 Prospect. 17 6 58.3 11
(132) TMZ 5/28 No NA   19 Prospect. 24 2.2 22 9.9
(137) TMZ 7/14 9 pts NA   64 II NA 5.5 43.8 NA
(138) TMZ q12h for 28 days No ≥1st   68 II 31 4.2 35 8.8
(139) TMZ 21/28 Yes NA   27 II 7 NA 0 5.1a
RESCUE study (140) TMZ cont. Yes 1st   91 II NA NA (all), 3.6 (early), 1.8 (extended), 3.7 (rechallenge) 23.9 (all), 27.3 (early), 7.4 (extended), 35.7 (rechallenge) 9.3 (all)
(141) TMZ cont. Yes 1st   38 II 5 3.9 32.5 9.4
(142) TMZ 21/28 Yes 1st   47 II 12.8 2.3 23 13
(143) TMZ 7/14 Yes 1st   27 II 11.1 4.2 33.3 6.9
(144) TMZ 21/28 Yes 1st   58 II 13 1.8 11 11.7
(145) TMZ 7/14 Yes ≥1st   40 II NA 1.8 10 5
Single-arm combination therapy trials
(146) TMZ + CIS No NA   20 II 10 4.9 35 NA
(147) TMZ + CIS No 1st   50 II 20.4 4.2 34 11.2
(148) TMZ + Lipo. DOX No 1st   22 II 18 3.6 32 8.2
(149) TMZ + IRI No NA   91 I 12 2.6 27.3 NA
(150) TMZ + O6-BG Yes NA   34 II 3 1.7 9 4.5
(151) TMZ + INF-α TMZ + PEG-INF-α2b No NA   63 IIb 12 vs. 3 3.6 vs. 4.4 31 vs. 38 7.2 vs. 10
(152) TMZ + BVZ NA 2nd, 3rd   15 I-II NA 2.4 6.7 3.6
(153) TMZ + Sorafenib Yes 1st to 6th   32 II 3 1.5 9.4 9.6
(154) TMZ + BVZ Yes 1st, 2nd, 3rd   32 II 28 3.7 18.8 8.6
(155) TMZ + BVZ Yes 1st   32 II 40.6 4.2 21.9 7.3
(156) TMZ + ABT-414 NA NA   18 I 22.2 NA NA NA
Randomized trials
(127) TMZ PCZ No 1st 225 II 5 vs. 5 2.8 vs. 1.9 21 vs. 8 NA
(157) TMZ 5/28 TMZ 21/28 PCV No 1st 330c II NA 5.0 vs. 4.2 vs. 3.6c NA 8.5 vs. 6.6 vs. 6.7c
(158) TMZ + BVZ BVZ + ETOP Yes NA   23 II 0 vs. 0 1.0 vs. 1.9 0 vs. 7.7 2.9 vs. 4.4
(159) TMZ 21/28 Afatinib TMZ 21/28 + Afatinib Yes 1st 119 II 19 vs. 2.4 vs. 5 1.9 vs. 1 vs. 1.5 23 vs. 3 vs. 10 10.6 vs. 9.8 vs. 8
DIRECTOR trial (160) TMZ 7/14 TMZ 21/28 Yes 1st 105 II 8 vs. 16 NA 17.1 vs. 25 9.8 vs. 10.6
Retrospective studies
(161) TMZ Yes 1st, 2nd     9 Retrosp. 44.4 7.0 NA 12
(162) TMZ + resection + MITOX TMZ + resection TMZ No 1st 276 Retrosp. NA 70.7 vs. 64 vs. 39.3 NA 11 vs. 8 vs. 5
(163) TMZ(PD during TMZ) TMZ(SD during TMZ) Yes 1st   47 Retrosp. 0.0 vs. 17.2 NA 26.3 vs. 28.6 6.6 vs. 5.3
(164) TMZ + CEL 24 pts 1st, 2nd   28 Retrosp. 10.7 4.2 43 16.8

ACNU, nimustine; BCNU, carmustine; CCNU, lomustine; CED, cediranib; CIS, cisplatin; cont., continuous; DOX, doxorubicin; ERL, erlotinib; FOT, fotemustine; Galun, galunisertib; IFN, interferon; IRI, irinotecan; Lipo., liposomal; MITOX, mitoxantrone; NA, not available; O6-BG, O6-benzylguanine; OS, median overall survival; P, placebo; PCV, procarbazine-lomustine-vincristine; PCZ, procarbazine; PD, progression disease; PFS, median progression-free survival; PFS6, 6-month PFS; Pts, patients; Retrosp., retrospective; RR, response rate; SD, stable disease; TMZ, temozolomide.

aOS from all patients: 27 patients with glioblastoma, 15 with anaplastic astrocytoma, and 5 with miscellaneous brain tumors; OS of glioblastoma patients was not reported separately. bTwo single-arm phase II studies: one with short-acting (IFN) and another with long-acting (pegylated) interferon α2b. cTwo hundred and seventy seven patients with glioblastoma, 53 with anaplastic astrocytoma, and 20 with miscellaneous brain tumors; data of glioblastoma patients was not reported separately.

Four randomized phase II clinical trials were conducted using single-agent TMZ (127, 159, 166, 167). A randomized trial comparing sdTMZ with procarbazine, in TMZ-naive patients, revealed a PFS6 of 21% versus 8%, with a median OS 1.5 months longer in the TMZ arm (127). Brada et al. compared two different TMZ schedules with PCV, before TMZ became first-line standard, in patients with recurrent high-grade glioma (no separate data for GBM patients were provided) (166). In this trial, TMZ (both arms combined) did not display a clear benefit compared with PCV. It also showed that TMZ dose-intense regimens do not provide a survival or PFS benefit compared with standard doses, in the treatment of TMZ-naive patients. The DIRECTOR trial compared two dose-dense regimens of TMZ (120 mg/m2/day, 1 week on/1 week off versus 80 mg/m2/day, 3 weeks on/1 week off), in patients with GBM at first progression, after TMZ chemoradiotherapy and at least two maintenance TMZ cycles (160). The outcome was comparable between arms regarding efficacy, safety, and tolerability. The most important result of this trial was the strong prognostic role of the MGMT promoter methylation status in patients rechallenged with TMZ. PFS6 was increased by 5.8-fold (39.7 % in patients with methylated MGMT versus 6.9 % in unmethylated tumours), and OS at 12 months by 2.4-fold. Also, a significantly improved outcome was demonstrated in patients with an interval above 2 months from previous TMZ, and largely confined to patients with MGMT methylated promoter (160). Wick et al. conducted a retrospective review of 80 patients with recurrent glioma (45 with GBM) rechallenged with various TMZ schedules (163). Upon progression, those who had stable disease and a TMZ-free interval of at least 8 weeks were treated with the same or an alternative regimen of TMZ; the group progressing under TMZ received an alternative regimen. The efficacy results were comparable between groups and no clear evidence of cumulative toxicity has emerged (163). Considering the small numbers of patients in most studies and the wide range of TMZ regimens tested, there was no evidence that one metronomic schedule was superior over the other in terms of efficacy or safety. Numerous other studies evaluated TMZ-based combination regimens in rGBM but have failed to deliver conclusive efficacy beyond single-agent activity of TMZ. Those combination partners prospectively evaluated in single-arm designs were bevacizumab, interferon-α2b, sorafenib, O6-benzylguanine, irinotecan, cisplatin, liposomal doxorubicine, and ABT-414 (146156, 158).

BEVACIZUMAB MONOTHERAPY AND COMBINATION REGIMENS

The first documented use of bevacizumab in GBM was a small series of patients with rGBM treated by Stark-Vance et al. (Table 4) (168). The authors used the combination of bevacizumab with irinotecan, which showed activity, with acceptable toxicity profile. Several prospective phase II studies were subsequently conducted. Two phase II studies, by Vredenburg et al., using the same combination, achieved a RR of 57–60.9%, PFS6 rate of 30–46%, and a median OS of 9 to 10 months (171, 172). Previous reports on salvage therapy for rGBM showed inferior efficacy results, with RR of 5–10%, PFS6 rate of 9–25%, and median OS of 5 to 6 months (108, 202, 203). In 2009, FDA approved bevacizumab for patients with rGBM, based on the results of two phase II prospective studies (64, 65). However, in Europe, bevacizumab was not approved because of lack of a bevacizumab-free control arm. The BRAIN study, a phase II noncomparative trial, randomized patients to bevacizumab plus irinotecan or bevacizumab monotherapy (64). RR was 37.8 and 28.2% for the combination and monotherapy arms, respectively, and PFS6 was similar between the groups (50.3 and 42.6%), which compared favorably with historical controls. Numerous other retrospective studies addressing the combination of bevacizumab plus irinotecan described similar results (191193, 196, 199, 201). Several phase II trials evaluated the combination of irinotecan with bevacizumab, and two trials added a third combination partner, cetuximab or carboplatin (171173, 176, 178, 179). RR ranged from 25 to 60.9%, PFS6 between 28 and 46.5%, and median OS between 6.7 and 9.7 months. A retrospective analysis by Nghiemphu et al. compared two groups: one with bevacizumab in combination with different chemotherapy agents, and the other, a control group, without bevacizumab. The authors found a significant improvement in PFS (P = 0.01) and OS (P = 0.04) in favor of the group treated with bevacizumab (195).

TABLE 4 Retrospective studies and clinical trials with bevacizumab in recurrent GBM

Reference Arm 1 Arm 2 Arm 3 Relapse n Phase RR (%) Arm 1 vs. 2 vs. 3 PFS (months) Arm 1 vs. 2 vs. 3 PFS6 (%) Arm 1 vs. 2 vs. 3 OS (months) Arm 1 vs. 2vs. 3
Single-arm monotherapy trials
(65) BVZ 1st   48 II 35 3.7 29 7.2
(169) BVZ ≥1st   50 II 24.5 25 6.5
(170) BVZ 1st   29 II 27.6 3.3 33.9 10.5
Single-arm combination therapy trials
(171) BVZ + IRI NA   35 II 57.0 5.5 46.0 9.7
(172) BVZ + IRI NA   23 II 60.9 4.6 30 9.2
(173) BVZ + IRI NA   57 II 37
(174) BVZ + IRI 1st to 6th   37 Prospect. 63.9 5.0 44.3 9.0
(175) BVZ + ETOP 1st, 2nd, 3rd   27 II 23 44.4 10.7
(176) BVZ + IRI + CETUX 1st   32 II 26 33 6.7
(177) BVZ + Erlotinib 1st, 2nd, 3rd   25 II 50 29.2 10.5
(178) BVZ + IRI NA   32 II 25 5.2 28 7.9
(179) BVZ + IRI + CARBO 1st, 2nd, 3rd   40 II 33 46.5 8.3
(154) BVZ + TMZ 1st, 2nd, 3rd   32 II 28 3.7 18.8 8.6
(180) BVZ + Tem NA   13 II 0 1.8 3.5
(181) BVZ + Sora NA   54 II 18.5 2.9 20.4 5.6
(182) BVZ + FOT 1st   54 II 52 5.2 42.6 9.1
(183) BVZ + Pano 1st, 2nd   24 II 29.2 5.0 30.4 9.0
(184) BVZ + VOR NA   40 II 22.5 30 10.4
Randomized trials
(64) BVZ BVZ + IRI 1st, 2nd 167 II 28.2 vs. 37.8 4.2 vs 5.6 42.6 vs. 50.3 9.2 vs. 8.7
(152) BVZ + TMZ 2nd, 3rd   15 I-II NA 2.4 6.7 3.6
(158) BVZ + TMZ BVZ + ETOP NA   23 II 0 vs. 0 0 vs. 7.7 2.9 vs. 4.4
BELOB trial(128) BVZ CCNU BVZ + CCNU 110/90 1st 148 II 38 vs. 5 vs. 63/34 3.0 vs. 1.0 vs. 11.0/4.0 16 vs. 13 vs. 50/41 8 vs. 8 vs. 16/11
(185) BVZ BVZ + CCNU 1st, 2nd, 3rd   68 II 4.1 vs. 4.3 Pts with 1st relapse:
8.8 vs. 13.1
(186) BVZ BVZ + CARBO 1st 110 II 6 vs. 14 3.5 vs. 3.5 18 vs. 15 7.5 vs. 6.9
(187) BVZ BVZ + VOR 1st   90 II 3.6 vs. 4.2 7.0 vs. 8.3
(188) BVZ + P BVZ + Dasa NA 121 II (1:2) 26.5 vs. 18.3 18.4 vs. 27.2 7.9 vs. 7.2
(155) BVZ + TMZ 1st   32 II 40.6 4.2 21.9 7.3
(189) BVZ + CCNU CCNU 1st 437 III (2:1) 4.2 vs. 1.5 9.1 vs. 8.6
Retrospective studies
(168) BVZ + IRI NA   21 Retrosp. 42.9b
(190) BVZ + IRI or CARBO or ETOP 1st to 6th   14 Retrosp. 40a
(191) BVZ + IRI 1st to 5th   13 Retrosp. 77 6.3
(192) BVZ + IRI 1st to 10th   27 Retrosp. 3.8 17 7.1
(193) BVZ + IRI ≥1st   20 Retrosp. 47.4b 4.2b 25b 7.0b
(194) BVZ + IRI or CARBO or CCNU or ETOP 1st to 7th   55 Retrosp. 34.1b 42a
(195) BVZ + IRI or CARBO or CCNU or ETOP Control group (various drugs with no BVZ) 1st to 3rd   20 Retrosp. 4.3 vs. 1.8a 41 vs. 18a 9.0 vs. 6.1a
(196) BVZ + IRI ≥1st   51 Retrosp. 67.6 7.6 63.7 11.5
(171) BVZ + CARBO or IRI or BCNU or CCNU or Erlotinib or ETOP 2nd (after BVZ)   54 Retrosp. 0a 2b
(172) BVZ + CARBO + ETOP 1st to 5th 6 Retrosp.   83 22 7.0
(197) BVZ 1st, 2nd   50 Retrosp. 42 42 8.5
(198) BVZ BVZ + IRI or CARBO or TMZ 2nd, 3rd   24 Retrosp. 50 vs. 19a 0 vs. 14a 1.5 vs. 5.2a
(199) BVZ + IRI 1st to 5th   93 Retrosp. 56 5.1 42 8.8
(200) BVZ ± IRI or CARBO 2nd, 3rd   14 Retrosp. 29a 4.0a 29a 7.8a
(201) BVZ + IRI PCV 1st   60 Retrosp. 66.0 vs 11.0 5.0 vs. 3.0 9.0 vs. 5.0

BCNU, carmustine; BVZ, bevacizumab; CARBO, carboplatin; CETUX, cetuximab; CCNU, lomustine; Dasa, Dasatinib; EVE, everolimus; ETOP, etoposide; FOT, fotemustine; IRI, irinotecan; NA, not available; OS, median overall survival; P, placebo; Pano, Panobinostat; PCV, procarbazine-lomustine-vincristine; PFS, median progression-free survival; PFS6, 6-month PFS; Pts, patients; Retrosp., retrospective; RR, response rate; Sora, Sorafenib; Tem, temsirolimus; TMZ, temozolomide; VOR, vorinostat.

aFor overall group. bIncludes patients with GBM and other high-grade gliomas.

Numerous studies have assessed bevacizumab in combination with other agents, namely etoposide, TMZ, fotemustine, dasatinib, temsirolimus, erlotinib, sorafenib, panobinostat, or vorinostat (154, 158, 175, 177, 179181, 183, 184, 187189). In randomized trials involving two arms—one with bevacizumab in combination with experimental agents (irinotecan, carboplatin, vorinostat, or dasatinib) and the other with bevacizumab alone—it was found that both arms showed comparable efficacy, leading to the conclusion of poor efficacy of the experimental agent, without valid evidence regarding the single-agent activity of bevacizumab. A recent Dutch, open-label, three-group multicenter phase II trial (BELOB) reported promising results with bevacizumab combined with lomustine (128). Improved OS at 9 months (59% vs. 43% vs. 38%) and PFS6 (41% vs. 13% vs. 16%) were seen in the combination arm compared with single-agent lomustine and single-agent bevacizumab, respectively. Effectively, this was the first trial with bevacizumab in rGBM to demonstrate an improvement in a primary OS endpoint, suggesting increased effectiveness with the combination of bevacizumab and lomustine versus each of these agents alone. Therefore, a randomized phase III trial was performed, comparing bevacizumab–lomustine with single-agent lomustine (189). Unfortunately, a benefit in OS was not observed, while the improvement in PFS for the combination arm was maintained. A crossover to bevacizumab occurred in 35.5% of patients, which may account for these results. To evaluate the efficacy of bevacizumab beyond the second-line treatment, Piccioni et al. performed a retrospective analysis of 468 GBM patients treated at different recurrences (first, second, third, or more), including 80 who were treated upfront. The authors found that PFS and OS were similar for all three recurrence groups (median 4.1 and 9.8 months, respectively) (204). These data suggest that bevacizumab could have a role in the treatment of GBM independent of the line of therapy, and that a deferred use of bevacizumab seems not to decrease effectiveness. When comparing the results of available phase II trials on bevacizumab, alone or in combination with irinotecan, with those of standard cytotoxic chemotherapy, in rGBM, several findings are clear: bevacizumab alone has activity and increases RR, PFS6, and median PFS; on the other hand, the impact on OS is less clear.

Tumor-Treating Electric Fields for Glioblastoma

CONCEPT

Tumor-Treating Fields (TTFields) has been called the “fourth cancer treatment modality,” after surgery, RT, and pharmacotherapy. It’s a locoregionally antimitotic treatment that delivers low-intensity, intermediate-frequency (200 kHz), alternating electric fields, through four transducer arrays, consisting of nine insulated electrodes applied to the shaved scalp and connected to a portable device (11). In vitro studies have shown that TTFields arrests cell division and kills tumor cells through multiple mechanisms, namely, misalignment of microtubule subunits during division, aberrant chromosomal segregation, and cytoplasmic blebbing during anaphase.

CLINICAL TRIALS

Clinical effectiveness and feasibility of TTFields was first tested in 10 patients with rGBM, with PFS6 and median survival doubling that of historic controls (205).

Two pivotal randomized trials studied TTFields in rGBM (EF-11) and nGBM (EF-14).

In EF-11 trial, a total of 237 rGBM patients, after initial treatment with RT-TMZ, were randomized 1:1 to either the novel TTFields therapy (120 patients) or to treatment according to investigator’s choice (117 patients). Although EF-11 did not meet its primary endpoint of improving OS, similar median OS, and PFS in both arms, it established TTFields as noninferior to chemotherapy (206). In addition, the favorable QoL and toxicity profile led to FDA approval, in 2011, of TTFields as a therapeutic option for use in rGBM. The EF-14 trial, an open-label phase 3 study, enrolled 695 patients and evaluated the efficacy and safety of TTFields in combination with TMZ maintenance treatment, after chemoradiation therapy for patients with nGBM. The trial was terminated based on the results of the preplanned interim analysis that evaluated the outcomes of the first 315 patients and showed a significant improvement in PFS and OS. The percentage of patients alive at 2 years was 43% in the TTFields/TMZ group and 29% in the TMZ alone group (P = 0.006) (207). In October 2015, the FDA approved TTFields for use in nGBM patients. National Comprehensive Cancer Network has further incorporated TTFields in their updated guidelines (208).

ISSUES

TTFields are particularly safe, since no additional systemic toxicity was observed with the addition of this technology. The most common side effects are mild to moderate skin reactions beneath the transducer arrays, observed in 44% of patients, and grade 3 skin reactions in 1–2 % of patients. Additional research is warranted, in order to identify which patients are most likely to be responsive to TTFields. Benefit was present across all subgroups studied (according to age, PS, MGMT methylation status, and EOR), but the follow-up remains short, and some subsets are rather small in number. Detailed subgroup analyses are to be performed on the final and validated dataset. Although approximately three-quarters of patients, in EF-14, had a treatment compliance of 75%, this is an important issue unique to this therapy, since it requires >18 h of usage per day. Another important point is the high cost of this therapeutic approach (about $20,000 monthly). Strong price regulation by health authorities could make this technology more affordable and consequently accessible to patients (209). TTFields plus TMZ represents the first major advance in the field of GBM therapy in roughly a decade, and it should be considered for patients with nGBM and no contraindications.

Although showing significant improvements in survival, the results still underscore that the majority of patients did not survive beyond 2 years, highlighting the need for additional improvements in GBM therapeutic strategies. Due to its unique and localized mechanism of action, and general absence of systemic toxicity, TTFields is particularly well suited for combination therapies, such as immunotherapy and targeted therapies (210).

Glioblastoma in the Elderly

GBM is diagnosed at a median age of 64 years, and the incidence peaks between 75 and 84 years (15.24/100,000) (211). With aging population, this incidence is expected to increase. The poor survival rates associated with GBM (about 5% at 5 years) get even poorer in patients over 65 years (less than 2.1% at 5 years) (212). Age has long been recognized as the most important prognostic factor. Elderly patients tend to have more comorbidities and worse PS than their younger counterparts diagnosed with GBM. Similarly, their tumors seldom have favorable molecular features (IDH mutations occur in less than 2% of the tumors) (213). As a result, these patients have frequently been undertreated and underrepresented in clinical trials.

SURGERY

Several retrospective studies have shown an increase in OS, in elderly patients submitted to surgical resection (as opposed to biopsy) (206, 214216). In the study by Keime-Guibert, and in the NOA-08 and the Nordic trials, the EOR was identified as an independent prognostic factor. As such, age alone should not preclude an attempt at complete resection (8, 206, 217).

RADIOTHERAPY

RT was associated with a statistically significant, although modest, gain in OS, when compared to best supportive care (BSC), in patients aged over 70 years (206). The study was interrupted after the first interim analysis due to superiority of RT. There was no difference between the two groups, regarding QoL and NCF. Roa et al. compared hypofractionated RT (HFRT; 40 Gy in 15 fractions over 3 weeks) with cfRT (60 Gy in 30 fractions over 6 weeks) in 100 patients aged 60 years or older (218). There was no difference in OS between the two groups and the patients treated with HFRT required less increment in post-therapy corticosteroid dose (23% vs. 49%; P = 0.02). Although the study could not show that the two treatments were equivalent, together these results led to the adoption of HFRT as a valid option in the treatment of elderly patients, particularly those with a poor PS.

CHEMOTHERAPY

In an ANOCEF phase II trial, 70 patients aged 70 years or older, with a KPS under 70%, received sdTMZ until disease progression (219). The 25 weeks median OS compared favorably with the 12–16 weeks expected with BSC alone. Furthermore, there was an improvement in functional status in 33% of patients. Patients with MGMT promoter methylation had longer PFS and OS. A previous study by Chinot and colleagues had shown similar survival results (220).

CHEMOTHERAPY AND RADIOTHERAPY

The 5-year analysis of the hallmark study by Stupp et al. (3) showed a survival benefit for the combination in all subgroups, including patients aged over 60 years, RPA class V, and unmethylated MGMT promoter (211). However, an analysis by age strata showed a diminishing benefit of TMZ association with increasing age, especially in patients older than 65 years (221). Caution should be made in interpreting these results as the group over the age of 65 years represented only 15% of the study population. The 2014 EANO guidelines reflected this concern by including the multimodality treatment as an option for fit elderly patients (9). A retrospective analysis of 293 patients over the age of 65 showed a benefit for the combined regimen (222). A retrospective survey of the National Cancer Database yielded similar results, with combined modality treatment showing superiority over both chemotherapy alone and RT alone, in a group of 16,717 patients, with nGBM, aged 65 years or older (223). Two prospective randomized studies addressed the question of which single modality treatment would be best for elderly patients. In the Nordic trial, patients over the age of 60, with ECOG PS 0-3, were randomized between three treatment arms: sdTMZ (up to six cycles), HFRT (34.0 Gy administered in 3.4 Gy fractions over 2 weeks), and cfRT (217). Fewer patients completed the course of RT in the standard group (72%) compared to the hypofractionated one (95%), which may partly account for the poor results obtained in the former group. TMZ and HFRT yielded similar survival results and, particularly in patients over the age of 70, these were significantly better than the ones for cfRT. MGMT promoter methylation was predictive of response to TMZ. In the NOA-08 trial, patients older than 65 years, with a KPS >50%, were randomized to receive TMZ (100 mg/m2, given on days 1–7 of 1 week on, 1 week off cycles) or cfRT (8). The trial showed noninferiority of TMZ and there were no differences regarding QoL either, between the treatment arms. MGMT promoter methylation was both prognostic and predictive of response to TMZ. Event-free survival was longer in patients with methylated MGMT promoter treated with TMZ than in those submitted to RT (8.4 vs. 4.6 months) and the opposite was true for the group of patients with an unmethylated promoter (3.3 vs. 4.6 months). Taken together, these results support the role of MGMT promoter methylation in the choice of single modality treatment, in elderly patients with nGBM.

The EORTC 26062-22061 trial was designed to assess whether the addition of TMZ to HFRT would translate into a survival benefit (224). A total of 562 patients, aged 65 years or older, were randomized, with the combined modality being associated with a longer median OS (9.3 vs. 7.6 months; P < 0.001) and PFS. Again, MGMT promoter methylation was predictive of response to TMZ (median OS: 13.5 months with RT-TMZ vs. 7.7 months with RT alone; P < 0.001). Although not reaching statistical significance, the combined therapy also offered a survival advantage to the group with an unmethylated MGMT status (median OS: 10.0 months vs. 7.9 months; P = 0.055). QoL was similar in both study groups. An unsolved question is which RT-TMZ scheme is better for fit elderly patients. Some retrospective studies have addressed this issue. Arvold and colleagues found no differences in OS, between cfRT-TMZ and HFRT-TMZ, after adjusting for selection bias (225). Minitti also found no differences in OS and PFS between the two groups (226). However, cfRT-TMZ was associated with more neurologic toxicity (P = 0.01), lowering of KPS scores over time (P = 0.01), and higher post-treatment dosing of corticosteroid (P = 0.02). There are numerous issues that make this a special and challenging group. The definition of elderly varies widely between studies limiting the extrapolation of results to our patients’ population. Several trials lack NCF and QoL evaluations necessary for us to understand the real impact of the current available therapies in the elderly patient. The assessment of MGMT promoter methylation, although proven useful in this population, is not readily available to all. Furthermore, these patients are frequently only submitted to biopsy, which may render insufficient samples to MGMT promoter status determination.

Elderly patients with GBM have a worse prognosis than their younger counterparts. This relates to several factors, namely, poorer PS, comorbidities, delay in diagnosis (symptoms are often interpreted as signs of depression or dementia), and IDH wild-type tumors. These patients tend to be undertreated solely based on their biological age and because, they are underrepresented in clinical trials, there’s a paucity of data guiding clinical decisions. Based on prospective trials, HFRT has become standard in this population and proven equivalent to TMZ, and MGMT promoter methylation status has a paramount importance in the choice of single modality therapy. In addition, there’s now evidence that the addition of TMZ to HFRT yields an increase in OS, representing an alternative to the Stupp regimen, in elderly patients with a good PS.

Supportive Care

The patient with GBM is, simultaneously, a patient with cancer and one with a progressive neurological disease. As such, there are certain specificities regarding not only the most frequent symptoms exhibited but also some end-of-life (EOL) care issues. Patients with primary brain tumors were found to have poorer PS, higher levels of nursing and social support, and more family overburden than other palliative care patients. Disorientation and confusion were also more frequent. Conversely, general EOL symptoms, such as dyspnoea, nausea, vomiting, anorexia, constipation, and pain, were experienced less frequently (227). Palliative care should aim at improving QoL, both for the patient and the caregiver, and is not limited to the EOL stage. The timing of its introduction, in the management of GBM patients, is an understudied issue. The experience with metastatic nonsmall cell lung cancer indicates improvements in QoL, mood, and symptom burden, as well as better EOL care and even extended survival, with early initiation of palliative care (228). Disease itself, along with GBM treatment side effects and symptomatic medication (namely antiepileptic drugs), affects cognition and impairs decision-making, very often early in the disease course (229, 230). As such, timely involvement of the patient in treatment decisions (including supportive measures ahead) is of paramount importance. Two systematic reviews of studies addressing the EOL phase, in high-grade gliomas, showed a high burden of symptoms, namely reduced consciousness (44–90%), dysphagia (10–85%), headache (36–62%), seizures (10–56%), focal neurological deficits (>50%), cognitive disturbances (>30%), confusion (15–51%), and poor communication (64–90%) (227, 231236).

SEIZURES

Approximately 30% of glioma patients have a seizure during the last week of life, regardless of having or not having a history of seizures (235, 237). As mentioned earlier, prophylactic use of anticonvulsant drugs is not indicated in patients without history of seizures (12). Enzyme inducers antiepileptic drugs should be avoided as they interact with commonly used cytotoxic agents and dexamethasone, having the potential to reduce their efficacy (238). Valproic acid is an enzyme inhibitor that may increase therapeutic levels of antineoplastic agents. Several reports also suggest a direct antitumor effect, but this is yet to be proven (239). Levetiracetam is the most studied of the recent anticonvulsants in this setting. It appears safe without major interactions with the commonly used drugs (240). At the EOL, dysphagia and altered consciousness are common and impair the administration of oral medication. As a result, half of the patients taper antiepileptic drugs in the last week of life, with one-third experiencing seizures (237). As the occurrence of seizures is associated with nonpeaceful death, it is important to maintain antiepileptic treatment throughout the EOL phase (231). This can be achieved by using alternative routes of administration. For patients in home care, rectal diazepam and buccal or intranasal midazolam are convenient alternatives (241).

DEPRESSION

Diagnosis can be difficult as all the symptoms of a major depressive disorder, with the exception of suicidal thoughts, can be attributed to the tumor, its treatment, or both (242). Selective serotonin reuptake inhibitors may be considered as first-line treatment of depression, as they have not shown increased toxicity in glioma patients and they are not associated with increased incidence of seizures in the general population (242, 243). The benefit and feasibility of psychotherapy in treating depression and anxiety in glioma patients is uncertain (242).

RAISED INTRACRANIAL PRESSURE

Raised intracranial pressure, as a result of tumor growth and cerebral edema, can cause headache, nausea, vomiting, somnolence, and visual disturbances. Corticosteroids are the treatment of choice. Dexamethasone is often used for its long half-life, anti-inflammatory activity, and absence of mineralocorticoid effect (244). Corticosteroids must be tapered as soon as possible and kept in the lowest dose capable of controlling symptoms. Attention must be given to the complications associated with prolonged steroid use.

CONFUSION

Confusion is a major cause of distress for the patient and his caregivers. It can arise from the tumor itself, or be caused by pain, infection, metabolic imbalances, symptomatic treatments, fecaloma, bladder retention, intracranial hemorrhage, or seizures (244). Neuroleptics, such as haloperidol, risperidone, and olanzapine, are often needed. Opioids and sedatives are also options if pain or sleep and behavioral disorders coexist.

ISSUES AT EOL CARE

During the EOL phase, dysphagia and altered consciousness will impair nutrition and hydration, and the administration of symptomatic medication, namely, corticosteroids and anticonvulsants. As mentioned before, these last ones should be kept, but steroids are often tapered and discontinued in the last days of life, when the patient is already unconscious, to avoid futile prolongation of life. Maintenance or withdrawal of artificial nutrition and hydration and symptomatic medications are EOL decisions common to all glioma patients. Another topic that can be discussed with the patient in advance is palliative sedation. Between 13 and 45% of patients were reported to have received it. Refractory seizures, agitation, and delirium are among the reasons that lead to its institution (245). The knowledge on palliative care in glioma patients is largely based on retrospective studies and on extrapolations from studies performed on other cancer patients. Properly conducted prospective and interventional investigations, specifically designed for glioma patients, addressing the specificities of this population are needed.

Conclusion

Despite maximal safe surgical resection and combined chemotherapy and RT, GBM retains a poor prognostic value. To date, excluding TTFields, no new agents improve survival when added to standard therapy. Although MGMT promoter methylation is predictive of response to TMZ, its role in the choice of first-line therapy is currently limited to the elderly GMB patients. No standard of care is established in the recurrent setting. Bevacizumab clearly impacts PFS, although its role in OS is less certain. TMZ rechallenge is a treatment option, especially for MGMT promoter-methylated rGBM. All in all, while efforts are being put in strategies to prolong OS, enhancing QoL for these patients must be a priority.

Conflict of interest: The authors declare no potential conflicts of interest with respect to research, authorship, and/or publication of this manuscript.

Copyright and permission statement: We confirm that the materials included in this chapter do not violate copyright laws. Where relevant, appropriate permissions have been obtained from the original copyright holder(s). All original sources have been appropriately acknowledged and/or referenced.

REFERENCES

  1. Thakkar JP, Dolecek TA, Horbinski C, Ostrom QT, Lightner DD, Barnholtz-Sloan JS, et al. Epidemiologic and molecular prognostic review of glioblastoma. Cancer Epidemiol Biomarkers Prev. 2014;23(10):1985–96. http://dx.doi.org/10.1158/1055-9965.EPI-14-0275
  2. Gilbert MR, Wang M, Aldape KD, Stupp R, Hegi ME, Jaeckle KA, et al. Dose-dense temozolomide for newly diagnosed glioblastoma: A randomized phase III clinical trial. J Clin Oncol. 2013;31(32): 4085–91. http://dx.doi.org/10.1200/JCO.2013.49.6968
  3. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–96. http://dx.doi.org/10.1056/NEJMoa043330
  4. Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370(8):709–22. http://dx.doi.org/10.1056/NEJMoa1308345
  5. Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370(8):699–708. http://dx.doi.org/10.1056/NEJMoa1308573
  6. Esteller M, Gaidano G, Goodman SN, Zagonel V, Capello D, Botto B, et al. Hypermethylation of the DNA repair gene O(6)-methylguanine DNA methyltransferase and survival of patients with diffuse large B-cell lymphoma. J Natl Cancer Inst. 2002;94(1):26–32. http://dx.doi.org/10.1093/jnci/94.1.26
  7. Esteller M, Garcia-Foncillas J, Andion E, Goodman SN, Hidalgo OF, Vanaclocha V, et al. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 2000;343(19):1350–4. http://dx.doi.org/10.1056/NEJM200011093431901
  8. Wick W, Platten M, Meisner C, Felsberg J, Tabatabai G, Simon M, 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. http://dx.doi.org/10.1016/S1470-2045(12)70164-X
  9. Weller M, van den Bent M, Hopkins K, Tonn JC, Stupp R, Falini A, et al. EANO guideline for the diagnosis and treatment of anaplastic gliomas and glioblastoma. Lancet Oncol. 2014;15(9):e395–403. http://dx.doi.org/10.1016/S1470-2045(14)70011-7
  10. Weller M, Cloughesy T, Perry JR, Wick W. Standards of care for treatment of recurrent glioblastoma— Are we there yet? Neuro Oncol. 2013;15(1):4–27. http://dx.doi.org/10.1093/neuonc/nos273
  11. Hottinger AF, Pacheco P, Stupp R. Tumor treating fields: A novel treatment modality and its use in brain tumors. Neuro Oncol. 2016;18(10):1338–49. http://dx.doi.org/10.1093/neuonc/now182
  12. Glantz MJ, Cole BF, Forsyth PA, Recht LD, Wen PY, Chamberlain MC, et al. Practice parameter: Anticonvulsant prophylaxis in patients with newly diagnosed brain tumors. Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2000;54(10):1886–93. http://dx.doi.org/10.1212/WNL.54.10.1886
  13. Wolbers JG. Novel strategies in glioblastoma surgery aim at safe, supra-maximum resection in conjunction with local therapies. Chin J Cancer. 2014;33(1):8–15. http://dx.doi.org/10.5732/cjc.013.10219
  14. Li YM, Suki D, Hess K, Sawaya R. The influence of maximum safe resection of glioblastoma on survival in 1229 patients: Can we do better than gross-total resection? J Neurosurg. 2016;124(4): 977–88. http://dx.doi.org/10.3171/2015.5.JNS142087
  15. 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. http://dx.doi.org/10.3171/2011.2.JNS10998
  16. Lacroix M, Abi-Said D, Fourney DR, Gokaslan ZL, Shi W, DeMonte F, et al. A multivariate analysis of 416 patients with glioblastoma multiforme: Prognosis, extent of resection, and survival. J Neurosurg. 2001;95(2):190–8. http://dx.doi.org/10.3171/jns.2001.95.2.0190
  17. Sanai N, Berger MS. Intraoperative stimulation techniques for functional pathway preservation and glioma resection. Neurosurg Focus. 2010;28(2):E1. http://dx.doi.org/10.3171/2009.12.FOCUS09266
  18. Chaichana KL, Jusue-Torres I, Navarro-Ramirez R, Raza SM, Pascual-Gallego M, Ibrahim A, et al. Establishing percent resection and residual volume thresholds affecting survival and recurrence for patients with newly diagnosed intracranial glioblastoma. Neuro Oncol. 2014;16(1):113–22. http://dx.doi.org/10.1093/neuonc/not137
  19. Grabowski MM, Recinos PF, Nowacki AS, Schroeder JL, Angelov L, Barnett GH, et al. Residual tumor volume versus extent of resection: Predictors of survival after surgery for glioblastoma. J Neurosurg. 2014;121(5):1115–23. http://dx.doi.org/10.3171/2014.7.JNS132449
  20. Pallud J, Rigaux-Viode O, Corns R, Muto J, Lopez Lopez C, Mellerio C, et al. Direct electrical bipolar electrostimulation for functional cortical and subcortical cerebral mapping in awake craniotomy. Practical considerations. Neurochirurgie. 2017 Jun;63(3):164–174. http://dx.doi.org/10.1016/j.neuchi.2016.08.009
  21. Mueller WM, Yetkin FZ, Hammeke TA, Morris GL, 3rd, Swanson SJ, Reichert K, et al. Functional magnetic resonance imaging mapping of the motor cortex in patients with cerebral tumors. Neurosurgery. 1996;39(3):515–20; discussion 20–1. http://dx.doi.org/10.1227/00006123-199609000-00015
  22. Szelenyi A, Bello L, Duffau H, Fava E, Feigl GC, Galanda M, et al. Intraoperative electrical stimulation in awake craniotomy: Methodological aspects of current practice. Neurosurg Focus. 2010;28(2):E7. http://dx.doi.org/10.3171/2009.12.FOCUS09237
  23. De Witt Hamer PC, Robles SG, Zwinderman AH, Duffau H, Berger MS. Impact of intraoperative stimulation brain mapping on glioma surgery outcome: A meta-analysis. J Clin Oncol. 2012;30(20): 2559–65. http://dx.doi.org/10.1200/JCO.2011.38.4818
  24. Panciani PP, Fontanella M, Garbossa D, Agnoletti A, Ducati A, Lanotte M. 5-aminolevulinic acid and neuronavigation in high-grade glioma surgery: Results of a combined approach. Neurocirugia (Astur). 2012;23(1):23–8. http://dx.doi.org/10.1016/j.neucir.2012.04.003
  25. Senders JT, Muskens IS, Schnoor R, Karhade AV, Cote DJ, Smith TR, et al. Agents for fluorescence-guided glioma surgery: A systematic review of preclinical and clinical results. Acta Neurochir (Wien). 2017;159(1):151–67. http://dx.doi.org/10.1007/s00701-016-3028-5
  26. Stummer W, Pichlmeier U, Meinel T, Wiestler OD, Zanella F, Reulen HJ, 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. http://dx.doi.org/10.1016/S1470-2045(06)70665-9
  27. Eljamel MS, Goodman C, Moseley H. ALA and Photofrin fluorescence-guided resection and repetitive PDT in glioblastoma multiforme: A single centre Phase III randomised controlled trial. Lasers Med Sci. 2008;23(4):361–7. http://dx.doi.org/10.1007/s10103-007-0494-2
  28. Stummer W, Tonn JC, Mehdorn HM, Nestler U, Franz K, Goetz C, et al. Counterbalancing risks and gains from extended resections in malignant glioma surgery: A supplemental analysis from the randomized 5-aminolevulinic acid glioma resection study. Clinical article. J Neurosurg. 2011;114(3): 613–23. http://dx.doi.org/10.3171/2010.3.JNS097
  29. Young RM, Jamshidi A, Davis G, Sherman JH. Current trends in the surgical management and treatment of adult glioblastoma. Ann Transl Med. 2015;3(9):121.
  30. Athanassiou H, Synodinou M, Maragoudakis E, Paraskevaidis M, Verigos C, Misailidou D, et al. Randomized phase II study of temozolomide and radiotherapy compared with radiotherapy alone in newly diagnosed glioblastoma multiforme. J Clin Oncol. 2005;23(10):2372–7. http://dx.doi.org/10.1200/JCO.2005.00.331
  31. Karacetin D, Okten B, Yalcin B, Incekara O. Concomitant temozolomide and radiotherapy versus radiotherapy alone for treatment of newly diagnosed glioblastoma multiforme. J BUON. 2011;16(1):133–7.
  32. Kocher M, Frommolt P, Borberg SK, Ruhl U, Steingraber M, Niewald M, et al. Randomized study of postoperative radiotherapy and simultaneous temozolomide without adjuvant chemotherapy for glioblastoma. Strahlenther Onkol. 2008;184(11):572–9. http://dx.doi.org/10.1007/s00066-008-1897-0
  33. Szczepanek D, Marchel A, Moskala M, Krupa M, Kunert P, Trojanowski T. Efficacy of concomitant and adjuvant temozolomide in glioblastoma treatment. A multicentre randomized study. Neurol Neurochir Pol. 2013;47(2):101–8. http://dx.doi.org/10.5114/ninp.2013.34398
  34. Feng E, Sui C, Wang T, Sun G. Temozolomide with or without radiotherapy in patients with newly diagnosed glioblastoma multiforme: A meta-analysis. Eur Neurol. 2017;77(3–4):201–10. http://dx.doi.org/10.1159/000455842
  35. Gorlia T, van den Bent MJ, Hegi ME, Mirimanoff RO, Weller M, Cairncross JG, et al. Nomograms for predicting survival of patients with newly diagnosed glioblastoma: Prognostic factor analysis of EORTC and NCIC trial 26981-22981/CE.3. Lancet Oncol. 2008;9(1):29–38. http://dx.doi.org/10.1016/S1470-2045(07)70384-4
  36. Hegi ME, Liu L, Herman JG, Stupp R, Wick W, Weller M, et al. Correlation of O6-methylguanine methyltransferase (MGMT) promoter methylation with clinical outcomes in glioblastoma and clinical strategies to modulate MGMT activity. J Clin Oncol. 2008;26(25):4189–99. http://dx.doi.org/10.1200/JCO.2007.11.5964
  37. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352(10):997–1003. http://dx.doi.org/10.1056/NEJMoa043331
  38. Wick W, Platten M, Weller M. New (alternative) temozolomide regimens for the treatment of glioma. Neuro Oncol. 2009;11(1):69–79. http://dx.doi.org/10.1215/15228517-2008-078
  39. Koukourakis GV, Kouloulias V, Zacharias G, Papadimitriou C, Pantelakos P, Maravelis G, et al. Temozolomide with radiation therapy in high grade brain gliomas: Pharmaceuticals considerations and efficacy; a review article. Molecules. 2009;14(4):1561–77. http://dx.doi.org/10.3390/molecules14041561
  40. Hart MG, Garside R, Rogers G, Stein K, Grant R. Temozolomide for high grade glioma. Cochrane Database Syst Rev. 2013;(4):CD007415. http://dx.doi.org/10.1002/14651858.cd007415.pub2
  41. Le Rhun E, Taillibert S, Chamberlain MC. The future of high-grade glioma: Where we are and where are we going. Surg Neurol Int. 2015;6(Suppl 1):S9–S44. http://dx.doi.org/10.4103/2152-7806.151331
  42. Barbagallo GM, Paratore S, Caltabiano R, Palmucci S, Parra HS, Privitera G, et al. Long-term therapy with temozolomide is a feasible option for newly diagnosed glioblastoma: A single-institution experience with as many as 101 temozolomide cycles. Neurosurg Focus. 2014;37(6):E4. http://dx.doi.org/10.3171/2014.9.FOCUS14502
  43. Darlix A, Baumann C, Lorgis V, Ghiringhelli F, Blonski M, Chauffert B, et al. Prolonged administration of adjuvant temozolomide improves survival in adult patients with glioblastoma. Anticancer Res. 2013;33(8):3467–74.
  44. Roldan Urgoiti GB, Singh AD, Easaw JC. Extended adjuvant temozolomide for treatment of newly diagnosed glioblastoma multiforme. J Neurooncol. 2012;108(1):173–7. http://dx.doi.org/10.1007/s11060-012-0826-3
  45. Seiz M, Krafft U, Freyschlag CF, Weiss C, Schmieder K, Lohr F, et al. Long-term adjuvant administration of temozolomide in patients with glioblastoma multiforme: Experience of a single institution. J Cancer Res Clin Oncol. 2010;136(11):1691–5. http://dx.doi.org/10.1007/s00432-010-0827-6
  46. Blumenthal DT, Gorlia T, Gilbert MR, Kim MM, Burt Nabors L, Mason WP, et al. Is more better? The impact of extended adjuvant temozolomide in newly diagnosed glioblastoma: A secondary analysis of EORTC and NRG Oncology/RTOG. Neuro-Oncology, Volume 19, Issue 8, 1 August 2017, Pages 1119–1126. http://dx.doi.org/10.1093/neuonc/nox025
  47. Nabors LB, Fink KL, Mikkelsen T, Grujicic D, Tarnawski R, Nam DH, et al. Two cilengitide regimens in combination with standard treatment for patients with newly diagnosed glioblastoma and unmethylated MGMT gene promoter: Results of the open-label, controlled, randomized phase II CORE study. Neuro Oncol. 2015;17(5):708–17. http://dx.doi.org/10.1093/neuonc/nou356
  48. Stupp R, Hegi ME, Gorlia T, Erridge SC, Perry J, Hong YK, et al. Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071-22072 study): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15(10):1100–8. http://dx.doi.org/10.1016/S1470-2045(14)70379-1
  49. Gramatzki D, Kickingereder P, Hentschel B, Felsberg J, Herrlinger U, Schackert G, et al. Limited role for extended maintenance temozolomide for newly diagnosed glioblastoma. Neurology. 2017;88(15):1422–30. http://dx.doi.org/10.1212/WNL.0000000000003809
  50. Bregy A, Shah AH, Diaz MV, Pierce HE, Ames PL, Diaz D, et al. The role of Gliadel wafers in the treatment of high-grade gliomas. Expert Rev Anticancer Ther. 2013;13(12):1453–61. http://dx.doi.org/10.1586/14737140.2013.840090
  51. Lawrence YR, Li XA, el Naqa I, Hahn CA, Marks LB, Merchant TE, et al. Radiation dose-volume effects in the brain. Int J Radiat Oncol Biol Phys. 2010;76(3 Suppl):S20–7. http://dx.doi.org/10.1016/j.ijrobp.2009.02.091
  52. Tosoni A, Franceschi E, Poggi R, Brandes AA. Relapsed glioblastoma: Treatment strategies for initial and subsequent recurrences. Curr Treat Options Oncol. 2016;17(9):49. http://dx.doi.org/10.1007/s11864-016-0422-4
  53. Herrlinger U, Schaefer N, Steinbach JP, Weyerbrock A, Hau P, Goldbrunner R, et al. Survival and quality of life in the randomized, multicenter GLARIUS trial investigating bevacizumab/irinotecan versus standard temozolomide in newly diagnosed, MGMT-non-methylated glioblastoma patients. J Clin Oncol. 2014;32(15_suppl):2042.
  54. Herrlinger U, Schafer N, Steinbach JP, Weyerbrock A, Hau P, Goldbrunner R, et al. Bevacizumab plus irinotecan versus temozolomide in newly diagnosed O6-methylguanine-DNA methyltransferase non-methylated glioblastoma: The randomized GLARIUS trial. J Clin Oncol. 2016;34(14):1611–19. http://dx.doi.org/10.1200/JCO.2015.63.4691
  55. Carlson JA, Reddy K, Gaspar LE, Ney D, Kavanagh BD, Damek D, et al. Hypofractionated-intensity modulated radiotherapy (hypo-IMRT) and temozolomide (TMZ) with or without bevacizumab (BEV) for newly diagnosed glioblastoma multiforme (GBM): A comparison of two prospective phase II trials. J Neurooncol. 2015;123(2):251–7. http://dx.doi.org/10.1007/s11060-015-1791-4
  56. Omuro A, Beal K, Gutin P, Karimi S, Correa DD, Kaley TJ, et al. Phase II study of bevacizumab, temozolomide, and hypofractionated stereotactic radiotherapy for newly diagnosed glioblastoma. Clin Cancer Res. 2014;20(19):5023–31. http://dx.doi.org/10.1158/1078-0432.CCR-14-0822
  57. Nicholas MK, Lukas RV, Amidei C, Vick N, Paleologos N, Malkin MG, et al. Final results of a single-arm phase II study of bevacizumab and temozolomide following radiochemotherapy in newly diagnosed adult glioblastoma patients. J Clin Oncol. 2013;31(15_suppl):2076.
  58. Raizer JJ, Giglio P, Hu JL, Groves MD, Merrell R, Conrad CA, et al. BTTC08-01: A phase II study of bevacizumab and erlotinib after radiation therapy and temozolomide in patients with newly diagnosed glioblastoma (GBM) without MGMT promoter methylation. J Clin Oncol. 2013;31(15_suppl):2019.
  59. Narayana A, Gruber D, Kunnakkat S, Golfinos JG, Parker E, Raza S, et al. A clinical trial of bevacizumab, temozolomide, and radiation for newly diagnosed glioblastoma. J Neurosurg. 2012;116(2):341–5. http://dx.doi.org/10.3171/2011.9.JNS11656
  60. Hainsworth JD, Shih KC, Shepard GC, Tillinghast GW, Brinker BT, Spigel DR. Phase II study of concurrent radiation therapy, temozolomide, and bevacizumab followed by bevacizumab/everolimus as first-line treatment for patients with glioblastoma. Clin Adv Hematol Oncol. 2012;10(4):240–6.
  61. Friedman HS, Desjardins A, Peters KB, Reardon DA, Kirkpatrick J, Herndon JE, et al. The addition of bevacizumab to temozolomide and radiation therapy followed by bevacizumab, temozolomide, and oral topotecan for newly diagnosed glioblastoma multiforme (GBM). J Clin Oncol. 2012;30(15_suppl):2090.
  62. Vredenburgh JJ, Desjardins A, Reardon DA, Peters KB, Herndon JE, 2nd, Marcello J, et al. The addition of bevacizumab to standard radiation therapy and temozolomide followed by bevacizumab, temozolomide, and irinotecan for newly diagnosed glioblastoma. Clin Cancer Res. 2011;17(12): 4119–24. http://dx.doi.org/10.1158/1078-0432.CCR-11-0120
  63. Lai A, Tran A, Nghiemphu PL, Pope WB, Solis OE, Selch M, et al. Phase II study of bevacizumab plus temozolomide during and after radiation therapy for patients with newly diagnosed glioblastoma multiforme. J Clin Oncol. 2011;29(2):142–8. http://dx.doi.org/10.1200/JCO.2010.30.2729
  64. Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009;27(28):4733–40. http://dx.doi.org/10.1200/JCO.2008.19.8721
  65. Kreisl TN, Kim L, Moore K, Duic P, Royce C, Stroud I, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol. 2009;27(5):740–5. http://dx.doi.org/10.1200/JCO.2008.16.3055
  66. Nabors LB, Mikkelsen T, Hegi ME, Ye X, Batchelor T, Lesser G, et al. A safety run-in and randomized phase 2 study of cilengitide combined with chemoradiation for newly diagnosed glioblastoma (NABTT 0306). Cancer. 2012;118(22):5601–7. http://dx.doi.org/10.1002/cncr.27585
  67. Reardon DA, Fink KL, Mikkelsen T, Cloughesy TF, O’Neill A, Plotkin S, et al. Randomized phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme. J Clin Oncol. 2008;26(34):5610–17. http://dx.doi.org/10.1200/JCO.2008.16.7510
  68. Stupp R, Hegi ME, Neyns B, Goldbrunner R, Schlegel U, Clement PM, et al. Phase I/IIa study of cilengitide and temozolomide with concomitant radiotherapy followed by cilengitide and temozolomide maintenance therapy in patients with newly diagnosed glioblastoma. J Clin Oncol. 2010;28(16): 2712–18. http://dx.doi.org/10.1200/JCO.2009.26.6650
  69. Montemurro N, Perrini P, Blanco MO, Vannozzi R. Second surgery for recurrent glioblastoma: A concise overview of the current literature. Clin Neurol Neurosurg. 2016;142:60–4. http://dx.doi.org/10.1016/j.clineuro.2016.01.010
  70. Carson KA, Grossman SA, Fisher JD, Shaw EG. Prognostic factors for survival in adult patients with recurrent glioma enrolled onto the new approaches to brain tumor therapy CNS consortium phase I and II clinical trials. J Clin Oncol. 2007;25(18):2601–6. http://dx.doi.org/10.1200/JCO.2006.08.1661
  71. Chan TA, Weingart JD, Parisi M, Hughes MA, Olivi A, Borzillary S, et al. Treatment of recurrent glioblastoma multiforme with GliaSite brachytherapy. Int J Radiat Oncol Biol Phys. 2005;62(4):1133–9. http://dx.doi.org/10.1016/j.ijrobp.2004.12.032
  72. Cuneo KC, Vredenburgh JJ, Sampson JH, Reardon DA, Desjardins A, Peters KB, et al. Safety and efficacy of stereotactic radiosurgery and adjuvant bevacizumab in patients with recurrent malignant gliomas. Int J Radiat Oncol Biol Phys. 2012;82(5):2018–24. http://dx.doi.org/10.1016/j.ijrobp.2010.12.074
  73. Gabayan AJ, Green SB, Sanan A, Jenrette J, Schultz C, Papagikos M, et al. GliaSite brachytherapy for treatment of recurrent malignant gliomas: A retrospective multi-institutional analysis. Neurosurgery. 2006;58(4):701–9; discussion 701–9.
  74. Gorlia T, Stupp R, Brandes AA, Rampling RR, Fumoleau P, Dittrich C, et al. New prognostic factors and calculators for outcome prediction in patients with recurrent glioblastoma: A pooled analysis of EORTC Brain Tumour Group phase I and II clinical trials. Eur J Cancer. 2012;48(8):1176–84. http://dx.doi.org/10.1016/j.ejca.2012.02.004
  75. Minniti G, Scaringi C, De Sanctis V, Lanzetta G, Falco T, Di Stefano D, et al. Hypofractionated stereotactic radiotherapy and continuous low-dose temozolomide in patients with recurrent or progressive malignant gliomas. J Neurooncol. 2013;111(2):187–94. http://dx.doi.org/10.1007/s11060-012-0999-9
  76. Park JK, Hodges T, Arko L, Shen M, Dello Iacono D, McNabb A, et al. Scale to predict survival after surgery for recurrent glioblastoma multiforme. J Clin Oncol. 2010;28(24):3838–43. http://dx.doi.org/10.1200/JCO.2010.30.0582
  77. Combs SE, Edler L, Rausch R, Welzel T, Wick W, Debus J. Generation and validation of a prognostic score to predict outcome after re-irradiation of recurrent glioma. Acta Oncol. 2013;52(1):147–52. http://dx.doi.org/10.3109/0284186X.2012.692882
  78. Fogh SE, Andrews DW, Glass J, Curran W, Glass C, Champ C, et al. Hypofractionated stereotactic radiation therapy: An effective therapy for recurrent high-grade gliomas. J Clin Oncol. 2010;28(18):3048–53. http://dx.doi.org/10.1200/JCO.2009.25.6941
  79. Park CK, Kim JH, Nam DH, Kim CY, Chung SB, Kim YH, et al. A practical scoring system to determine whether to proceed with surgical resection in recurrent glioblastoma. Neuro Oncol. 2013;15(8):1096–101. http://dx.doi.org/10.1093/neuonc/not069
  80. Chang SM, Parney IF, McDermott M, Barker FG, 2nd, Schmidt MH, Huang W, et al. Perioperative complications and neurological outcomes of first and second craniotomies among patients enrolled in the Glioma Outcome Project. J Neurosurg. 2003;98(6):1175–81. http://dx.doi.org/10.3171/jns.2003.98.6.1175
  81. Brandes AA, Franceschi E, Tosoni A, Bartolini S, Bacci A, Agati R, et al. O(6)-methylguanine DNA-methyltransferase methylation status can change between first surgery for newly diagnosed glioblastoma and second surgery for recurrence: Clinical implications. Neuro Oncol. 2010;12(3): 283–8. http://dx.doi.org/10.1093/neuonc/nop050
  82. Suchorska B, Weller M, Tabatabai G, Senft C, Hau P, Sabel MC, et al. Complete resection of contrast-enhancing tumor volume is associated with improved survival in recurrent glioblastoma-results from the DIRECTOR trial. Neuro Oncol. 2016;18(4):549–56. http://dx.doi.org/10.1093/neuonc/nov326
  83. Ringel F, Pape H, Sabel M, Krex D, Bock HC, Misch M, et al. Clinical benefit from resection of recurrent glioblastomas: Results of a multicenter study including 503 patients with recurrent glioblastomas undergoing surgical resection. Neuro Oncol. 2016;18(1):96–104. http://dx.doi.org/10.1093/neuonc/nov145
  84. Nieder C, Adam M, Molls M, Grosu AL. Therapeutic options for recurrent high-grade glioma in adult patients: Recent advances. Crit Rev Oncol Hematol. 2006;60(3):181–93. http://dx.doi.org/10.1016/j.critrevonc.2006.06.007
  85. Mayer R, Sminia P. Reirradiation tolerance of the human brain. Int J Radiat Oncol Biol Phys. 2008;70(5):1350–60. http://dx.doi.org/10.1016/j.ijrobp.2007.08.015
  86. Shaw E, Scott C, Souhami L, Dinapoli R, Kline R, Loeffler J, et al. Single dose radiosurgical treatment of recurrent previously irradiated primary brain tumors and brain metastases: Final report of RTOG protocol 90-05. Int J Radiat Oncol Biol Phys. 2000;47(2):291–8. http://dx.doi.org/10.1016/S0360-3016(99)00507-6
  87. Shepherd SF, Laing RW, Cosgrove VP, Warrington AP, Hines F, Ashley SE, et al. Hypofractionated stereotactic radiotherapy in the management of recurrent glioma. Int J Radiat Oncol Biol Phys. 1997;37(2):393–8. http://dx.doi.org/10.1016/S0360-3016(96)00455-5
  88. Cabrera AR, Cuneo KC, Desjardins A, Sampson JH, McSherry F, Herndon JE, 2nd, et al. Concurrent stereotactic radiosurgery and bevacizumab in recurrent malignant gliomas: A prospective trial. Int J Radiat Oncol Biol Phys. 2013;86(5):873–9. http://dx.doi.org/10.1016/j.ijrobp.2013.04.029
  89. Minniti G, Armosini V, Salvati M, Lanzetta G, Caporello P, Mei M, et al. Fractionated stereotactic reirradiation and concurrent temozolomide in patients with recurrent glioblastoma. J Neurooncol. 2011;103(3):683–91. http://dx.doi.org/10.1007/s11060-010-0446-8
  90. Niyazi M, Ganswindt U, Schwarz SB, Kreth FW, Tonn JC, Geisler J, et al. Irradiation and bevacizumab in high-grade glioma retreatment settings. Int J Radiat Oncol Biol Phys. 2012;82(1):67–76. http://dx.doi.org/10.1016/j.ijrobp.2010.09.002
  91. Combs SE, Thilmann C, Edler L, Debus J, Schulz-Ertner D. Efficacy of fractionated stereotactic reirradiation in recurrent gliomas: Long-term results in 172 patients treated in a single institution. J Clin Oncol. 2005;23(34):8863–9. http://dx.doi.org/10.1200/JCO.2005.03.4157
  92. Darakchiev BJ, Albright RE, Breneman JC, Warnick RE. Safety and efficacy of permanent iodine-125 seed implants and carmustine wafers in patients with recurrent glioblastoma multiforme. J Neurosurg. 2008;108(2):236–42. http://dx.doi.org/10.3171/JNS/2008/108/2/0236
  93. Combs SE, Debus J, Schulz-Ertner D. Radiotherapeutic alternatives for previously irradiated recurrent gliomas. BMC Cancer. 2007;7:167. http://dx.doi.org/10.1186/1471-2407-7-167
  94. Moeller BJ, Cao Y, Li CY, Dewhirst MW. Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: Role of reoxygenation, free radicals, and stress granules. Cancer Cell. 2004;5(5):429–41. http://dx.doi.org/10.1016/S1535-6108(04)00115-1
  95. Moeller BJ, Dewhirst MW. Raising the bar: How HIF-1 helps determine tumor radiosensitivity. Cell Cycle. 2004;3(9):1107–10. http://dx.doi.org/10.4161/cc.3.9.1099
  96. Moeller BJ, Dreher MR, Rabbani ZN, Schroeder T, Cao Y, Li CY, et al. Pleiotropic effects of HIF-1 blockade on tumor radiosensitivity. Cancer Cell. 2005;8(2):99–110. http://dx.doi.org/10.1016/j.ccr.2005.06.016
  97. Boothe D, Young R, Yamada Y, Prager A, Chan T, Beal K. Bevacizumab as a treatment for radiation necrosis of brain metastases post stereotactic radiosurgery. Neuro Oncol. 2013;15(9):1257–63. http://dx.doi.org/10.1093/neuonc/not085
  98. Levin VA, Bidaut L, Hou P, Kumar AJ, Wefel JS, Bekele BN, et al. Randomized double-blind placebo-controlled trial of bevacizumab therapy for radiation necrosis of the central nervous system. Int J Radiat Oncol Biol Phys. 2011;79(5):1487–95. http://dx.doi.org/10.1016/j.ijrobp.2009.12.061
  99. Torcuator R, Zuniga R, Mohan YS, Rock J, Doyle T, Anderson J, et al. Initial experience with bevacizumab treatment for biopsy confirmed cerebral radiation necrosis. J Neurooncol. 2009;94(1):63–8. http://dx.doi.org/10.1007/s11060-009-9801-z
  100. Greenspoon JN, Sharieff W, Hirte H, Overholt A, Devillers R, Gunnarsson T, et al. Fractionated stereotactic radiosurgery with concurrent temozolomide chemotherapy for locally recurrent glioblastoma multiforme: A prospective cohort study. Onco Targets Ther. 2014;7:485–90. http://dx.doi.org/10.2147/OTT.S60358
  101. Grosu AL, Weber WA, Franz M, Stark S, Piert M, Thamm R, et al. Reirradiation of recurrent high-grade gliomas using amino acid PET (SPECT)/CT/MRI image fusion to determine gross tumor volume for stereotactic fractionated radiotherapy. Int J Radiat Oncol Biol Phys. 2005;63(2):511–19. http://dx.doi.org/10.1016/j.ijrobp.2005.01.056
  102. Gutin PH, Iwamoto FM, Beal K, Mohile NA, Karimi S, Hou BL, et al. Safety and efficacy of bevacizumab with hypofractionated stereotactic irradiation for recurrent malignant gliomas. Int J Radiat Oncol Biol Phys. 2009;75(1):156–63. http://dx.doi.org/10.1016/j.ijrobp.2008.10.043
  103. Hundsberger T, Brugge D, Putora PM, Weder P, Weber J, Plasswilm L. Re-irradiation with and without bevacizumab as salvage therapy for recurrent or progressive high-grade gliomas. J Neurooncol. 2013;112(1):133–9. http://dx.doi.org/10.1007/s11060-013-1044-3
  104. Park KJ, Kano H, Iyer A, Liu X, Niranjan A, Flickinger JC, et al. Salvage gamma knife stereotactic radiosurgery followed by bevacizumab for recurrent glioblastoma multiforme: A case-control study. J Neurooncol. 2012;107(2):323–33. http://dx.doi.org/10.1007/s11060-011-0744-9
  105. Torcuator RG, Thind R, Patel M, Mohan YS, Anderson J, Doyle T, et al. The role of salvage reirradiation for malignant gliomas that progress on bevacizumab. J Neurooncol. 2010;97(3):401–7. http://dx.doi.org/10.1007/s11060-009-0034-y
  106. Shapiro LQ, Beal K, Goenka A, Karimi S, Iwamoto FM, Yamada Y, et al. Patterns of failure after concurrent bevacizumab and hypofractionated stereotactic radiation therapy for recurrent high-grade glioma. Int J Radiat Oncol Biol Phys. 2013;85(3):636–42. http://dx.doi.org/10.1016/j.ijrobp.2012.05.031
  107. Minniti G, Agolli L, Falco T, Scaringi C, Lanzetta G, Caporello P, et al. Hypofractionated stereotactic radiotherapy in combination with bevacizumab or fotemustine for patients with progressive malignant gliomas. J Neurooncol. 2015;122(3):559–66. http://dx.doi.org/10.1007/s11060-015-1745-x
  108. Lamborn KR, Yung WK, Chang SM, Wen PY, Cloughesy TF, DeAngelis LM, et al. Progression-free survival: An important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol. 2008;10(2):162–70. http://dx.doi.org/10.1215/15228517-2007-062
  109. Takimoto CH, Calvo E. Principles of oncologic pharmacotherapy. In: Cancer management: A multidisciplinary approach. 11th ed. 2007. Available from: http://www.cancernetwork.com/articles/principles-oncologic-pharmacotherapy-0.
  110. Brandes AA, Tosoni A, Amista P, Nicolardi L, Grosso D, Berti F, et al. How effective is BCNU in recurrent glioblastoma in the modern era? A phase II trial. Neurology. 2004;63(7):1281–4. http://dx.doi.org/10.1212/01.WNL.0000140495.33615.CA
  111. Scoccianti S, Detti B, Sardaro A, Iannalfi A, Meattini I, Leonulli BG, et al. Second-line chemotherapy with fotemustine in temozolomide-pretreated patients with relapsing glioblastoma: A single institution experience. Anticancer Drugs. 2008;19(6):613–20. http://dx.doi.org/10.1097/CAD.0b013e3283005075
  112. Brandes AA, Tosoni A, Franceschi E, Blatt V, Santoro A, Faedi M, et al. Fotemustine as second-line treatment for recurrent or progressive glioblastoma after concomitant and/or adjuvant temozolomide: A phase II trial of Gruppo Italiano Cooperativo di Neuro-Oncologia (GICNO). Cancer Chemother Pharmacol. 2009;64(4):769–75. http://dx.doi.org/10.1007/s00280-009-0926-8
  113. Fabrini MG, Silvano G, Lolli I, Perrone F, Marsella A, Scotti V, et al. A multi-institutional phase II study on second-line Fotemustine chemotherapy in recurrent glioblastoma. J Neurooncol. 2009;92(1): 79–86. http://dx.doi.org/10.1007/s11060-008-9739-6
  114. Addeo R, Caraglia M, De Santi MS, Montella L, Abbruzzese A, Parlato C, et al. A new schedule of fotemustine in temozolomide-pretreated patients with relapsing glioblastoma. J Neurooncol. 2011;102(3):417–24. http://dx.doi.org/10.1007/s11060-010-0329-z
  115. Brandes AA, Tosoni A, Basso U, Reni M, Valduga F, Monfardini S, et al. Second-line chemotherapy with irinotecan plus carmustine in glioblastoma recurrent or progressive after first-line temozolomide chemotherapy: A phase II study of the Gruppo Italiano Cooperativo di Neuro-Oncologia (GICNO). J Clin Oncol. 2004;22(23):4779–86. http://dx.doi.org/10.1200/JCO.2004.06.181
  116. Prados MD, Yung WK, Fine HA, Greenberg HS, Junck L, Chang SM, et al. Phase 2 study of BCNU and temozolomide for recurrent glioblastoma multiforme: North American Brain Tumor Consortium study. Neuro Oncol. 2004;6(1):33–7. http://dx.doi.org/10.1215/S1152851703000309
  117. Gaviani P, Salmaggi A, Silvani A. Combined chemotherapy with temozolomide and fotemustine in recurrent glioblastoma patients. J Neurooncol. 2011;104(2):617–18. http://dx.doi.org/10.1007/s11060-010-0515-z
  118. van den Bent MJ, Brandes AA, Rampling R, Kouwenhoven MC, Kros JM, Carpentier AF, et al. Randomized phase II trial of erlotinib versus temozolomide or carmustine in recurrent glioblastoma: EORTC brain tumor group study 26034. J Clin Oncol. 2009;27(8):1268–74. http://dx.doi.org/10.1200/JCO.2008.17.5984
  119. Wick W, Puduvalli VK, Chamberlain MC, van den Bent MJ, Carpentier AF, Cher LM, et al. Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J Clin Oncol. 2010;28(7):1168–74. http://dx.doi.org/10.1200/JCO.2009.23.2595
  120. Batchelor TT, Mulholland P, Neyns B, Nabors LB, Campone M, Wick A, et al. Phase III randomized trial comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, versus lomustine alone in patients with recurrent glioblastoma. J Clin Oncol. 2013;31(26):3212–18. http://dx.doi.org/10.1200/JCO.2012.47.2464
  121. Brandes AA, Carpentier AF, Kesari S, Sepulveda-Sanchez JM, Wheeler HR, Chinot O, et al. A Phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma. Neuro Oncol. 2016;18(8):1146–56. http://dx.doi.org/10.1093/neuonc/now009
  122. Kappelle AC, Postma TJ, Taphoorn MJ, Groeneveld GJ, van den Bent MJ, van Groeningen CJ, et al. PCV chemotherapy for recurrent glioblastoma multiforme. Neurology. 2001;56(1):118–20. http://dx.doi.org/10.1212/WNL.56.1.118
  123. Schmidt F, Fischer J, Herrlinger U, Dietz K, Dichgans J, Weller M. PCV chemotherapy for recurrent glioblastoma. Neurology. 2006;66(4):587–9. http://dx.doi.org/10.1212/01.wnl.0000197792.73656.c2
  124. Happold C, Roth P, Wick W, Steinbach JP, Linnebank M, Weller M, et al. ACNU-based chemotherapy for recurrent glioma in the temozolomide era. J Neurooncol. 2009;92(1):45–8. http://dx.doi.org/10.1007/s11060-008-9728-9
  125. Reithmeier T, Graf E, Piroth T, Trippel M, Pinsker MO, Nikkhah G. BCNU for recurrent glioblastoma multiforme: Efficacy, toxicity and prognostic factors. BMC Cancer. 2010;10:30. http://dx.doi.org/10.1186/1471-2407-10-30
  126. Brada M, Hoang-Xuan K, Rampling R, Dietrich PY, Dirix LY, Macdonald D, et al. Multicenter phase II trial of temozolomide in patients with glioblastoma multiforme at first relapse. Ann Oncol. 2001;12(2):259–66. http://dx.doi.org/10.1023/A:1008382516636
  127. Yung WK, Albright RE, Olson J, Fredericks R, Fink K, Prados MD, et al. A phase II study of temozolomide vs. procarbazine in patients with glioblastoma multiforme at first relapse. Br J Cancer. 2000;83(5):588–93. http://dx.doi.org/10.1054/bjoc.2000.1316
  128. Taal W, Oosterkamp HM, Walenkamp AM, Dubbink HJ, Beerepoot LV, Hanse MC, et al. Single-agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): A randomised controlled phase 2 trial. Lancet Oncol. 2014;15(9):943–53. http://dx.doi.org/10.1016/S1470-2045(14)70314-6
  129. Brandes AA, Ermani M, Basso U, Amista P, Berti F, Scienza R, et al. Temozolomide as a second-line systemic regimen in recurrent high-grade glioma: A phase II study. Ann Oncol. 2001;12(2):255–7. http://dx.doi.org/10.1023/A:1008336732273
  130. Brandes AA, Ermani M, Basso U, Paris MK, Lumachi F, Berti F, et al. Temozolomide in patients with glioblastoma at second relapse after first line nitrosourea-procarbazine failure: A phase II study. Oncology. 2002;63(1):38–41. http://dx.doi.org/10.1159/000065718
  131. Chan DT, Poon WS, Chan YL, Ng HK. Temozolomide in the treatment of recurrent malignant glioma in Chinese patients. Hong Kong Med J. 2005;11(6):452–6.
  132. Nagane M, Kobayashi K, Ohnishi A, Shimizu S, Shiokawa Y. Prognostic significance of O6-methylguanine-DNA methyltransferase protein expression in patients with recurrent glioblastoma treated with temozolomide. Jpn J Clin Oncol. 2007;37(12):897–906. http://dx.doi.org/10.1093/jjco/hym132
  133. Khan RB, Raizer JJ, Malkin MG, Bazylewicz KA, Abrey LE. A phase II study of extended low-dose temozolomide in recurrent malignant gliomas. Neuro Oncol. 2002;4(1):39–43. http://dx.doi.org/10.1215/15228517-4-1-39
  134. Wick W, Steinbach JP, Kuker WM, Dichgans J, Bamberg M, Weller M. One week on/one week off: A novel active regimen of temozolomide for recurrent glioblastoma. Neurology. 2004;62(11):2113–15. http://dx.doi.org/10.1212/01.WNL.0000127617.89363.84
  135. Brandes AA, Tosoni A, Cavallo G, Bertorelle R, Gioia V, Franceschi E, et al. Temozolomide 3 weeks on and 1 week off as first-line therapy for recurrent glioblastoma: Phase II study from gruppo italiano cooperativo di neuro-oncologia (GICNO). Br J Cancer. 2006;95(9):1155–60. http://dx.doi.org/10.1038/sj.bjc.6603376
  136. Kong DS, Lee JI, Kim WS, Son MJ, Lim DH, Kim ST, et al. A pilot study of metronomic temozolomide treatment in patients with recurrent temozolomide-refractory glioblastoma. Oncol Rep. 2006;16(5):1117–21. http://dx.doi.org/10.3892/or.16.5.1117
  137. Wick A, Felsberg J, Steinbach JP, Herrlinger U, Platten M, Blaschke B, et al. Efficacy and tolerability of temozolomide in an alternating weekly regimen in patients with recurrent glioma. J Clin Oncol. 2007;25(22):3357–61. http://dx.doi.org/10.1200/JCO.2007.10.7722
  138. Balmaceda C, Peereboom D, Pannullo S, Cheung YK, Fisher PG, Alavi J, et al. Multi-institutional phase II study of temozolomide administered twice daily in the treatment of recurrent high-grade gliomas. Cancer. 2008;112(5):1139–46. http://dx.doi.org/10.1002/cncr.23167
  139. Berrocal A, Perez Segura P, Gil M, Balana C, Garcia Lopez J, Yaya R, et al. Extended-schedule dose-dense temozolomide in refractory gliomas. J Neurooncol. 2010;96(3):417–22. http://dx.doi.org/10.1007/s11060-009-9980-7
  140. Perry JR, Belanger K, Mason WP, Fulton D, Kavan P, Easaw J, et al. Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study. J Clin Oncol. 2010;28(12): 2051–7. http://dx.doi.org/10.1200/JCO.2009.26.5520
  141. Kong DS, Lee JI, Kim JH, Kim ST, Kim WS, Suh YL, et al. Phase II trial of low-dose continuous (metronomic) treatment of temozolomide for recurrent glioblastoma. Neuro Oncol. 2010;12(3): 289–96. http://dx.doi.org/10.1093/neuonc/nop030
  142. Hammond S, Norden AD, Lesser GJ, Drappatz J, Fadul CE, Batchelor T, et al. Phase II study of dose-intense temozolomide in recurrent glioblastoma. J Clin Oncol. 2011;29(Suppl 15):2038. http://dx.doi.org/10.1200/jco.2011.29.15_suppl.2038
  143. Santoni M, Paccapelo A, Burattini L, Bianconi M, Cardinali M, Fabbietti L, et al. Protracted low doses of temozolomide for the treatment of patients with recurrent glioblastoma: A phase II study. Oncol Lett. 2012;4(4):799–801. http://dx.doi.org/10.3892/ol.2012.788
  144. Norden AD, Lesser GJ, Drappatz J, Ligon KL, Hammond SN, Lee EQ, et al. Phase 2 study of dose-intense temozolomide in recurrent glioblastoma. Neuro Oncol. 2013;15(7):930–5. http://dx.doi.org/10.1093/neuonc/not040
  145. Han SJ, Rolston JD, Molinaro AM, Clarke JL, Prados MD, Chang SM, et al. Phase II trial of 7 days on/7 days off temozolmide for recurrent high-grade glioma. Neuro Oncol. 2014;16(9):1255–62. http://dx.doi.org/10.1093/neuonc/nou044
  146. Silvani A, Eoli M, Salmaggi A, Lamperti E, Maccagnano E, Broggi G, et al. Phase II trial of cisplatin plus temozolomide, in recurrent and progressive malignant glioma patients. J Neurooncol. 2004; 66(1–2):203–8. http://dx.doi.org/10.1023/B:NEON.0000013479.64348.69
  147. Brandes AA, Basso U, Reni M, Vastola F, Tosoni A, Cavallo G, et al. First-line chemotherapy with cisplatin plus fractionated temozolomide in recurrent glioblastoma multiforme: A phase II study of the Gruppo Italiano Cooperativo di Neuro-Oncologia. J Clin Oncol. 2004;22(9):1598–604. http://dx.doi.org/10.1200/JCO.2004.11.019
  148. Chua SL, Rosenthal MA, Wong SS, Ashley DM, Woods AM, Dowling A, et al. Phase 2 study of temozolomide and Caelyx in patients with recurrent glioblastoma multiforme. Neuro Oncol. 2004;6(1): 38–43. http://dx.doi.org/10.1215/S1152851703000188
  149. Reardon DA, Quinn JA, Rich JN, Desjardins A, Vredenburgh J, Gururangan S, et al. Phase I trial of irinotecan plus temozolomide in adults with recurrent malignant glioma. Cancer. 2005;104(7): 1478–86. http://dx.doi.org/10.1002/cncr.21316
  150. Quinn JA, Jiang SX, Reardon DA, Desjardins A, Vredenburgh JJ, Rich JN, et al. Phase II trial of temozolomide plus o6-benzylguanine in adults with recurrent, temozolomide-resistant malignant glioma. J Clin Oncol. 2009;27(8):1262–7. http://dx.doi.org/10.1200/JCO.2008.18.8417
  151. Groves MD, Puduvalli VK, Gilbert MR, Levin VA, Conrad CA, Liu VH, et al. Two phase II trials of temozolomide with interferon-alpha2b (pegylated and non-pegylated) in patients with recurrent glioblastoma multiforme. Br J Cancer. 2009;101(4):615–20. http://dx.doi.org/10.1038/sj.bjc.6605189
  152. Verhoeff JJ, Lavini C, van Linde ME, Stalpers LJ, Majoie CB, Reijneveld JC, et al. Bevacizumab and dose-intense temozolomide in recurrent high-grade glioma. Ann Oncol. 2010;21(8):1723–7. http://dx.doi.org/10.1093/annonc/mdp591
  153. Reardon DA, Vredenburgh JJ, Desjardins A, Peters K, Gururangan S, Sampson JH, et al. Effect of CYP3A-inducing anti-epileptics on sorafenib exposure: Results of a phase II study of sorafenib plus daily temozolomide in adults with recurrent glioblastoma. J Neurooncol. 2011;101(1):57–66. http://dx.doi.org/10.1007/s11060-010-0217-6
  154. Desjardins A, Reardon DA, Coan A, Marcello J, Herndon JE, 2nd, Bailey L, et al. Bevacizumab and daily temozolomide for recurrent glioblastoma Cancer. 2012;118(5):1302–12. http://dx.doi.org/10.1002/cncr.26381
  155. Sepulveda JM, Belda-Iniesta C, Gil-Gil M, Perez-Segura P, Berrocal A, Reynes G, et al. A phase II study of feasibility and toxicity of bevacizumab in combination with temozolomide in patients with recurrent glioblastoma. Clin Transl Oncol. 2015;17(9):743–50. http://dx.doi.org/10.1007/s12094-015-1304-0
  156. Gan HK, Fichtel L, Lassman AB, Merrell R, van den Bent MJ, Kumthekar P, et al. A phase 1 study evaluating ABT-414 in combination with temozolomide (TMZ) for subjects with recurrent or unresectable glioblastoma (GBM). J Clin Oncol. 2014;32(15_suppl):2021.
  157. Bower M, Newlands ES, Bleehen NM, Brada M, Begent RJ, Calvert H, et al. Multicentre CRC phase II trial of temozolomide in recurrent or progressive high-grade glioma. Cancer Chemother Pharmacol. 1997;40(6):484–8. http://dx.doi.org/10.1007/s002800050691
  158. Reardon DA, Desjardins A, Peters K, Gururangan S, Sampson J, Rich JN, et al. Phase II study of metronomic chemotherapy with bevacizumab for recurrent glioblastoma after progression on bevacizumab therapy. J Neurooncol. 2011;103(2):371–9. http://dx.doi.org/10.1007/s11060-010-0403-6
  159. Reardon DA, Nabors LB, Mason WP, Perry JR, Shapiro W, Kavan P, et al. Phase I/randomized phase II study of afatinib, an irreversible ErbB family blocker, with or without protracted temozolomide in adults with recurrent glioblastoma. Neuro Oncol. 2015;17(3):430–9.
  160. Weller M, Tabatabai G, Kastner B, Felsberg J, Steinbach JP, Wick A, et al. MGMT promoter methylation is a strong prognostic biomarker for benefit from dose-intensified temozolomide rechallenge in progressive glioblastoma: The DIRECTOR trial. Clin Cancer Res. 2015;21(9):2057–64. http://dx.doi.org/10.1158/1078-0432.CCR-14-2737
  161. Franceschi E, Omuro AM, Lassman AB, Demopoulos A, Nolan C, Abrey LE. Salvage temozolomide for prior temozolomide responders. Cancer. 2005;104(11):2473–6. http://dx.doi.org/10.1002/cncr.21564
  162. Boiardi A, Silvani A, Eoli M, Lamperti E, Salmaggi A, Gaviani P, et al. Treatment of recurrent glioblastoma: Can local delivery of mitoxantrone improve survival? J Neurooncol. 2008;88(1):105–13. http://dx.doi.org/10.1007/s11060-008-9540-6
  163. Wick A, Pascher C, Wick W, Jauch T, Weller M, Bogdahn U, et al. Rechallenge with temozolomide in patients with recurrent gliomas. J Neurol. 2009;256(5):734–41. http://dx.doi.org/10.1007/s00415-009-5006-9
  164. Stockhammer F, Misch M, Koch A, Czabanka M, Plotkin M, Blechschmidt C, et al. Continuous low-dose temozolomide and celecoxib in recurrent glioblastoma. J Neurooncol. 2010;100(3):407–15. http://dx.doi.org/10.1007/s11060-010-0192-y
  165. van den Bent MJ, Taal W. Are we done with dose-intense temozolomide in recurrent glioblastoma? Neuro Oncol. 2014;16(9):1161–3. http://dx.doi.org/10.1093/neuonc/nou157
  166. Brada M, Stenning S, Gabe R, Thompson LC, Levy D, Rampling R, et al. Temozolomide versus procarbazine, lomustine, and vincristine in recurrent high-grade glioma. J Clin Oncol. 2010;28(30): 4601–8. http://dx.doi.org/10.1200/JCO.2009.27.1932
  167. Weller M, Tabatabai G, Reifenberger G, Herrlinger U, Pichler J, Schnell O, et al. Dose-intensified rechallenge with temozolomide: One week on/one week off versus 3 weeks on/one week off in patients with progressive or recurrent glioblastoma (DIRECTOR). J Clin Oncol. 2010;28(Suppl. 15):TPS154. http://dx.doi.org/10.1200/jco.2010.28.15_suppl.tps154
  168. Stark-Vance V. Bevacizumab and CPT-11 in the treatment of relapsed malignant glioma. Neuro Oncology. 2005;7(3):369.
  169. Raizer JJ, Grimm S, Chamberlain MC, Nicholas MK, Chandler JP, Muro K, et al. A phase 2 trial of single-agent bevacizumab given in an every-3-week schedule for patients with recurrent high-grade gliomas. Cancer. 2010;116(22):5297–305. http://dx.doi.org/10.1002/cncr.25462
  170. Nagane M, Nishikawa R, Narita Y, Kobayashi H, Takano S, Shinoura N, et al. Phase II study of single-agent bevacizumab in Japanese patients with recurrent malignant glioma. Jpn J Clin Oncol. 2012;42(10):887–95. http://dx.doi.org/10.1093/jjco/hys121
  171. Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, Marcello J, Reardon DA, Quinn JA, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 2007;25(30):4722–9. http://dx.doi.org/10.1200/JCO.2007.12.2440
  172. Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, Dowell JM, Reardon DA, Quinn JA, et al. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007;13(4): 1253–9. http://dx.doi.org/10.1158/1078-0432.CCR-06-2309
  173. Gilbert MR, Wang M, Aldape K, Lassman A, Sorensen AG, Mikkelson T, et al. RTOG 0625: A phase II study of bevacizumab with irinotecan in recurrent glioblastoma (GBM). J Clin Oncol. 2009; 27(15_suppl):89s.
  174. Narayana A, Kelly P, Golfinos J, Parker E, Johnson G, Knopp E, et al. Antiangiogenic therapy using bevacizumab in recurrent high-grade glioma: Impact on local control and patient survival. J Neurosurg. 2009;110(1):173–80. http://dx.doi.org/10.3171/2008.4.17492
  175. Reardon DA, Desjardins A, Vredenburgh JJ, Gururangan S, Sampson JH, Sathornsumetee S, et al. Metronomic chemotherapy with daily, oral etoposide plus bevacizumab for recurrent malignant glioma: A phase II study. Br J Cancer. 2009;101(12):1986–94. http://dx.doi.org/10.1038/sj.bjc.6605412
  176. Hasselbalch B, Lassen U, Hansen S, Holmberg M, Sorensen M, Kosteljanetz M, et al. Cetuximab, bevacizumab, and irinotecan for patients with primary glioblastoma and progression after radiation therapy and temozolomide: A phase II trial. Neuro Oncol. 2010;12(5):508–16.
  177. Sathornsumetee S, Desjardins A, Vredenburgh JJ, McLendon RE, Marcello J, Herndon JE, et al. Phase II trial of bevacizumab and erlotinib in patients with recurrent malignant glioma. Neuro Oncol. 2010;12(12):1300–10. http://dx.doi.org/10.1093/neuonc/noq099
  178. Moller S, Grunnet K, Hansen S, Schultz H, Holmberg M, Sorensen M, et al. A phase II trial with bevacizumab and irinotecan for patients with primary brain tumors and progression after standard therapy. Acta Oncol. 2012;51(6):797–804. http://dx.doi.org/10.3109/0284186X.2012.681063
  179. Reardon DA, Desjardins A, Peters KB, Gururangan S, Sampson JH, McLendon RE, et al. Phase II study of carboplatin, irinotecan, and bevacizumab for bevacizumab naive, recurrent glioblastoma. J Neurooncol. 2012;107(1):155–64. http://dx.doi.org/10.1007/s11060-011-0722-2
  180. Lassen U, Sorensen M, Gaziel TB, Hasselbalch B, Poulsen HS. Phase II study of bevacizumab and temsirolimus combination therapy for recurrent glioblastoma multiforme. Anticancer Res. 2013;33(4):1657–60.
  181. Galanis E, Anderson SK, Lafky JM, Uhm JH, Giannini C, Kumar SK, et al. Phase II study of bevacizumab in combination with sorafenib in recurrent glioblastoma (N0776): A north central cancer treatment group trial. Clin Cancer Res. 2013;19(17):4816–23. http://dx.doi.org/10.1158/1078-0432.CCR-13-0708
  182. Soffietti R, Trevisan E, Bertero L, Cassoni P, Morra I, Fabrini MG, et al. Bevacizumab and fotemustine for recurrent glioblastoma: A phase II study of AINO (Italian Association of Neuro-Oncology). J Neurooncol. 2014;116(3):533–41. http://dx.doi.org/10.1007/s11060-013-1317-x
  183. Lee EQ, Reardon DA, Schiff D, Drappatz J, Muzikansky A, Grimm SA, et al. Phase II study of panobinostat in combination with bevacizumab for recurrent glioblastoma and anaplastic glioma. Neuro Oncol. 2015;17(6):862–7. http://dx.doi.org/10.1093/neuonc/nou350
  184. Ghiaseddin A, Reardon DA, Massey W, Mannerino A, Lipp ES, Herndon JE, et al. Phase II study of bevacizumab and vorinostat for recurrent glioblastoma. J Clin Oncol. 2015;33(15_suppl):2034.
  185. Weathers SP, Han X, Liu DD, Conrad CA, Gilbert MR, Loghin ME, et al. A randomized phase II trial of standard dose bevacizumab versus low dose bevacizumab plus lomustine (CCNU) in adults with recurrent glioblastoma. J Neurooncol. 2016;129(3):487–94. http://dx.doi.org/10.1007/s11060-016-2195-9
  186. Field KM, Simes J, Nowak AK, Cher L, Wheeler H, Hovey EJ, et al. Randomized phase 2 study of carboplatin and bevacizumab in recurrent glioblastoma. Neuro Oncol. 2015;17(11):1504–13. http://dx.doi.org/10.1093/neuonc/nov104
  187. Puduvalli VK, Wu J, Yuan Y, Armstrong TS, Groves MD, Raizer JJ, et al. Brain tumor trials collaborative Bayesian adaptive randomized phase II trial of bevacizumab plus vorinostat versus bevacizumab alone in adults with recurrent glioblastoma (BTTC-1102). J Clin Oncol. 2015;33(15_suppl):2012.
  188. Galanis E, Anderson SK, Anastasiadis P, Tran DD, Jeyapalan SA, Anderson DM, et al. NCCTG N0872 (Alliance): A randomized placebo-controlled phase II trial of bevacizumab plus dasatinib in patients with recurrent glioblastoma (GBM). J Clin Oncol. 2015;33(15_suppl):2004.
  189. Wick W, Brandes AA, Gorlia T, Bendszus M, Sahm F, Taal W, et al. Phase III trial exploring the combination of bevacizumab and lomustine in patients with first recurrence of a glioblastoma: The EORTC 26101 trial. Neuro Oncol. 2015;17(suppl_5):v1.
  190. Pope WB, Lai A, Nghiemphu P, Mischel P, Cloughesy TF. MRI in patients with high-grade gliomas treated with bevacizumab and chemotherapy. Neurology. 2006;66(8):1258–60. http://dx.doi.org/10.1212/01.wnl.0000208958.29600.87
  191. Ali SA, McHayleh WM, Ahmad A, Sehgal R, Braffet M, Rahman M, et al. Bevacizumab and irinotecan therapy in glioblastoma multiforme: A series of 13 cases. J Neurosurg. 2008;109(2):268–72. http://dx.doi.org/10.3171/JNS/2008/109/8/0268
  192. Kang TY, Jin T, Elinzano H, Peereboom D. Irinotecan and bevacizumab in progressive primary brain tumors, an evaluation of efficacy and safety. J Neurooncol. 2008;89(1):113–18. http://dx.doi.org/10.1007/s11060-008-9599-0
  193. Bokstein F, Shpigel S, Blumenthal DT. Treatment with bevacizumab and irinotecan for recurrent high-grade glial tumors. Cancer. 2008;112(10):2267–73. http://dx.doi.org/10.1002/cncr.23401
  194. Norden AD, Young GS, Setayesh K, Muzikansky A, Klufas R, Ross GL, et al. Bevacizumab for recurrent malignant gliomas: Efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70(10):779–87. http://dx.doi.org/10.1212/01.wnl.0000304121.57857.38
  195. Nghiemphu PL, Liu W, Lee Y, Than T, Graham C, Lai A, et al. Bevacizumab and chemotherapy for recurrent glioblastoma: A single-institution experience. Neurology. 2009;72(14):1217–22. http://dx.doi.org/10.1212/01.wnl.0000345668.03039.90
  196. Zuniga RM, Torcuator R, Jain R, Anderson J, Doyle T, Ellika S, et al. Efficacy, safety and patterns of response and recurrence in patients with recurrent high-grade gliomas treated with bevacizumab plus irinotecan. J Neurooncol. 2009;91(3):329–36. http://dx.doi.org/10.1007/s11060-008-9718-y
  197. Chamberlain MC, Johnston SK. Salvage therapy with single agent bevacizumab for recurrent glioblastoma. J Neurooncol. 2010;96(2):259–69. http://dx.doi.org/10.1007/s11060-009-9957-6
  198. Scott BJ, Quant EC, McNamara MB, Ryg PA, Batchelor TT, Wen PY. Bevacizumab salvage therapy following progression in high-grade glioma patients treated with VEGF receptor tyrosine kinase inhibitors. Neuro Oncol. 2010;12(6):603–7. http://dx.doi.org/10.1093/neuonc/nop073
  199. Gil MJ, de Las Penas R, Reynes G, Balana C, Perez-Segura P, Garcia-Velasco A, et al. Bevacizumab plus irinotecan in recurrent malignant glioma shows high overall survival in a multicenter retrospective pooled series of the Spanish Neuro-Oncology Research Group (GEINO). Anticancer Drugs. 2012;23(6):659–65. http://dx.doi.org/10.1097/CAD.0b013e3283534d3e
  200. Goldlust SA, Cavaliere R, Newton HB, Hsu M, Deangelis LM, Batchelor TT, et al. Bevacizumab for glioblastoma refractory to vascular endothelial growth factor receptor inhibitors. J Neurooncol. 2012;107(2):407–11. http://dx.doi.org/10.1007/s11060-011-0768-1
  201. Carvalho BF, Fernandes AC, Almeida DS, Sampaio LV, Costa A, Caeiro C, et al. Second-line chemotherapy in recurrent glioblastoma: A 2-cohort study. Oncol Res Treat. 2015;38(7–8):348–54. http://dx.doi.org/10.1159/000431236
  202. Ballman KV, Buckner JC, Brown PD, Giannini C, Flynn PJ, LaPlant BR, et al. The relationship between six-month progression-free survival and 12-month overall survival end points for phase II trials in patients with glioblastoma multiforme. Neuro Oncol. 2007;9(1):29–38. http://dx.doi.org/10.1215/15228517-2006-025
  203. Wong ET, Hess KR, Gleason MJ, Jaeckle KA, Kyritsis AP, Prados MD, et al. Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J Clin Oncol. 1999;17(8): 2572–8. http://dx.doi.org/10.1200/JCO.1999.17.8.2572
  204. Piccioni DE, Lai A. Deferred use of bevacizumab for recurrent glioblastoma is not associated with diminished efficacy. Neuro Oncol. 2014;16(10):1427–8. http://dx.doi.org/10.1093/neuonc/nou214
  205. Kirson ED, Dbaly V, Tovarys F, Vymazal J, Soustiel JF, Itzhaki A, et al. Alternating electric fields arrest cell proliferation in animal tumor models and human brain tumors. Proc Natl Acad Sci U S A. 2007;104(24):10152–7. http://dx.doi.org/10.1073/pnas.0702916104
  206. Keime-Guibert F, Chinot O, Taillandier L, Cartalat-Carel S, Frenay M, Kantor G, et al. Radiotherapy for glioblastoma in the elderly. N Engl J Med. 2007;356(15):1527–35. http://dx.doi.org/10.1056/NEJMoa065901
  207. Stupp R, Taillibert S, Kanner AA, Kesari S, Steinberg DM, Toms SA, 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. http://dx.doi.org/10.1001/jama.2015.16669
  208. Nabors LB, Portnow J, Ammirati M, Baehring J, Brem H, Brown P, et al. National comprehensive cancer network clinical practice guidelines in oncology, Central Nervous System cancers. Version 1. 2016. Available from: http://www.nccn.org/professionals/physician_gls/f_guidelines.asp.
  209. Bernard-Arnoux F, Lamure M, Ducray F, Aulagner G, Honnorat J, Armoiry X. The cost-effectiveness of tumor-treating fields therapy in patients with newly diagnosed glioblastoma. Neuro Oncol. 2016;18(8):1129–36. http://dx.doi.org/10.1093/neuonc/now102
  210. Mehta M, Wen P, Nishikawa R, Reardon D, Peters K. Critical review of the addition of tumor treating fields (TTFields) to the existing standard of care for newly diagnosed glioblastoma patients. Crit Rev Oncol Hematol. 2017;111:60–5. http://dx.doi.org/10.1016/j.critrevonc.2017.01.005
  211. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, 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. http://dx.doi.org/10.1016/S1470-2045(09)70025-7
  212. Ostrom QT, Gittleman H, Fulop J, Liu M, Blanda R, Kromer C, et al. CBTRUS statistical report: Primary brain and Central Nervous System tumors diagnosed in the United States in 2008–2012. Neuro Oncol. 2015;17(Suppl 4):iv1–iv62. http://dx.doi.org/10.1093/neuonc/nov189
  213. Holdhoff M, Ye X, Blakeley JO, Blair L, Burger PC, Grossman SA, et al. Use of personalized molecular biomarkers in the clinical care of adults with glioblastomas. J Neurooncol. 2012;110(2):279–85. http://dx.doi.org/10.1007/s11060-012-0968-3
  214. Chaichana KL, Garzon-Muvdi T, Parker S, Weingart JD, Olivi A, Bennett R, et al. Supratentorial glioblastoma multiforme: The role of surgical resection versus biopsy among older patients. Ann Surg Oncol. 2011;18(1):239–45. http://dx.doi.org/10.1245/s10434-010-1242-6
  215. Ewelt C, Goeppert M, Rapp M, Steiger HJ, Stummer W, Sabel M. Glioblastoma multiforme of the elderly: The prognostic effect of resection on survival. J Neurooncol. 2011;103(3):611–18. http://dx.doi.org/10.1007/s11060-010-0429-9
  216. Stark AM, Hedderich J, Held-Feindt J, Mehdorn HM. Glioblastoma—The consequences of advanced patient age on treatment and survival. Neurosurg Rev. 2007;30(1):56–61; discussion 61–2.
  217. Malmstrom A, Gronberg BH, Marosi C, Stupp R, Frappaz D, Schultz H, 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. http://dx.doi.org/10.1016/S1470-2045(12)70265-6
  218. Roa W, Brasher PM, Bauman G, Anthes M, Bruera E, Chan A, 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. http://dx.doi.org/10.1200/JCO.2004.06.082
  219. Gallego Perez-Larraya J, Ducray F, Chinot O, Catry-Thomas I, Taillandier L, Guillamo JS, et al. Temozolomide in elderly patients with newly diagnosed glioblastoma and poor performance status: An ANOCEF phase II trial. J Clin Oncol. 2011;29(22):3050–5. http://dx.doi.org/10.1200/JCO.2011.34.8086
  220. Chinot OL, Barrie M, Frauger E, Dufour H, Figarella-Branger D, Palmari J, et al. Phase II study of temozolomide without radiotherapy in newly diagnosed glioblastoma multiforme in an elderly populations. Cancer. 2004;100(10):2208–14. http://dx.doi.org/10.1002/cncr.20224
  221. Laperriere N, Weller M, Stupp R, Perry JR, Brandes AA, Wick W, et al. Optimal management of elderly patients with glioblastoma. Cancer Treat Rev. 2013;39(4):350–7. http://dx.doi.org/10.1016/j.ctrv.2012.05.008
  222. Behm T, Horowski A, Schneider S, Bock HC, Mielke D, Rohde V, et al. Concomitant and adjuvant temozolomide of newly diagnosed glioblastoma in elderly patients. Clin Neurol Neurosurg. 2013;115(10):2142–6. http://dx.doi.org/10.1016/j.clineuro.2013.08.002
  223. Rusthoven CG, Koshy M, Sher DJ, Ney DE, Gaspar LE, Jones BL, et al. Combined-modality therapy with radiation and chemotherapy for elderly patients with glioblastoma in the temozolomide era: A national cancer database analysis. JAMA Neurol. 2016;73(7):821–8. http://dx.doi.org/10.1001/jamaneurol.2016.0839
  224. Perry JR, Laperriere N, O’Callaghan CJ, Brandes AA, Menten J, Phillips C, et al. Short-course radiation plus temozolomide in elderly patients with glioblastoma. N Engl J Med. 2017;376(11):1027–37. http://dx.doi.org/10.1056/NEJMoa1611977
  225. Arvold ND, Tanguturi SK, Aizer AA, Wen PY, Reardon DA, Lee EQ, et al. Hypofractionated versus standard radiation therapy with or without temozolomide for older glioblastoma patients. Int J Radiat Oncol Biol Phys. 2015;92(2):384–9. http://dx.doi.org/10.1016/j.ijrobp.2015.01.017
  226. Minniti G, Scaringi C, Lanzetta G, Terrenato I, Esposito V, Arcella A, et al. Standard (60 Gy) or short-course (40 Gy) irradiation plus concomitant and adjuvant temozolomide for elderly patients with glioblastoma: A propensity-matched analysis. Int J Radiat Oncol Biol Phys. 2015;91(1):109–15. http://dx.doi.org/10.1016/j.ijrobp.2014.09.013
  227. Ostgathe C, Gaertner J, Kotterba M, Klein S, Lindena G, Nauck F, et al. Differential palliative care issues in patients with primary and secondary brain tumours. Support Care Cancer. 2010;18(9): 1157–63. http://dx.doi.org/10.1007/s00520-009-0735-y
  228. Temel JS, Greer JA, Muzikansky A, Gallagher ER, Admane S, Jackson VA, et al. Early palliative care for patients with metastatic non-small-cell lung cancer. N Engl J Med. 2010;363(8):733–42. http://dx.doi.org/10.1056/NEJMoa1000678
  229. Sizoo EM, Pasman HR, Buttolo J, Heimans JJ, Klein M, Deliens L, et al. Decision-making in the end-of-life phase of high-grade glioma patients. Eur J Cancer. 2012;48(2):226–32. http://dx.doi.org/10.1016/j.ejca.2011.11.010
  230. Triebel KL, Martin RC, Nabors LB, Marson DC. Medical decision-making capacity in patients with malignant glioma. Neurology. 2009;73(24):2086–92. http://dx.doi.org/10.1212/WNL.0b013e3181c67bce
  231. Bausewein C, Hau P, Borasio GD, Voltz R. How do patients with primary brain tumours die? Palliat Med. 2003;17(6):558–9. http://dx.doi.org/10.1177/026921630301700615
  232. Faithfull S, Cook K, Lucas C. Palliative care of patients with a primary malignant brain tumour: Case review of service use and support provided. Palliat Med. 2005;19(7):545–50. http://dx.doi.org/10.1191/0269216305pm1068oa
  233. Oberndorfer S, Lindeck-Pozza E, Lahrmann H, Struhal W, Hitzenberger P, Grisold W. The end-of-life hospital setting in patients with glioblastoma. J Palliat Med. 2008;11(1):26–30. http://dx.doi.org/10.1089/jpm.2007.0137
  234. Pace A, Di Lorenzo C, Guariglia L, Jandolo B, Carapella CM, Pompili A. End of life issues in brain tumor patients. J Neurooncol. 2009;91(1):39–43. http://dx.doi.org/10.1007/s11060-008-9670-x
  235. Sizoo EM, Braam L, Postma TJ, Pasman HR, Heimans JJ, Klein M, et al. Symptoms and problems in the end-of-life phase of high-grade glioma patients. Neuro Oncol. 2010;12(11):1162–6. http://dx.doi.org/10.1093/neuonc/nop045
  236. Sizoo EM, Pasman HR, Dirven L, Marosi C, Grisold W, Stockhammer G, et al. The end-of-life phase of high-grade glioma patients: A systematic review. Support Care Cancer. 2014;22(3):847–57. http://dx.doi.org/10.1007/s00520-013-2088-9
  237. Sizoo EM, Koekkoek JA, Postma TJ, Heimans JJ, Pasman HR, Deliens L, et al. Seizures in patients with high-grade glioma: A serious challenge in the end-of-life phase. BMJ Support Palliat Care. 2014;4(1):77–80. http://dx.doi.org/10.1136/bmjspcare-2013-000456
  238. Vecht CJ, Wagner GL, Wilms EB. Treating seizures in patients with brain tumors: Drug interactions between antiepileptic and chemotherapeutic agents. Semin Oncol. 2003;30(6 Suppl 19):49–52. http://dx.doi.org/10.1053/j.seminoncol.2003.11.030
  239. Weller M, Gorlia T, Cairncross JG, van den Bent MJ, Mason W, Belanger K, et al. Prolonged survival with valproic acid use in the EORTC/NCIC temozolomide trial for glioblastoma. Neurology. 2011;77(12):1156–64. http://dx.doi.org/10.1212/WNL.0b013e31822f02e1
  240. Usery JB, Michael LM, Sills AK, Finch CK. A prospective evaluation and literature review of levetiracetam use in patients with brain tumors and seizures. J Neurooncol. 2010;99(2):251–60. http://dx.doi.org/10.1007/s11060-010-0126-8
  241. Koekkoek JA, Postma TJ, Heimans JJ, Reijneveld JC, Taphoorn MJ. Antiepileptic drug treatment in the end-of-life phase of glioma patients: A feasibility study. Support Care Cancer. 2016;24(4):1633–8. http://dx.doi.org/10.1007/s00520-015-2930-3
  242. Rooney AG, Brown PD, Reijneveld JC, Grant R. Depression in glioma: A primer for clinicians and researchers. J Neurol Neurosurg Psychiatry. 2014;85(2):230–5. http://dx.doi.org/10.1136/jnnp-2013-306497
  243. Alper K, Schwartz KA, Kolts RL, Khan A. Seizure incidence in psychopharmacological clinical trials: An analysis of Food and Drug Administration (FDA) summary basis of approval reports. Biol Psychiatry. 2007;62(4):345–54. http://dx.doi.org/10.1016/j.biopsych.2006.09.023
  244. Taillandier L, Blonski M, Darlix A, Hoang Xuan K, Taillibert S, Cartalat Carel S, et al. Supportive care in neurooncology. Rev Neurol (Paris). 2011;167(10):762–72. http://dx.doi.org/10.1016/j.neurol.2011.08.008
  245. Walbert T, Khan M. End-of-life symptoms and care in patients with primary malignant brain tumors: A systematic literature review. J Neurooncol. 2014;117(2):217–24. http://dx.doi.org/10.1007/s11060-014-1393-6