Skip to main content
Current Oncology logoLink to Current Oncology
. 2011 Jun;18(3):e126–e136. doi: 10.3747/co.v18i3.755

Canadian recommendations for the treatment of recurrent or progressive glioblastoma multiforme

JC Easaw , WP Mason, J Perry, N Laperrière, DD Eisenstat, R Del Maestro, K Bélanger, D Fulton, D Macdonald; for the Canadian Glioblastoma Recommendations Committee*
PMCID: PMC3108873  PMID: 21655151

Abstract

Recommendation 1: Multidisciplinary Approach

To optimize treatment outcomes, the management of patients with recurrent glioblastoma should be individualized and should involve a multidisciplinary team approach, including neurosurgery, neuropathology, radiation oncology, neuro-oncology, and allied health professions.

Recommendation 2: Imaging

The standard imaging modality for assessment of recurrent glioblastoma is Gd-enhanced magnetic resonance imaging (mri). Tumour recurrence should be assessed according to the criteria set out by the Response Assessment in Neuro-Oncology Working Group. The optimal timing and frequency of mri after chemoradiation and adjunctive therapy have not been established.

Recommendation 3: Pseudo-progression

Progression observed by mri after chemoradiation can be pseudo-progression. Accordingly, treated patients should not be classified as having progressive disease by Gd-enhancing mri within the first 12 weeks after the end of radiotherapy unless new enhancement is observed outside the radiotherapy field or viable tumour is confirmed by pathology at the time of a required re-operation. Adjuvant temozolomide should be continued and follow-up imaging obtained.

Recommendation 4: Repeat Surgery

Surgery can play a role in providing symptom relief and confirming tumour recurrence, pseudo-progression, or radiation necrosis. However, before surgical intervention, it is essential to clearly define treatment goals and the expected impact on prognosis and the patient’s quality of life. In the absence of level 1 evidence, the decision to re-operate should be made according to individual circumstances, in consultation with the multidisciplinary team and the patient.

Recommendation 5: Re-irradiation

Re-irradiation is seldom recommended, but can be considered in carefully selected cases of recurrent glioblastoma.

Recommendation 6: Systemic Therapy

Clinical trials, when available, should be offered to all eligible patients. In the absence of a trial, systemic therapy, including temozolomide rechallenge or anti-angiogenic therapy, may be considered. Combination therapy is still experimental; optimal drug combinations and sequencing have not been established.

Keywords: Glioblastoma multiforme, guidelines, pseudo-progression, re-irradiation, re-operation, chemotherapy, recurrence, progression

1. INTRODUCTION

Glioblastoma multiforme (gbm) is a World Health Organization grade iv astrocytoma and the most common type of primary brain tumour in adults; its estimated age-adjusted incidence in North America is 3.0 per 100,000 population 1,2. An aggressive malignancy, gbm has an estimated 2-year survival rate of 8.7% in the absence of therapy 1. The median duration of survival with maximal treatment is 12–18 months 3.

Glioblastomas are characterized by high mitotic activity, microvascular proliferation, and necrosis 3. De novo (primary) gbm is more common in older patients (mean age: 55 years) 4, and the tumours are typically characterized by loss of heterozygosity on chromosome 10, overexpression or mutation of the epidermal growth factor receptor (egfr), and alteration or loss of the tumour-suppressor protein pten (phosphatase and tensin homolog) 59. Secondary gbm develops more slowly from lower-grade tumours and typically occurs in younger patients. Genetic alterations may include TP53 mutation or overexpression of platelet-derived growth factor receptor (pdgfr) alpha 10.

In newly-diagnosed glioblastoma, methylation of the O6-methylguanine dna methyltransferase (mgmt) promoter has been shown to predict response to alkylating agents such as temozolomide, 1,3-bis(2-chloroethyl)-1-nitrosourea (bcnu, carmustine), and cyclophosphamide 11,12. After phase iii study of combined radiotherapy and temozolomide in gbm 13, an analysis of mgmt promoter methylation status found that median survival was longer with than without promoter methylation (18.2 months vs. 12.2 months respectively) 14.

Accordingly, current Canadian guidelines recommend that newly-diagnosed glioblastoma be treated with maximal tumour resection, postoperative external-beam radiotherapy (60 Gy in 2-Gy fractions) with concurrent temozolomide (75 mg/m2), and adjuvant temozolomide (150–200 mg/m2) for 6 cycles 15.

Despite optimal treatment, the estimated recurrence rate is in excess of 90%, with most patients recurring fewer than 4 cm from the site of the original tumour 1618. “Recurrent glioblastoma” has been variously defined and may be difficult to distinguish from progression. Because overall prognosis seems to depend little on the ability to make a distinction between recurrent and progressive disease, those two terms are used interchangeably for the purposes of the present recommendations.

Because of a paucity of clinical trials at the time of writing, the management of recurrent glioblastoma was not adequately addressed by the previously-published Canadian recommendations. In the intervening period, new data on the use of agents such as temozolomide and bevacizumab in recurrent glioblastoma have altered the treatment paradigm. The recommendations that follow were developed by a multidisciplinary panel of Canadian neuro-oncologists, neurosurgeons, and radiation oncologists in accordance with the levels of evidence set out by the American Society for Clinical Oncology (Table i) 19. They are meant to guide the optimization of patient management in recurrent or progressive glioblastoma.

TABLE I.

Evidence levels and recommendation grades used in the consensus meetinga

Item Source or quality
Evidence
i Meta-analysis of well-designed controlled studies; high-quality randomized trial
ii At least one well-designed study; lower-quality randomized trial
iii Quasi-experimental study—for example, nonrandomized, uncontrolled, case–control
iv Non-experimental study—for example, comparative, case studies
v Case reports and clinical examples
Recommendation
A Type i or consistent findings from multiple studies of types ii, iii, or iv
B Type ii, iii, or iv, findings generally consistent
C Type ii, iii, or iv, inconsistent findings
D Little or no empiric evidence
a

Adapted from Somerfield et al., 2000 19.

2. METHODS

The Canadian Glioblastoma Recommendations Committee, comprising medical oncologists, surgical oncologists, radiation oncologists, and medical imaging specialists met in March 2010 to develop recommendations for the management of recurrent or progressive glioblastoma. Draft guidelines were based on expert opinion and a literature review. For the systematic literature review, the medline database was searched for all published studies before June 2010, and that search was supplemented by a search of the American Society for Clinical Oncology annual meeting abstracts for 2005–2010. Search terms included “glioblastoma”; “gbm” (glioblastoma multiforme); “progressive”; “recurrent”; “surgery”; “radiotherapy”; “pseudoprogression”; “stereotactic radiosurgery” and its abbreviation “srs”; “fractionated”; “imrt” (intensity-modulated radiotherapy); and generic and brand names of agents for chemotherapy and biologic therapy. Because of the continuing paucity of randomized controlled trials, relevant articles necessarily included retrospective analyses and case series. Draft recommendations were prepared by JCE and further refined at a committee meeting in May 2010. Revisions by the contributing author were coordinated by JCE into a final manuscript for submission.

3. RECOMMENDATIONS

3.1. Multidisciplinary Approach

To optimize treatment outcomes, the management of patients with recurrent glioblastoma should be individualized and should involve a multidisciplinary team approach, including neurosurgery, neuropathology, radiation oncology, neuro-oncology, and allied health professions.

The care path of patients with recurrent glioblastoma is complex, and cooperation and integration of services from multiple health care specialties and institutions are required. Factors that will influence the management approach include patient age, performance status, histology, extent of initial resection, response to initial therapy, time since diagnosis, and whether the recurrence is local or diffuse. To better inform decision-making, patients should receive a brain tumour information package that will help them understand glioblastoma and their treatment options. We encourage tumour banking whenever possible.

3.2. Imaging

The standard imaging modality for assessment of recurrent glioblastoma is Gd-enhanced magnetic resonance imaging (mri) (grade of recommendation: A). The optimal timing and frequency of mri in the adjuvant setting have not been established, but scans are often performed every 2–3 months while the patient is on therapy.

We recommend that a radiology evaluation be conducted using the recently published Response Assessment in Neuro-Oncology (rano) criteria 20. While incorporating many of the elements from the previously used Macdonald criteria 21, the rano criteria

  • modify the definition of measurable disease, addressing subcentimetre lesions, tumour cysts, and surgical cavities;

  • include evaluation of non-enhancing mri (T2-weighted or fluid attenuation inversion recovery tumour volume) changes; and

  • operationalize the definition of pseudo-progression (see the pseudo-progression recommendation, next subsection).

As previously described in the Macdonald criteria, treatment response is defined as a minimum decrease of 50% in tumour area (defined as the product of the maximal cross-sectional enhancing diameters). Progression is defined as a 25% increase.

3.3. Pseudo-progression

After chemoradiation, the diagnosis by mri of radiologic tumour progression can be challenging. Tumour recurrence should be assessed according to the rano criteria 19 (grade of recommendation: A). Progression observed by mri after chemoradiation can be pseudo-progression in 20%–50% of cases, particularly in patients treated with concurrent radiation and temozolomide 2224.

Pseudo-progression is diagnosed retrospectively when the post-radiotherapy mri shows increased tumour enhancement that stabilizes or improves with the same or no further therapy. This phenomenon was first described by Hoffman et al., who reported clinical deterioration suggestive of progression in 49% of patients treated with radiotherapy and carmustine, among whom 28% subsequently improved with the same or no further therapy 25. It has been suggested that radiation-related vascular effects result in increased capillary permeability, which in turn is associated with increased enhancement and fluid leakage into the interstitial space and brain edema 26,27.

A recent Canadian study examined pseudo-progression in 104 evaluable glioblastoma patients 24. Pseudo-progression was defined as early progression, with disease stabilization in the absence of salvage therapy for at least 6 months after completion of chemoradiotherapy with temozolomide. Early progression was observed in 26% of the patients, 32% of whom had pseudo-progression. Median survival was significantly prolonged for patients with pseudo-progression as compared with those showing true progression (124.9 weeks vs. 36.0 weeks). Pseudo-progression appears to be more common in patients with mgmt promoter methylation. Brandes et al. reported lesion enlargement at first mri in 50 of 103 patients who had received a regimen of radiotherapy with temozolomide and subsequent maintenance temozolomide; 32 of the 50 were subsequently classified as having pseudo-progression 28. Of 23 patients with mgmt promoter methylation, 21 (91%) showed pseudo-progression; among patients lacking such methylation (n = 27), 11 (41%) showed pseudo-progression.

If improperly characterized, pseudo-progression may lead to either premature termination of therapy or unnecessary debulking surgeries. Accordingly, adjuvant temozolomide 150 mg/m2 on a 5-days-in-28-days schedule (200 mg/m2 at the second cycle if well tolerated) should be continued for a minimum of 3 cycles, after which Gd-enhancing mri should be used to ascertain progression. In the presence of new enhancement outside the radiotherapy field in the first 3 months of adjuvant temozolomide (which is suggestive of true progression), alternative adjuvant regimens should be considered.

Other imaging techniques—such as proton magnetic resonance spectroscopy as it becomes more widely available—may assist in differentiating pseudo-progression from true progression. High choline levels generally indicate tumour cell proliferation and disease progression; low choline levels have been reported in radiation necrosis 29. Weybright et al. observed that the ratios of choline/creatine and choline/N-acetylaspartate are higher in tumour than in radiation injury 30. Assessment by diffusion tensor imaging of the mean apparent diffusion coefficient may also help to differentiate tumour from radiation-induced changes 31.

3.4. Repeat Surgery

Repeat surgery may play a role in debulking tumour, providing symptom relief, and differentiating tumour recurrence from pseudo-progression or radiation necrosis (grade of recommendation: B). However, before surgical intervention, it is essential to clearly define treatment goals and the effect on prognosis and quality of life for the patient. In the absence of level 1 evidence, the decision to re-operate should be made according to individual circumstances and in consultation with the multidisciplinary team and the patient.

A number of case series have reported modest benefits after re-operation in selected patients, with the caveat that patient selection bias may have influenced the results. In general, patients with a high Karnofsky performance status (kps) score (>70) and those with a tumour in a favourable location appear to be candidates for repeat surgery.

In an early review of 55 consecutive patients with glioblastoma or anaplastic astrocytoma (aa) undergoing repeat surgery, Ammirati et al. reported a median survival of 36 weeks with a mortality rate of 1.6% and a morbidity rate of 16% 32. The patient’s kps score before surgery and the extent of surgical resection were independent factors for survival post surgery. Other groups have reported survival times of 36–76 weeks 3336. However, Guyotat and co-authors noted that, even in carefully selected glioblastoma patients, the improvement in survival was only 3 months (5 months with repeat surgery vs. 2 months without) 36. Other authors have suggested that repeat surgery should be considered only in patients who are candidates for salvage chemotherapy or srs 37.

Compared with surgery alone, implantation of biodegradable chemotherapy wafers (for example, wafers with carmustine) at the time of repeat surgery may prolong survival; however, this practice remains highly controversial 38. Preliminary evidence suggests that survival may be improved in patients with mgmt promoter hypermethylation at recurrence 39. Survival in those cases is reported to be in the range of 25–35 weeks 40, but may be adversely affected by postoperative complications such as bone marrow suppression, infection, and poor wound healing 41,42.

3.5. Re-irradiation

Radiation therapy is seldom recommended, but may be considered in carefully selected cases of recurrent glioblastoma (grade of recommendation: C).

Radiosurgery [Gamma Knife (Elekta, Stockholm, Sweden), CyberKnife (Accuray, Sunnyvale, CA, U.S.A.), linear accelerator) delivers a radiation dose in one or several fractions (“fractionated srs”). Although studies have been conducted, the data do not support the use of re-irradiation as a standard treatment for recurrent gbm. The choice to re-irradiate depends on several factors, including the size and location of the tumour, prior radiotherapy dose, time since last radiation, and target volume. As a general rule, an increase in the fraction size is associated with an increased risk of adverse effects 43.

Stereotactic radiosurgery has the advantages of sparing normal tissue, of shortening recovery time, and potentially of being delivered on an outpatient basis in selected patients. Median survival after srs is in the range of 8–16 months 4448. Potential adverse effects include radiation necrosis, edema, hydrocephalus, and worsening of previous symptoms. Hypofractionated srs has similar survival outcomes, in the range of 9–12 months 4951.

Newer approaches include imrt and three-dimensional conformal radiation therapy. Intensity-modulated radiotherapy is able to deliver highly conformal radiation doses with a reduced dose to areas adjacent to critical tissues, such as the brainstem and optic chiasm 52,53. The imrt technique may minimize adverse effects, but compared with srs, it is more costly and has not been shown to improve outcomes 5456.

3.6. Systemic Therapy

Clinical trials, when available, should be offered to all eligible patients. In the absence of a trial, systemic therapy may be considered, including temozolomide rechallenge (grade of recommendation: B) and anti-angiogenic therapy such as bevacizumab (grade of recommendation: B).

In the pre-temozolomide era, Wong et al. 57 reported the pooled results of eight phase ii chemotherapy trials in recurrent glioblastoma or aa (n = 375). The chemotherapeutic regimens assessed were interferon-β (ifnβ), ifn-β with 13-cis-retinoic acid, menogaril, carboplatin, and carboplatin–fluorouracil/procarbazine. The overall 6-month progression-free survival (pfs) rate was 15% in gbm. The 1-year overall survival (os) rate was 32%, and median os was 30 weeks.

3.6.1. Temozolomide

Since the emergence of reports showing a survival benefit with the addition of temozolomide to radiotherapy in the first-line setting 13, temozolomide has been the most studied agent in recurrent glioblastoma, either as monotherapy or as the backbone of a combination regimen. Many trials evaluating temozolomide in the recurrent setting have also included anaplastic glioma, which appears to be highly responsive to repeat temozolomide therapy 58. Table ii summarizes data from trials that evaluated temozolomide in recurrent gbm only or that separated out the effect on a gbm subgroup. Several dosing regimens have been tested, including the standard temozolomide dosing regimen of 150–200 mg/m2 for the first 5 days of a 28-day cycle 5961,63,66 and novel schedules such as 150 mg/m2 daily, 1 week on, 1 week off 67,70; 75 mg/m2 daily, 3 weeks on, 1 week off 64; and 75 mg/m2 daily for 42 of 70 days 62. An alternative approach has been to administer continuous low-dose temozolomide at 40–50 mg/m2 daily 65,66,69 or to start with a 200 mg/m2 loading dose followed by a lower-dose regimen (for example, 90 mg/m2 every 12 hours 68). With these various approaches, the estimated 6-month pfs rate has been reported to be 24%–44%.

TABLE II.

Temozolomide (tmz) monotherapy trials in recurrent or progressive glioblastoma multiforme (gbm)a

Study tmz regimen Pts (n) 6-Monthpfs (%)
Yung et al., 2000 59 150–200 mg/m2 daily × 5 days every 28 days 112 21
Brandes et al., 2001 60 150 mg/m2 daily × 5 days every 28 days 22 31.8
Brandes et al., 2002 61 150 mg/m2 daily × 5 days every 28 days 42 24
Khan et al., 2002 62 75 mg/m2 daily × 42 days every 70 days 28 19
Chan et al., 2005 63 200 mg/m2 daily × 5 days every 28 days 13 21.0
Brandes et al., 2006 64 75 mg/m2 daily × 21 days every 28 days 33 30.3
Kong et al., 2006 65 40 mg/m2 daily (3 months) 12 58.3
Nagane et al., 2007 66 150–200 mg/m2 daily × 5 days every 28 days 30 22.2
Wick et al., 2007 67 150 mg/m2 on days 1–7 and days 15–21 every 28 days (1 week on, 1 week off) 64 43.8
Balmaceda et al., 2008 68 200 mg/m2 initial dose, then 9 × 90–100 mg/m2 every 12 hours every 28 days 68 35
Perry et al., 2010 58 50 mg/m2 daily, continuous 91 23.9
Kong et al., 2010 69 40–50 mg/m2 daily 38 32.5
Wick et al., 2009 70,b 75 mg/m2 daily (days 1–42 during rt), plus 150–200 mg/m2 daily × 5 days every 28 days;
OR
150–200 mg/m2 daily × 5 days every 28 days;
OR
150 mg/m2 daily × 1 week on, 1 week off;
OR
75 mg/m2 daily × 21 days every 28 days;
OR
40 mg/m2 daily, continuousc
47 27.7
a

Data presented for gbm patients only.

b

Retrospective review.

c

Eleven patients also received 13-cis-retinoic acid or pegylated liposomal doxorubicin.

Pts = patients; rt = radiotherapy.

The rationale for using metronomic chemotherapy (that is, a continuous low-dose regimen) is that this approach may deplete mgmt. The prognostic value of mgmt promoter methylation at progression is unclear. Some of the available data suggest that mgmt status influences the pattern of recurrence 71, but a retrospective analysis by Brandes et al. 72 found that mgmt methylation status changed from first to second surgery in 37% of patients and was no longer predictive of outcome after the second surgery. In an analysis of patients treated with radiotherapy alone or radiotherapy and temozolomide in the joint studies by the European Organisation for Research and Treatment of Cancer (26981, 22981) and the National Cancer Institute of Canada (ce.3), recurrence patterns were found to be independent of mgmt promoter methylation 73. In the phase ii rescue trial, 6-month pfs results were also independent of the mgmt status of patients 58. A phase ii trial of temozolomide in combination with the mgmt pseudo-substrate O6-benzylguanine did not produce superior efficacy in recurrent glioblastoma 74.

A further hypothesis is that metronomic temozolomide may limit endothelial cell recovery and upregulate thrombospondin 1, leading to an anti-angiogenic effect 7578. In vitro studies have indicated that low-dose temozolomide, at a concentration equivalent to 20 mg/m2 every 8 hours, inhibits angiogenesis 79. Preliminary studies have reported that continuous low-dose temozolomide plus a cyclooxygenase 2 inhibitor has anti-angiogenic effects and is well tolerated 80,81. Additional research in this area is required.

The rescue trial examined response to continuous temozolomide at a low dose (50 mg/m2 daily, 28 of 28 days) in patients previously treated with the standard temozolomide adjuvant regimen 58. The best responses were seen in patients with early progression (before completion of 6 cycles of adjuvant therapy—6-month pfs: 27.3%) and in previous responders who progressed more than 2 months after completing adjuvant therapy (6-month pfs: 35.7%). Patients who progressed while receiving extended adjuvant temozolomide had a poor response (6-month pfs: 7.4%) and would therefore be candidates for alternative salvage chemotherapy.

Accordingly, treatment with temozolomide (for example, 50 mg/m2 daily) is an option for patients who have completed a 6-month course of adjuvant temozolomide and have experienced a drug-free period of at least 2 months, or for those who progress 3–6 months after completing adjuvant temozolomide therapy. Other agents should be considered in patients who progress after receiving prolonged (>1 year) adjuvant temozolomide.

An alternative dosing schedule used in one phase ii trial was temozolomide 150 mg/m2 on days 1–7 and 15–21 in a 28-day cycle (1 week on, 1 week off) 67. The 6-month pfs with that regimen was 43.8%, but it is important to note that only 9 of 64 subjects had received prior temozolomide. At entry, 22 patients were chemotherapy-naïve, 30 had received prior nimustine–teniposide, 3 had received procarbazine–lomustine–vincristine (pcv), and 9 had received lomustine–temozolomide. A retrospective review by the same authors reported a 6-month pfs of 27.7% for gbm patients rechallenged with temozolomide 70, results that are comparable to those seen with the continuous low-dose temozolomide regimen.

New trials will undoubtedly evaluate new cytotoxic regimens in recurrent gbm. One of the key lessons from the rescue study is that recurrent patients cannot be considered a homogeneous group. Patients who recur with gbm typically do so during the first 6 months of conventional temozolomide adjuvant therapy, after a break from conventional therapy, or immediately after prolonged adjuvant treatment. The rescue study demonstrated that survival rates were different in these 3 patient populations. Failure to recognize the different subgroups of recurrent patients may underestimate the potential benefits of cytotoxic agents that may have activity confined to discrete patient cohorts.

3.6.2. Anti-angiogenic Therapies

Glioblastomas are highly vascularized tumours, which express vascular endothelial growth factor (vegf) and vegf receptor, providing a rationale for the use of anti-angiogenic agents such as bevacizumab 82. A phase ii trial comparing bevacizumab 10 mg/kg alone or in combination with irinotecan 340 mg/m2 or 125 mg/m2 every 2 weeks in 167 glioblastoma patients demonstrated 6-month pfs rates of 42.6% with monotherapy and 50.3% with combination therapy 83. The median duration of response was 4.3–5.6 months. Importantly, with bevacizumab, the use of steroids either stabilized or decreased in this patient population. Bevacizumab was generally well tolerated, although grade 3 or greater side effects were common (46.4% of patients in the monotherapy arm); adverse effects included hypertension, seizure, and thromboembolic events.

Alternative dosing regimens using bevacizumab have been studied (10 mg/kg every 2 weeks, or 15 mg/kg every 3 weeks plus irinotecan) with reported 6-month pfs rates of 29%–64% 8486. In addition to being used as monotherapy, bevacizumab has been combined with other drugs in the recurrent setting. Recently, a combination of bevacizumab and oral etoposide was observed to be no more effective and more toxic than bevacizumab monotherapy 87.

Another anti-angiogenic agent, cediranib, was recently evaluated in a phase ii trial of 31 subjects with recurrent gbm, and an encouraging 6-month pfs of 25.8% was observed 88. Grades 3 and 4 toxicities included hypertension, diarrhea, and fatigue. Toxicities were generally manageable, with dose reductions or drug interruptions being reported in 15 of 31 patients. The phase iii regal trial of cediranib in combination with lomustine is ongoing and will further clarify the role of that agent in recurrent gbm. Other anti-angiogenic agents, such as thalidomide and pazopanib, appear to offer only modest benefits 89,90.

Although the foregoing results indicate an encouraging clinical effect with selected anti-angiogenic agents, some concerns have also been raised about their use. A pooled analysis of recurrent glioblastoma patients treated with bevacizumab or cediranib found that anti-angiogenic therapies benefited pfs but not os 91. Also, anti-angiogenic agents may directly interfere with Gd uptake in tumours, making it difficult to ascertain tumour margins and to evaluate clinical response 92. A further concern is the effect of anti-angiogenic agents on tumour biology. A preliminary study found that, when exposed to anti-angiogenic therapy, glioblastoma upregulates other pro-angiogenic factors and invades normal brain tissue through upregulation of matrix metalloproteinases, thereby shifting glioblastoma to a more infiltrative phenotype that is undetectable with enhancing mri 93,94. Indeed, a non-enhancing pattern of tumour progression appears to be correlated with worse survival 95. Overall, early data regarding the use of anti-angiogenic agents are promising, but additional research is needed to clarify the effects of these agents used alone or in combination with conventional chemotherapies.

3.6.3. Combination Therapy and Nitrosourea-Based Regimens

Combination therapy is still experimental, and optimal drug combinations and sequencing have not been established.

Two phase ii trials reported improved efficacy with the combination of temozolomide 150–200 mg/m2 daily for 5 of 30 days and either short-acting ifnα2b 4×106 U for 3 of 7 days (6-month pfs: 31%) or pegylated ifn 0.5 μg/kg weekly (6-month pfs: 38%) 96. Use of this regimen may be limited by the frequency of grades 3 and 4 toxicities such as fatigue, leucopenia, and thrombocytopenia. Temozolomide has also been combined with conventional chemotherapeutic agents such as mitoxantrone 97, irinotecan 98, and pegylated doxorubicin, and appears to be well tolerated 99. A phase ii trial of cisplatin (40 mg/m2 on days 1 and 2) and temozolomide (200 mg/m2 on days 2–6) every 4 weeks in heavily pretreated patients with recurrent glioblastoma reported a 6-month pfs of 35%, but grades 4 and 5 side effects were not uncommon 100. Another report evaluating that combination suggested that temozolomide was better tolerated when fractionated (70 mg/m2 every 12 hours, days 2–6 every 4 weeks), although it should be noted that subjects in that study were chemotherapy-naïve 101. An important clinical consideration is that temozolomide does not appear to be cross-resistant with other chemotherapeutic agents 102104. Thus, selected patients with continued progression on a temozolomide regimen may respond to salvage chemotherapy or may be considered for entry into a clinical trial.

Adjuvant nitrosourea-based regimens such as carmustine, lomustine, and pcv were commonly used before the advent of temozolomide. Some studies have reported a combined complete and partial response rate as high as 11% and a 25% rate of stable disease with adjuvant pcv 105, but a large trial by the U.K. Medical Research Council found no benefit with pcv plus radiotherapy as compared with radiotherapy alone 106.

Several recent studies have investigated salvage nitrosoureas in progressive glioblastoma post-temozolomide. Fotemustine has been studied most extensively in that setting, with the 6-month pfs reported to be 20.9%–52% 104,107,108. The combination of fotemustine–procarbazine may provide some benefit with respect to partial response and stable disease, but it does not appear to improve 6-month pfs 103. The use of nimustine is not advised because of its modest efficacy and high rate of hematologic toxicity 109. Salvage cyclophosphamide at the time of first or second recurrence post-temozolomide has also been reported to have modest efficacy (6-month pfs: 20%) with more acceptable toxicity 110.

3.6.4. Novel Therapies

A number of novel therapies have been investigated, but have demonstrated little clinical benefit. A subset of glioblastomas exhibit overexpression of egfr and EGFR gene amplification 111, and several trials have investigated the egfr tyrosine kinase inhibitors erlotinib, gefitinib, and lapatinib. However, a phase ii trial that compared erlotinib with active controls (temozolomide or carmustine) reported a 6-month pfs of only 11.4% as compared with 24% for controls 112. Other trials have reported little or no benefit for erlotinib used as a single agent or in combination with carboplatin or sirolimus, a mammalian target of rapamycin (mtor) inhibitor 113115. Similarly, little benefit was observed with gefitinib alone or in combination with the mtor inhibitor everolimus 116,117. A Canadian phase i/ii trial of lapatinib was stopped early because of a lack of efficacy 118.

Other targeted therapies, such as the histone deacetylase inhibitor vorinostat 119 and the protein kinase C and phosphoinositide 3 kinase (pi3k)/Akt inhibitor enzastaurin 120,121, have demonstrated little antitumour effect when used as monotherapy. More promising is cilengitide, an inhibitor of αvβ3 and αvβ5 integrin receptors. A phase ii trial reported a 6-month pfs of 15% with cilengitide 2000 mg twice weekly, and in vitro data suggest cilengitide may promote temozolomide delivery to tumour cells when used in a combination approach 122,123. Other novel therapies currently being investigated target pi3k/Akt (to overcome radioresistance), tumour cell growth (by inhibiting the farnesyl transferase pathway—examples include tipifarnib, lonafarnib), and the angiogenesis and angiopoietin pathways (for example, pdgfr, Src, mtor, Ras). Additional research is needed to determine the effectiveness of these agents alone and in combination with current therapies.

4. SUMMARY

Numerous genetic alterations that influence tumour cell growth and proliferation have been identified in newly-diagnosed and recurrent glioblastoma. These alterations may be targets for novel therapies. Significant research is now being conducted and is likely to provide important insights into treatment strategies that target multiple pathways and that better control tumour infiltration and progression. Currently, selected patients may benefit from repeat surgery, re-irradiation, salvage chemotherapy, and biologic agents. The recommendations presented here are consistent with those produced by the National Comprehensive Cancer Network 124. Taking into account efficacy, ease of administration, and toxicity, many Canadian centers have opted for a metronomic dose schedule of temozolomide (for example, 50 mg/m2 daily) as the initial choice of treatment. However, anti-angiogenic therapies are also promising, and further studies will help to clarify the controversies outlined earlier. Using advances in molecular profiling, clinicians will be able to stratify patients by their response to alkylating chemotherapies, thus highlighting those who would benefit from an alternative approach.

5. ACKNOWLEDGMENT

Funding for the Canadian Glioblastoma Recommendations Committee meeting was provided by Merck Canada. The authors thank Steven Manners for his help with manuscript preparation.

Footnotes

6. CONFLICT OF INTEREST DISCLOSURES

All the authors declare that they have no conflicts to report.

7. REFERENCES

  • 1.Barnholtz-Sloan JS, Sloan AE, Schwartz AG. Cancer of the brain and other central nervous system. In: Ries LAG, Young JL, Keel GE, Eisner MP, Lin YD, Horner MJ, editors. Cancer Survival Among Adults: U.S. SEER Program, 1988–2001, Patient and Tumor Characteristics. Bethesda, MD: National Cancer Institute, Surveillance, Epidemiology, and End Results program; 2007. SEER Survival Monograph. Publication No. 07-6215. [Available online at: seer.cancer.gov/publications/survival/surv_brain.pdf; cited March 25, 2010] [Google Scholar]
  • 2.Deorah S, Lynch CF, Sibenaller ZA, Ryken TCR. Trends in brain cancer incidence and survival in the United States: Surveillance, Epidemiology, and End Results program, 1973 to 2001. Neurosurg Focus. 2006;20(4):E1. doi: 10.3171/foc.2006.20.4.E1. [DOI] [PubMed] [Google Scholar]
  • 3.Hou LC, Veeravagu A, Hsu AR, Tse VC. Recurrent glioblastoma multiforme: a review of natural history and management options. Neurosurg Focus. 2006;20(4):E5. doi: 10.3171/foc.2006.20.4.2. [DOI] [PubMed] [Google Scholar]
  • 4.DeAngelis LM. Brain tumors. N Engl J Med. 2001;344:114–23. doi: 10.1056/NEJM200101113440207. [DOI] [PubMed] [Google Scholar]
  • 5.Wooten EC, Fults D, Duggirala R, et al. A study of loss of heterozygosity at 70 loci in anaplastic astrocytoma and glioblastoma multiforme with implications for tumor evolution. Neuro Oncol. 1999;1:169–76. doi: 10.1093/neuonc/1.3.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Rasheed BK, McLendon RE, Friedman HS, et al. Chromosome 10 deletion mapping in human gliomas: a common deletion region in 10q25. Oncogene. 1995;10:2243–6. [PubMed] [Google Scholar]
  • 7.Watanabe K, Tachibana O, Sata K, Yonekawa Y, Kleihues P, Ohgaki H. Overexpression of the egf receptor and p53 mutations are mutually exclusive in the evolution of primary and secondary glioblastomas. Brain Pathol. 1996;6:217–24. doi: 10.1111/j.1750-3639.1996.tb00848.x. [DOI] [PubMed] [Google Scholar]
  • 8.Ohgaki H, Kleihues P. Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas. J Neuropathol Exp Neurol. 2005;64:479–89. doi: 10.1093/jnen/64.6.479. [DOI] [PubMed] [Google Scholar]
  • 9.Biernat W, Tohma Y, Yonekawa Y, Kleihues P, Ohgaki H. Alterations of cell cycle regulatory genes in primary (de novo) and secondary glioblastomas. Acta Neuropathol. 1997;94:303–9. doi: 10.1007/s004010050711. [DOI] [PubMed] [Google Scholar]
  • 10.Hermanson M, Funa K, Koopmann J, et al. Association of loss of heterozygosity on chromosome 17p with high platelet-derived growth factor alpha receptor expression in human malignant gliomas. Cancer Res. 1996;56:164–71. [PubMed] [Google Scholar]
  • 11.Esteller M, Garcia–Foncillas J, Andion E, et al. Inactivation of the dna-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 2000;343:1350–4. doi: 10.1056/NEJM200011093431901. [DOI] [PubMed] [Google Scholar]
  • 12.Esteller M, Gaidano G, Goodman SN, et al. Hypermethylation of the dna repair gene O6-methylguanine dna methyltransferase and survival of patients with diffuse large B-cell lymphoma. J Natl Cancer Inst. 2002;94:26–32. doi: 10.1093/jnci/94.1.26. [DOI] [PubMed] [Google Scholar]
  • 13.Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–96. doi: 10.1056/NEJMoa043330. [DOI] [PubMed] [Google Scholar]
  • 14.Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352:997–1003. doi: 10.1056/NEJMoa043331. [DOI] [PubMed] [Google Scholar]
  • 15.Mason WP, Del Maestro R, Eisenstat D, et al. Canadian recommendations for the treatment of glioblastoma multiforme. Curr Oncol. 2007;14:110–17. doi: 10.3747/co.2007.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Choucair AK, Levin VA, Gutin PH, et al. Development of multiple lesions during radiation therapy and chemotherapy in patients with gliomas. J Neurosurg. 1986;65:654–8. doi: 10.3171/jns.1986.65.5.0654. [DOI] [PubMed] [Google Scholar]
  • 17.Gaspar LE, Fisher BJ, Macdonald DR, et al. Supratentorial malignant glioma: patterns of recurrence and implications for external beam local treatment. Int J Radiat Oncol Biol Phys. 1992;24:55–7. doi: 10.1016/0360-3016(92)91021-E. [DOI] [PubMed] [Google Scholar]
  • 18.Lee SW, Fraass BA, Marsh LH, et al. Patterns of failure following high-dose 3-D conformal radiotherapy for high-grade astrocytomas: a quantitative dosimetric study. Int J Radiat Oncol Biol Phys. 1999;43:79–88. doi: 10.1016/S0360-3016(98)00266-1. [DOI] [PubMed] [Google Scholar]
  • 19.Somerfield M, Padberg J, Pfister D, et al. asco clinical practice guidelines: process, progress, pitfalls, and prospects. Classic Pap Curr Comments. 2000;4:881–6. [Google Scholar]
  • 20.Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010;28:1963–72. doi: 10.1200/JCO.2009.26.3541. [DOI] [PubMed] [Google Scholar]
  • 21.Macdonald DR, Cascino TL, Schold SC, Jr, Cairncross JG. Response criteria for phase ii studies of supratentorial malignant glioma. J Clin Oncol. 1990;8:1277–80. doi: 10.1200/JCO.1990.8.7.1277. [DOI] [PubMed] [Google Scholar]
  • 22.Roldán GB, Scott JN, McIntyre JB, et al. Population-based study of pseudoprogression after chemoradiotherapy in gbm. Can J Neurol Sci. 2009;36:617–22. doi: 10.1017/s0317167100008131. [DOI] [PubMed] [Google Scholar]
  • 23.Taal W, Brandsma D, de Bruin HG, et al. Incidence of early pseudo-progression in a cohort of malignant glioma patients treated with chemoirradiation with temozolomide. Cancer. 2008;113:405–10. doi: 10.1002/cncr.23562. [DOI] [PubMed] [Google Scholar]
  • 24.Sanghera P, Perry J, Sahgal A, et al. Pseudoprogression following chemoradiotherapy for glioblastoma multiforme. Can J Neurol Sci. 2010;37:36–42. doi: 10.1017/s0317167100009628. [DOI] [PubMed] [Google Scholar]
  • 25.Hoffman WF, Levin VA, Wilson CB. Evaluation of malignant glioma patients during the postirradiation period. J Neurosurg. 1979;50:624–8. doi: 10.3171/jns.1979.50.5.0624. [DOI] [PubMed] [Google Scholar]
  • 26.Brandes AA, Tosoni A, Spagnolli F, et al. Disease progression or pseudoprogression after concomitant radiochemotherapy treatment: pitfalls in neurooncology. Neuro Oncol. 2008;10:361–7. doi: 10.1215/15228517-2008-008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.DeAngelis LM, Delattre JY, Posner JB. Radiation-induced dementia in patients cured of brain metastases. Neurology. 1989;39:789–96. doi: 10.1212/wnl.39.6.789. [DOI] [PubMed] [Google Scholar]
  • 28.Brandes AA, Francheschi E, Tosoni A, et al. mgmt promoter methylation status can predict the incidence and outcome of pseudoprogression after concomitant radiochemotherapy in newly diagnosed glioblastoma patients. J Clin Oncol. 2008;26:2192–7. doi: 10.1200/JCO.2007.14.8163. [DOI] [PubMed] [Google Scholar]
  • 29.Wald LL, Nelson SJ, Day MR, et al. Serial proton magnetic resonance spectroscopy imaging of glioblastoma multiforme after brachytherapy. J Neurosurg. 1997;87:525–34. doi: 10.3171/jns.1997.87.4.0525. [DOI] [PubMed] [Google Scholar]
  • 30.Weybright P, Sundgren PC, Maly P, et al. Differentiation between brain tumor recurrence and radiation injury using mr spectroscopy. AJR Am J Roentgenol. 2005;185:1471–6. doi: 10.2214/AJR.04.0933. [DOI] [PubMed] [Google Scholar]
  • 31.Sundgren PC, Fan X, Weybright P, et al. Differentiation of recurrent brain tumor versus radiation injury using diffusion tensor imaging in patients with new contrast-enhancing lesions. Magn Reson Imaging. 2006;24:1131–42. doi: 10.1016/j.mri.2006.07.008. [DOI] [PubMed] [Google Scholar]
  • 32.Ammirati M, Galicich JH, Arbit E, Liao Y. Reoperation in the treatment of recurrent intracranial malignant gliomas. Neurosurgery. 1987;21:607–14. doi: 10.1227/00006123-198711000-00001. [DOI] [PubMed] [Google Scholar]
  • 33.Barker FG, 2nd, Chang SM, Gutin PH, et al. Survival and functional status after resection of recurrent glioblastoma multiforme. Neurosurgery. 1998;42:709–23. doi: 10.1097/00006123-199804000-00013. [DOI] [PubMed] [Google Scholar]
  • 34.Daneyemez M, Gezen F, Canakci Z, Kahraman S. Radical surgery and reoperation in supratentorial malignant glial tumors. Minim Invasive Neurosurg. 1998;41:209–13. doi: 10.1055/s-2008-1052044. [DOI] [PubMed] [Google Scholar]
  • 35.Pinsker M, Lumenta C. Experiences with reoperation on recurrent glioblastoma multiforme. Zentralbl Neurochir. 2001;62:43–7. doi: 10.1055/s-2002-19477. [DOI] [PubMed] [Google Scholar]
  • 36.Guyotat J, Signorelli F, Frappaz D, Madarassy G, Ricci AC, Bret P. Is reoperation for recurrence of glioblastoma justified? Oncol Rep. 2000;7:899–904. doi: 10.3892/or.7.4.899. [DOI] [PubMed] [Google Scholar]
  • 37.Mandl ES, Dirven CM, Buis DR, Postma TJ, Vandertop WP. Repeated surgery for glioblastoma multiforme: only in combination with other salvage therapy. Surg Neurol. 2008;69:506–9. doi: 10.1016/j.surneu.2007.03.043. [DOI] [PubMed] [Google Scholar]
  • 38.Brem H, Piantadosi S, Burger PC, et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer-Brain Tumor Treatment Group. Lancet. 1995;345:1008–12. doi: 10.1016/S0140-6736(95)90755-6. [DOI] [PubMed] [Google Scholar]
  • 39.Metellus P, Coulibaly B, Nanni I, et al. Prognostic impact of O6-methylguanine-dna methyltransferase silencing in patients with recurrent glioblastoma multiforme who undergo surgery and carmustine wafer implantation: a prospective patient cohort. Cancer. 2009;115:4783–94. doi: 10.1002/cncr.24546. [DOI] [PubMed] [Google Scholar]
  • 40.Nieder C, Grosu AL, Molls M. A comparison of treatment results for recurrent malignant gliomas. Cancer Treat Rev. 2000;26:397–409. doi: 10.1053/ctrv.2000.0191. [DOI] [PubMed] [Google Scholar]
  • 41.Subach BR, Witham TF, Kondziolka D, Lunsford LD, Bozik M, Schiff D. Morbidity and survival after 1,3-bis(2-chloroethyl)- 1-nitrosourea wafer implantation for recurrent glioblastoma: a retrospective case-matched cohort series. Neurosurgery. 1999;45:17–22. doi: 10.1097/00006123-199907000-00004. [DOI] [PubMed] [Google Scholar]
  • 42.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. doi: 10.1186/1471-2407-10-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shaw E, Scott C, Souhami L, 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;47:291–8. doi: 10.1016/S0360-3016(99)00507-6. [DOI] [PubMed] [Google Scholar]
  • 44.Combs SE, Widmer V, Thilmann C, Hof H, Debus J, Schulz–Ertner D. Stereotactic radiosurgery (srs): treatment option for recurrent glioblastoma multiforme (gbm) Cancer. 2005;104:2168–73. doi: 10.1002/cncr.21429. [DOI] [PubMed] [Google Scholar]
  • 45.Cho KH, Hall WA, Gerbi BJ, Higgins PD, McGuire WA, Clark HB. Single dose versus fractionated stereotactic radiotherapy for recurrent high-grade gliomas. Int J Radiat Oncol Biol Phys. 1999;45:1133–41. doi: 10.1016/S0360-3016(99)00336-3. [DOI] [PubMed] [Google Scholar]
  • 46.Hall WA, Djalilian HR, Sperduto PW, et al. Stereotactic radiosurgery for recurrent malignant gliomas. J Clin Oncol. 1995;13:1642–8. doi: 10.1200/JCO.1995.13.7.1642. [DOI] [PubMed] [Google Scholar]
  • 47.Kondziolka D, Flickinger JC, Bissonette DJ, Bozik M, Lunsford LD. Survival benefit of stereotactic radiosurgery for patients with malignant glial neoplasms. Neurosurgery. 1997;41:776–85. doi: 10.1097/00006123-199710000-00004. [DOI] [PubMed] [Google Scholar]
  • 48.Shrieve DC, Alexander E, 3rd, Wen PY, et al. Comparison of stereotactic radiosurgery and brachytherapy in the treatment of recurrent glioblastoma multiforme. Neurosurgery. 1995;36:275–82. doi: 10.1227/00006123-199502000-00006. [DOI] [PubMed] [Google Scholar]
  • 49.Hudes RS, Corn BW, Werner–Wasik M, et al. A phase i dose escalation study of hypofractionated stereotactic radiotherapy as salvage therapy for persistent or recurrent malignant glioma. Int J Radiat Oncol Biol Phys. 1999;43:293–8. doi: 10.1016/S0360-3016(98)00416-7. [DOI] [PubMed] [Google Scholar]
  • 50.Shepherd SF, Laing RW, Cosgrove VP, et al. Hypofractionated stereotactic radiotherapy in the management of recurrent glioma. Int J Radiat Oncol Biol Phys. 1997;37:393–8. doi: 10.1016/S0360-3016(96)00455-5. [DOI] [PubMed] [Google Scholar]
  • 51.Vordermark D, Kölbl O, Ruprecht K, Vince GH, Bratengeier K, Flentje M. Hypofractionated stereotactic re-irradiation: treatment option in recurrent malignant glioma. BMC Cancer. 2005;5:55. doi: 10.1186/1471-2407-5-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hermanto U, Frija EK, Lii MJ, Chang EL, Mahajan A, Woo SY. Intensity-modulated radiotherapy (imrt) and conventional three-dimensional conformal radiotherapy for high-grade gliomas: does imrt increase the integral dose to normal brain? Int J Radiat Oncol Biol Phys. 2007;67:1135–44. doi: 10.1016/j.ijrobp.2006.10.032. [DOI] [PubMed] [Google Scholar]
  • 53.MacDonald SM, Ahmad S, Kachris S, et al. Intensity modulated radiation therapy versus three-dimensional conformal radiation therapy for the treatment of high grade glioma: a dosimetric comparison. J Appl Clin Med Phys. 2007;8:47–60. doi: 10.1120/jacmp.v8i2.2423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Fuller CD, Choi M, Forthuber B, et al. Standard fractionation intensity modulated radiation therapy (imrt) of primary and recurrent glioblastoma multiforme. Radiat Oncol. 2007;2:26. doi: 10.1186/1748-717X-2-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Narayana A, Yamada J, Berry S, et al. Intensity-modulated radiotherapy in high-grade gliomas: clinical and dosimetric results. Int J Radiat Oncol Biol Phys. 2006;64:892–7. doi: 10.1016/j.ijrobp.2005.05.067. [DOI] [PubMed] [Google Scholar]
  • 56.Arslan M, Karadeniz AN, Aksu G, et al. Postoperative hypofractionated radiotherapy in glioblastoma multiforme. J BUON. 2006;11:39–42. [PubMed] [Google Scholar]
  • 57.Wong ET, Hess KR, Gleason MJ, et al. Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase ii clinical trials. J Clin Oncol. 1999;17:2572–8. doi: 10.1200/JCO.1999.17.8.2572. [DOI] [PubMed] [Google Scholar]
  • 58.Perry JR, Bélanger K, Mason WP, et al. Phase ii trial of continuous dose-intense temozolomide in recurrent malignant glioma: rescue study. J Clin Oncol. 2010;28:2051–7. doi: 10.1200/JCO.2009.26.5520. [DOI] [PubMed] [Google Scholar]
  • 59.Yung WK, Albright RE, Olson J, et al. A phase ii study of temozolomide vs. procarbazine in patients with glioblastoma multiforme at first relapse. Br J Cancer. 2000;83:588–93. doi: 10.1054/bjoc.2000.1316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Brandes AA, Ermani M, Basso U, et al. Temozolomide as a second-line systemic regimen in recurrent high-grade glioma: a phase ii study. Ann Oncol. 2001;12:255–7. doi: 10.1023/A:1008336732273. [DOI] [PubMed] [Google Scholar]
  • 61.Brandes AA, Ermani M, Basso U, et al. Temozolomide in patients with glioblastoma at second relapse after first line nitrosourea-procarbazine failure: a phase ii study. Oncology. 2002;63:38–41. doi: 10.1159/000065718. [DOI] [PubMed] [Google Scholar]
  • 62.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;4:39–43. doi: 10.1215/15228517-4-1-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Chan DT, Poon WS, Chan YL, Ng HK. Temozolomide in the treatment of recurrent malignant glioma in Chinese patients. Hong Kong Med J. 2005;11:452–6. [PubMed] [Google Scholar]
  • 64.Brandes AA, Tosoni A, Cavallo G, 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;95:1155–60. doi: 10.1038/sj.bjc.6603376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kong DS, Lee JI, Kim WS, et al. A pilot study of metronomic temozolomide treatment in patients with recurrent temozolomide- refractory glioblastoma. Oncol Rep. 2006;16:1117–21. [PubMed] [Google Scholar]
  • 66.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;37:897–906. doi: 10.1093/jjco/hym132. [DOI] [PubMed] [Google Scholar]
  • 67.Wick A, Felsberg J, Steinbach JP, et al. Efficacy and tolerability of temozolomide in an alternating weekly regimen in patients with recurrent glioma. J Clin Oncol. 2007;25:3357–61. doi: 10.1200/JCO.2007.10.7722. [DOI] [PubMed] [Google Scholar]
  • 68.Balmaceda C, Peereboom D, Pannullo S, et al. Multi-institutional phase ii study of temozolomide administered twice daily in the treatment of recurrent high-grade gliomas. Cancer. 2008;112:1139–46. doi: 10.1002/cncr.23167. [DOI] [PubMed] [Google Scholar]
  • 69.Kong DS, Lee JI, Kim JH, et al. Phase ii trial of low-dose continuous (metronomic) treatment of temozolomide for recurrent glioblastoma. Neuro Oncol. 2010;12:289–96. doi: 10.1093/neuonc/nop030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wick A, Pascher C, Wick W, et al. Rechallenge with temozolomide in patients with recurrent gliomas. J Neurol. 2009;256:734–41. doi: 10.1007/s00415-009-5006-9. [DOI] [PubMed] [Google Scholar]
  • 71.Brandes AA, Tosoni A, Franceschi E, et al. Recurrence pattern after temozolomide concomitant with and adjuvant to radiotherapy in newly diagnosed patients with glioblastoma: correlation with mgmt promoter methylation status. J Clin Oncol. 2009;27:1275–9. doi: 10.1200/JCO.2008.19.4969. [DOI] [PubMed] [Google Scholar]
  • 72.Brandes AA, Franceschi E, Tosoni A, et al. O6-Methylguanine dna-methyltransferase methylation status can change between first surgery for newly diagnosed glioblastoma and second surgery for recurrence: clinical implications. Neuro Oncol. 2010;12:283–8. doi: 10.1093/neuonc/nop050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Wick W, Stupp R, Beule AC, et al. on behalf of the European Organisation for Research and Treatment of Cancer and the National Cancer Institute of Canada Clinical Trials Group A novel tool to analyze mri recurrence patterns in glioblastoma. Neuro Oncol. 2008;10:1019–24. doi: 10.1215/15228517-2008-058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Quinn JA, Jiang SX, Reardon DA, et al. Phase ii trial of temozolomide plus O6-benzylguanine in adults with recurrent, temozolomide-resistant malignant glioma. J Clin Oncol. 2009;27:1262–7. doi: 10.1200/JCO.2008.18.8417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kerbel RS, Kamen BA. The anti-angiogenic basis of metronomic chemotherapy. Nat Rev Cancer. 2004;4:423–36. doi: 10.1038/nrc1369. [DOI] [PubMed] [Google Scholar]
  • 76.Bertolini F, Paul S, Mancuso P, et al. Maximum tolerable dose and low-dose metronomic chemotherapy have opposite effects on the mobilization and viability of circulating endothelial progenitor cells. Cancer Res. 2003;63:4342–6. [PubMed] [Google Scholar]
  • 77.Emmenegger U, Man S, Shaked Y, et al. A comparative analysis of low-dose metronomic cyclophosphamide reveals absent or low-grade toxicity on tissues highly sensitive to the toxic effects of maximum tolerated dose regimens. Cancer Res. 2004;64:3994–4000. doi: 10.1158/0008-5472.CAN-04-0580. [DOI] [PubMed] [Google Scholar]
  • 78.Bocci G, Francia G, Man S, Lawler J, Kerbel RS. Thrombospondin 1, a mediator of the antiangiogenic effects of low-dose metronomic chemotherapy. Proc Natl Acad Sci U S A. 2003;100:12917–22. doi: 10.1073/pnas.2135406100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kurzen H, Schmitt S, Näher H, Möhler T. Inhibition of angiogenesis by non-toxic doses of temozolomide. Anticancer Drugs. 2003;14:515–22. doi: 10.1097/00001813-200308000-00003. [DOI] [PubMed] [Google Scholar]
  • 80.Tuettenberg J, Grobholz R, Korn T, Wenz F, Erber R, Vajkoczy P. Continuous low-dose chemotherapy plus inhibition of cyclooxygenase- 2 as an antiangiogenic therapy of glioblastoma multiforme. J Cancer Res Clin Oncol. 2005;131:31–40. doi: 10.1007/s00432-004-0620-5. [DOI] [PubMed] [Google Scholar]
  • 81.Stockhammer F, Misch M, Koch A, et al. Continuous low-dose temozolomide and celecoxib in recurrent glioblastoma. J Neurooncol. 2010. [E-pub ahead of print] [DOI] [PubMed]
  • 82.Chamberlain MC, Johnston SK. Salvage therapy with single agent bevacizumab for recurrent glioblastoma. J Neurooncol. 2010;96:259–69. doi: 10.1007/s11060-009-9957-6. [DOI] [PubMed] [Google Scholar]
  • 83.Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009;27:4733–40. doi: 10.1200/JCO.2008.19.8721. [DOI] [PubMed] [Google Scholar]
  • 84.Kriesl TN, Kim L, Moore K, et al. Phase ii trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol. 2009;27:740–5. doi: 10.1200/JCO.2008.16.3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 2007;25:4722–9. doi: 10.1200/JCO.2007.12.2440. [DOI] [PubMed] [Google Scholar]
  • 86.Zuniga RM, Torcuator R, Jain R, 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;91:329–36. doi: 10.1007/s11060-008-9718-y. [DOI] [PubMed] [Google Scholar]
  • 87.Reardon DA, Desjardins A, Vredenburgh JJ, et al. Metronomic chemotherapy with daily, oral etoposide plus bevacizumab for recurrent malignant glioma: a phase ii study. Br J Cancer. 2009;101:1986–94. doi: 10.1038/sj.bjc.6605412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Batchelor TT, Duda DG, di Tomaso E, et al. Phase ii study of cediranib, an oral pan-vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma. J Clin Oncol. 2010;28:2817–23. doi: 10.1200/JCO.2009.26.3988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Fadul CE, Kingman LS, Meyer LP, et al. A phase ii study of thalidomide and irinotecan for treatment of glioblastoma multiforme. J Neurooncol. 2008;90:229–35. doi: 10.1007/s11060-008-9655-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Iwamoto FM, Lamborn KR, Robins HI, et al. Phase ii trial of pazopanib (GW786034), an oral multi-targeted angiogenesis inhibitor, for adults with recurrent glioblastoma (North American Brain Tumor Consortium Study 06-02) Neuro Oncol. 2010;12:855–61. doi: 10.1093/neuonc/noq025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Norden AD, Drappatz J, Muzikansky A, et al. An exploratory survival analysis of anti-angiogenic therapy for recurrent malignant glioma. J Neurooncol. 2009;92:149–55. doi: 10.1007/s11060-008-9745-8. [DOI] [PubMed] [Google Scholar]
  • 92.Verhoeff JJ, van Tellingen O, Claes A, et al. Concerns about anti-angiogenic treatment in patients with glioblastoma multiforme. BMC Cancer. 2009;9:444. doi: 10.1186/1471-2407-9-444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Lucio–Eterovic AK, Piao Y, de Groot JF. Mediators of glioblastoma resistance and invasion during antivascular endothelial growth factor therapy. Clin Cancer Res. 2009;15:4589–99. doi: 10.1158/1078-0432.CCR-09-0575. [DOI] [PubMed] [Google Scholar]
  • 94.Tuettenberg J, Grobholz R, Seiz M, et al. Recurrence pattern in glioblastoma multiforme patients treated with anti-angiogenic chemotherapy. J Cancer Res Clin Oncol. 2009;135:1239–44. doi: 10.1007/s00432-009-0565-9. [DOI] [PubMed] [Google Scholar]
  • 95.Iwamoto FM, Abrey LE, Beal K, et al. Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma. Neurology. 2009;73:1200–6. doi: 10.1212/WNL.0b013e3181bc0184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Groves MD, Puduvalli VK, Gilbert MR, 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;101:615–20. doi: 10.1038/sj.bjc.6605189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Boiardi A, Eoli M, Salmaggi A, et al. Systemic temozolomide combined with loco-regional mitoxantrone in treating recurrent glioblastoma. J Neurooncol. 2005;75:215–20. doi: 10.1007/s11060-005-3030-x. [DOI] [PubMed] [Google Scholar]
  • 98.Reardon DA, Quinn JA, Rich JN, et al. Phase i trial of irinotecan plus temozolomide in adults with recurrent malignant glioma. Cancer. 2005;104:1478–86. doi: 10.1002/cncr.21316. [DOI] [PubMed] [Google Scholar]
  • 99.Chua SL, Rosenthal MA, Wong SS, et al. Phase 2 study of temozolomide and Caelyx in patients with recurrent glioblastoma multiforme. Neuro Oncol. 2004;6:38–43. doi: 10.1215/S1152851703000188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Silvani A, Eoli M, Salmaggi A, et al. Phase ii trial of cisplatin plus temozolomide, in recurrent and progressive malignant glioma patients. J Neurooncol. 2004;66:203–8. doi: 10.1023/B:NEON.0000013479.64348.69. [DOI] [PubMed] [Google Scholar]
  • 101.Brandes AA, Basso U, Reni M, 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;22:1598–604. doi: 10.1200/JCO.2004.11.019. [DOI] [PubMed] [Google Scholar]
  • 102.Brandes AA, Tosoni A, Basso U, 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;22:4779–86. doi: 10.1200/JCO.2004.06.181. [DOI] [PubMed] [Google Scholar]
  • 103.Silvani A, Lamperti E, Gaviani P, et al. Salvage chemotherapy with procarbazine and fotemustine combination in the treatment of temozolomide treated recurrent glioblastoma patients. J Neurooncol. 2008;87:143–51. doi: 10.1007/s11060-007-9427-y. [DOI] [PubMed] [Google Scholar]
  • 104.Scoccianti S, Detti B, Sardaro A, et al. Second-line chemotherapy with fotemustine in temozolomide-pretreated patients with relapsing glioblastoma: a single institution experience. Anticancer Drugs. 2008;19:613–20. doi: 10.1097/CAD.0b013e3283005075. [DOI] [PubMed] [Google Scholar]
  • 105.Kappelle AC, Postma TJ, Taphoorn MJ, et al. pcv chemotherapy for recurrent glioblastoma multiforme. Neurology. 2001;56:118–20. doi: 10.1212/wnl.56.1.118. [DOI] [PubMed] [Google Scholar]
  • 106.Medical Research Council Brain Tumor Working Party Randomized trial of procarbazine, lomustine, and vincristine in the adjuvant treatment of high-grade astrocytoma: a Medical Research Council trial. J Clin Oncol. 2001;19:509–18. doi: 10.1200/JCO.2001.19.2.509. [DOI] [PubMed] [Google Scholar]
  • 107.Brandes AA, Tosoni A, Franceschi E, 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;64:769–75. doi: 10.1007/s00280-009-0926-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Fabrini MG, Silvano G, Lolli I, et al. A multi-institutional phase ii study on second-line fotemustine chemotherapy in recurrent glioblastoma. J Neurooncol. 2009;92:79–86. doi: 10.1007/s11060-008-9739-6. [DOI] [PubMed] [Google Scholar]
  • 109.Happold C, Roth P, Wick W, et al. acnu-based chemotherapy for recurrent glioma in the temozolomide era. J Neurooncol. 2009;92:45–8. doi: 10.1007/s11060-008-9728-9. [DOI] [PubMed] [Google Scholar]
  • 110.Chamberlain MC, Tsao–Wei DD. Salvage chemotherapy with cyclophosphamide for recurrent, temozolomide-refractory glioblastoma multiforme. Cancer. 2004;100:1213–20. doi: 10.1002/cncr.20072. [DOI] [PubMed] [Google Scholar]
  • 111.Toth J, Egervari K, Klekner A, et al. Analysis of EGFR gene amplification, protein over-expression and tyrosine kinase domain mutation in recurrent glioblastoma. Pathol Oncol Res. 2009;15:225–9. doi: 10.1007/s12253-008-9082-4. [DOI] [PubMed] [Google Scholar]
  • 112.van den Bent MJ, Brandes AA, Rampling R, 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;27:1268–74. doi: 10.1200/JCO.2008.17.5984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Raizer JJ, Abrey LE, Lassman AB, et al. on behalf of the North American Brain Tumor Consortium A phase ii trial of erlotinib in patients with recurrent malignant gliomas and nonprogressive glioblastoma multiforme postradiation therapy. Neuro Oncol. 2010;12:95–103. doi: 10.1093/neuonc/nop015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.de Groot JF, Gilbert MR, Aldape K, et al. Phase ii study of carboplatin and erlotinib (Tarceva, OSI-774) in patients with recurrent glioblastoma. J Neurooncol. 2008;90:89–97. doi: 10.1007/s11060-008-9637-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Reardon DA, Desjardins A, Vredenburgh JJ, et al. Phase 2 trial of erlotinib plus sirolimus in adults with recurrent glioblastoma. J Neurooncol. 2010;96:219–30. doi: 10.1007/s11060-009-9950-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Franceschi E, Cavallo G, Lonardi S, et al. Gefitinib in patients with progressive high-grade gliomas: a multicentre phase ii study by Gruppo Italiano Cooperativo di Neuro-Oncologia (gicno) Br J Cancer. 2007;96:1047–51. doi: 10.1038/sj.bjc.6603669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kreisl TN, Lassman AB, Mischel PS, et al. A pilot study of everolimus and gefitinib in the treatment of recurrent glioblastoma (gbm) J Neurooncol. 2009;92:99–105. doi: 10.1007/s11060-008-9741-z. [DOI] [PubMed] [Google Scholar]
  • 118.Thiessen B, Stewart C, Tsao M, et al. A phase i/ii trial of GW572016 (lapatinib) in recurrent glioblastoma multiforme: clinical outcomes, pharmacokinetics and molecular correlation. Cancer Chemother Pharmacol. 2009. [E-pub ahead of print] [DOI] [PubMed]
  • 119.Galanis E, Jaeckle KA, Maurer MJ, et al. Phase ii trial of vorinostat in recurrent glioblastoma multiforme: a North Central Cancer Treatment Group study. J Clin Oncol. 2009;27:2052–8. doi: 10.1200/JCO.2008.19.0694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wick W, Puduvalli VK, Chamberlain MC, et al. Phase iii study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J Clin Oncol. 2010;28:1168–74. doi: 10.1200/JCO.2009.23.2595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kreisl TN, Kotliarova S, Butman JA, et al. A phase i/ii trial of enzastaurin in patients with recurrent high-grade gliomas. Neuro Oncol. 2010;12:181–9. doi: 10.1093/neuonc/nop042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Reardon DA, Fink KL, Mikkelsen T, et al. Randomized phase ii study of cilengitidse, an integrin-targeting arginine-glycineaspartic acid peptide, in recurrent glioblastoma multiforme. J Clin Oncol. 2008;26:5610–17. doi: 10.1200/JCO.2008.16.7510. [DOI] [PubMed] [Google Scholar]
  • 123.Maurer GD, Tritschler I, Adams B, et al. Cilengitide modulates attachment and viability of human glioma cells, but not sensitivity to irradiation or temozolomide in vitro. Neuro Oncol. 2009;11:747–56. doi: 10.1215/15228517-2009-012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.National Comprehensive Cancer Network (nccn) NCCN Clinical Practice Guidelines in Oncology: Central Nervous System Cancers. Fort Washington, PA: NCCN; 2010. Ver. 1.2011. [Google Scholar]

Articles from Current Oncology are provided here courtesy of Multidisciplinary Digital Publishing Institute (MDPI)

RESOURCES