144
Views
4
CrossRef citations to date
0
Altmetric
Review

The impact of bevacizumab treatment on survival and quality of life in newly diagnosed glioblastoma patients

, , , , &
Pages 373-387 | Published online: 26 Sep 2014

Abstract

Glioblastoma multiforme (GBM) remains one of the most devastating tumors, and patients have a median survival of 15 months despite aggressive local and systemic therapy, including maximal surgical resection, radiation therapy, and concomitant and adjuvant temozolomide. The purpose of antineoplastic treatment is therefore to prolong life, with a maintenance or improvement of quality of life. GBM is a highly vascular tumor and overexpresses the vascular endothelial growth factor A, which promotes angiogenesis. Preclinical data have suggested that anti-angiogenic treatment efficiently inhibits tumor growth. Bevacizumab is a humanized monoclonal antibody against vascular endothelial growth factor A, and treatment has shown impressive response rates in recurrent GBM. In addition, it has been shown that response is correlated to prolonged survival and improved quality of life. Several investigations in newly diagnosed GBM patients have been performed during recent years to test the hypothesis that newly diagnosed GBM patients should be treated with standard multimodality treatment, in combination with bevacizumab, in order to prolong life and maintain or improve quality of life. The results of these studies along with relevant preclinical data will be described, and pitfalls in clinical and paraclinical endpoints will be discussed.

Introduction

Glioblastoma multiforme (GBM) remains one of the most lethal tumors, and newly diagnosed patients have a median survival of 15 months despite aggressive therapy including maximal surgical resection, radiation therapy (RT), and concomitant and adjuvant temozolomide.Citation1 GBM is characterized by excessive and aberrant angiogenesis, which is thought to be linked to expression of the prominent angiogenic promoter – the vascular endothelial growth factor A (VEGF).Citation2,Citation3 Bevacizumab (BVZ) (Avastin®; Genentech, Inc., South San Francisco, CA, USA), a humanized monoclonal antibody targeting VEGF, has been suggested as a feasible anti-angiogenic drug. BVZ inhibits angiogenesis by clearing circulating VEGF and thereby preventing interaction of VEGF with its target receptors on the surface of endothelial cells. Consequently, BVZ does not have to cross the blood–brain barrier (BBB) in order to be active. Preclinical and clinical studies suggest that anti-angiogenic therapies such as BVZ, in part elicit their anti-tumor activity by transiently “normalizing” the tumor vasculature by inhibiting the formation of new blood vessels, thereby improving the efficacy of both chemotherapy and RT.Citation4,Citation5

Several phase II studies in GBM patients with recurrent disease have shown impressive response rates varying from 30% to 50%, prolongation of life in responders, as well as improved quality of life (QOL).Citation6Citation10 These studies have led to a number of investigations in newly diagnosed GBM patients,Citation11Citation17 including well conducted prospective randomized phase III trialsCitation18Citation20 testing the hypothesis that GBM patients for first line therapy should be treated with standard multimodality treatment in combination with BVZ. In this paper we will discuss the results of these recent investigations of combining BVZ and standard treatment in newly diagnosed patients with primary GBM.

Anti-angiogenic therapy rationale

GBM tumor cells are known to produce VEGF primarily as a result of hypoxia, glucose deprivation, oxidative and mechanical stress, and mutations in both oncogenes and tumor suppressor genes.Citation2 VEGF binds to its receptors on endothelial cells, resulting in new blood vessel formation, which facilitates tumor growth. Therefore, 4 decades ago, Judah Folkman hypothesized that anti-angiogenic therapy by vessel pruning would be an effective treatment strategy in cancer.Citation21 Now, this hypothesis is widely accepted, and a variety of drugs targeting VEGF or its receptors have been shown to effectively prevent or reduce the growth of experimental mouse tumors and xenografts.Citation22

However, the promising results from preclinical animal studies in GBM have not been translated into the clinic, suggesting that the mechanisms of angiogenesis are complicated and that escape mechanisms during single anti-angiogenic drug treatment with drugs such as BVZ occur.Citation22Citation25 More recently, Rakesh K Jain formulated a new hypothesis which suggested that anti-angiogenic therapy elicits its anti-tumor activity by transiently normalizing the abnormal structure and function of tumor vasculature to make it more efficient for delivery of oxygen and chemotherapeutic agents.Citation26 Evidence from several preclinical studies has confirmed that the temporary vascular normalization as a result of anti-angiogenic therapy, improves tumor blood perfusion, which consequently increases the delivery of chemotherapy and oxygen, and this potentially enhances the efficacy of both chemotherapy and RT.Citation27Citation30 Recently, Batchelor et al provided clinical proof of concept that improved tumor blood perfusion, as a consequence of vascular normalization, was related to longer overall survival (OS) in newly diagnosed GBM patients treated with a combination of anti-angiogenic therapy (Cediranib) (Recentin™; AstraZeneca plc, London, UK) and standard treatment.Citation5 These data remain to be validated, but strongly suggest that part of the beneficial response mechanisms of anti-angiogenic therapy in GBM patients is due to vascular normalization.

In addition to the paracrine stimulation of endothelial cells, recent data indicate that the GBM cells themselves also possess VEGF receptors and that autocrine stimulation of these receptors promotes tumor growth (Michaelsen et al, unpublished data, 2014).Citation31 However, whether BVZ therapy has a directly disrupting effect on the autocrine growth stimulation of glioma cells in the perivascular niche, remains to be studied further.

BVZ in newly diagnosed GBM

Efficacy

Over the years, seven phase II (including two randomized)Citation11Citation17 and three phase III randomized studiesCitation18Citation20 with BVZ as part of the treatment of newly diagnosed GBM patients have been published. In three studies, BVZ was given as neoadjuvant treatment,Citation11,Citation12,Citation14 while it was given as concomitant and adjuvant treatment in combination with RT in the remaining studiesCitation13,Citation15Citation20 (). The response rates were comparable in both the neoadjuvant and adjuvant setting, and 23% to 38% responded to treatment, which is comparable to patients treated for recurrent disease.Citation32

Table 1 Efficacy of bevacizumab in newly diagnosed GBM

Progression free survival (PFS) at 6 months varied from 52% to 85%, and median PFS from 7.3 to 14.2 months. Finally, median OS varied from 11.7 to 23 months. The studiesCitation11Citation20 varied in baseline characteristics such as patient age, performance status, degree of surgical resection, steroid use, and epigenetic silencing of MGMT (O6-methylguanine-DNA methyltransferase), which have been shown previously to have prognostic value,Citation33,Citation34 and this may explain some of the observed variations in response, PFS, and OS.

Among the published studiesCitation11Citation20 were two randomized Phase III studies, AVAGlioCitation18 and RTOG 0825,Citation19 which compared standard treatment versus standard treatment plus BVZ. Both studies included newly diagnosed GBM patients in good performance status having MGMT methylated as well as non-methylated tumors. Known prognostic factors were comparable in the two studies except surgery, since more patients had debulking surgery in the RTOG 0825 study compared to the AVAGlio study. Both studies found significant increase in median PFS in the BVZ treatment arm compared to the placebo treatment arm (AVAGlio: 6.2 months to 10.6 months, ∼71% increase, and RTOG 0825: 7.3 months to 10.7 months, ∼47% increase). In contrast, no difference in OS was observed in either of the studies.

In both the AVAGlioCitation18 and the RTOG 0825Citation19 studies, the patients were allowed to cross over to BVZ-containing treatment at progression, and this occurred frequently. However, independently of whether BVZ was given as early treatment or as late treatment at recurrence, only a marginal, clinical difference in median OS was seen as compared to the OS observed following standard treatment, as originally published by Stupp et al.Citation1 This indicates that BVZ in general does not influence OS in patients with GBM. Whether subgroup analysis may define patients more sensitive to BVZ treatment still remains to be examined.

In contrast to comparable results for the effects of BVZ on PFS and OS, the AVAGlio and RTOG 0825 studies found contradicting results when evaluating QOL. The AVAGlio studyCitation18 showed that increased PFS in the BVZ arm was accompanied by a better QOL, while the opposite if any change in QOL was found in the RTOG 0825 study.Citation19

In addition to the above, a third Phase III randomized study, GLARIUS, has been published, in which standard treatment was compared to RT combined with concomitant and adjuvant BVZ, plus irinotecan.Citation20 The study included non-methylated MGMT GBMs only, who are patients with supposedly worse prognoses as compared to patients with methylated MGMT. The data showed increased median OS in both arms as compared to published data from non-methylated groups,Citation35 which indicates that some sort of selection other than MGMT methylation status might have occurred. Nevertheless, RT and concomitant and adjuvant BVZ plus irinotecan increased PFS, but not median OS, as compared to standard treatment. In addition, the QOL study did not show any difference.Citation20 In conclusion, three well conducted, randomized Phase III studiesCitation18Citation20 could not demonstrate any survival benefit of adding BVZ to standard treatment in patients with newly diagnosed GBM.

Safety of BVZ in the first line setting

The Phase II studiesCitation11Citation17 and the three randomized studiesCitation18Citation20 were comparable in terms of safety and toxicity according to the National Cancer Institute Common Terminology Criteria for Adverse Events (). In general, concomitant RT and TMZ with and without BVZ was well tolerated, with hematotoxicity being the most common toxicity in both treatment groups.Citation11Citation20 The rates of serious adverse events were higher in the BVZ group than in the control group in the three randomized studies.Citation18Citation20 Notably, it was observed in the randomized studies that grade >3 hypertension and thromboembolic events were significantly more frequent in patients treated with BVZ, as compared to standard treatment.Citation18Citation20 In addition, in some studies, gastrointestinal perforation, cerebral bleeding, fatigue, wound healing complications, and proteinuria appeared more frequently in patients treated with BVZ, as compared to patients not receiving the drug.Citation11,Citation18Citation20

Table 2 Toxicity of bevacizumab in newly diagnosed GBM

Recurrence pattern

The recurrence pattern is shown in . The data available are very heterogeneous and in general, the major studiesCitation18,Citation36Citation38 do not uniformly support the assumption that BVZ treatment induces migratory growth at time of recurrence, resulting in more diffuse tumors.Citation39,Citation40

Table 3 Patterns of recurrence in bevacizumab treated GBM patients

Discussion

Only a subset or approximately one third of patients with newly diagnosed or recurrent GBM respond to BVZ treatment, and some studies report a better QOL while others do not.Citation9,Citation18Citation20 Several factors could be responsible for this, including molecular, cellular, and phenotypic resistance; problems in evaluating clinical and paraclinical response; and nonstandard procedure of reporting QOL data. Some important issues will be discussed in the following sections.

Resistance to BVZ

Multiple mechanisms of resistance to anti-angiogenic therapy have been described, and modes of resistance to anti-angiogenic therapy can be classified into intrinsic resistance (where tumors fail to respond from the onset of treatment) and adaptive resistance (where tumors initially respond, and then progress whilst still on treatment).Citation23 The molecular, cellular, and phenotypic bases of these two modes of resistance are still not well understood, and no molecular features have been identified which in clinical practice can predict which patient should be treated with anti-angiogenic therapy.Citation41Citation43

Although increased blood flow might occur during a short normalization window,Citation5 several studies have described hypoxia due to vascular pruning as being a central hallmark in resistance to long-term, anti-angiogenic therapy.Citation24,Citation40,Citation44,Citation45 Among other mechanisms, hypoxia induces upregulation of alternative angiogenic growth factors such as fibroblast growth factor 2, delta-like ligand 4, stromal cell-derived factor 1, platelet-derived growth factor alpha, angiopoietins, and placental growth factor. These and other angiogenic factors interact between tumor cells, endothelial cells, and stromal cells (eg, inflammatory cells and pericytes) in a complex network in order to adapt to anti-angiogenic therapy. Some factors, such as fibroblast growth factor 2, directly interact with receptors on endothelial cells to stimulate angiogenesis while others, such as stromal cell-derived factor 1, indirectly stimulate angiogenesis by recruiting pro-angiogenic, bone marrow-derived cells (BMDCs).Citation23,Citation24,Citation46

Also, myeloid BMDCs may be involved in the resistant phenotype. Preclinical studies have shown an increase in the number of tumor-infiltrating, pro-angiogenic BMDCs during tumor progression while on anti-angiogenic therapy.Citation23,Citation25,Citation47 Myeloid BMDCs consist of a variety of cell subpopulations including tumor-associated macrophages, which appear to constitute the largest subpopulation in GBM patient tumors after progression on anti-angiogenic therapy.Citation47 Tumor-associated macrophages are believed to induce resistance by the release of potent pro-angiogenic factors, a variety of cytokines, growth factors, and vascular-modulating enzymes.Citation48 Similarly, other subpopulations of BMDCs have been described as being involved in mechanisms of resistance to anti-angiogenic therapy in GBM.Citation49Citation51 A common feature of most studies describing BMDCs in GBM is the use of xenograft models, which are characterized by the lack of a normal immune system,Citation25,Citation50Citation52 and this may have influenced the observations of these studies. Future studies on BMDCs and other immune cells from patient GBM tissue or preclinical models other than the xenograft model might provide new knowledge of the immune system’s role in resistance to anti-angiogenic therapy in GBM patients.

Several studies have reported tumor invasion as being an important resistance mechanism to BVZ.Citation24,Citation40,Citation44,Citation45 Among the theories for development of an invasive BVZ-resistant tumor phenotype is the involvement of the tyrosine kinase c-mesenchymal-epithelial transition factor (c-Met), which in both preclinical animal studies and in patient GBM tumors has been shown to be upregulated during progression on anti-angiogenic therapy.Citation25,Citation53,Citation54 c-Met seems to be involved in resistance to anti-angiogenic therapy by promoting tumor invasion and importantly, this resistance mechanism was blocked by inhibiting c-Met in combination with anti-angiogenic therapy.Citation53 Although the recurrence patterns from the clinical studies described in do not support increased migratory growth during anti-angiogenic therapy,Citation18,Citation36Citation38 these findings suggest that the combination of anti-angiogenic therapy and a c-Met inhibitor may produce a more efficacious therapeutic effect in GBM.

Moreover, the expression of VEGF receptor 2 on GBM cells has been suggested as a resistance mechanism to BVZ.Citation31 In some GBM cells, the receptor is believed to be activated in an autocrine manner by VEGF, and has been linked to promotion of cell proliferation and tumor growth, in addition to the formation of vessel-like structures from the tumor cells themselves, thereby making the tumors independent of the normal endothelial cell-derived vasculature.Citation31,Citation55,Citation56 As BVZ can only cross the BBB in areas where the BBB is disrupted, and BVZ has to diffuse against a high intratumoral pressure gradient, autocrine VEGF stimulation will probably not be targeted.Citation57 In addition, new data from our laboratory suggest that the receptors in GBM are not solely dependent on VEGF, but are additionally activated by the growth factor vascular endothelial growth factor C (Michaelsen et al, unpublished data, 2014), which also could explain resistance to BVZ.

Additionally, recent data presented at the American Society of Clinical Oncology (ASCO) indicate that different molecular subtypes of GBM may represent tumor phenotypes with different resistance profiles to BVZ. This study performed subgroup analysis on GBM tissue taken from the AVAGlio study.Citation58 A total of 99 patients (56 receiving placebo treatment and 43 BVZ treatment) were classified into four subtypes by gene expression analysis: proneural, mesenchymal, proliferative, and unclassified.Citation59,Citation60 In addition, IDH1 mutation status analysis was performed. It was demonstrated that patients with proneural tumors without IDH1 mutation had a significantly longer median OS in the BVZ treatment group compared to the control treatment group (17.1 and 12.1 months, respectively; hazard ratio 0.63, 95% confidence interval: 0.4–0.99; P=0.043). Although this potential predictor needs validation in an independent dataset, it could potentially be used for patient and therapy stratification in the future, and it provides more information on BVZ resistance and mode of action.

In conclusion, many mechanisms have been suggested as responsible for BVZ resistance. However, no clear molecular or adaptive data definitively explain why only some GBM patients respond to BVZ.

Dosing and scheduling

Different dosing and scheduling strategies of BVZ were used in the three randomized phase III trials (). In all three studies, BVZ was administered concomitant and adjuvant at a dose of 10 mg/kg every 2 weeks, starting at week 4 of RT in the RTOG 0825Citation19 and day 1 of RT in the AVAGlioCitation18 and GLARIUSCitation20 trials. The adjuvant treatment phase in the three studies varied in length, dosing, and scheduling. The dosing and scheduling were based on previously reported trials on recurrent GBM and other non-GBM malignancies.Citation32,Citation61 Although BVZ is most often used at a dose of 10 mg/kg administered every 2 weeks in recurrent GBM until toxicity or progression occurs, its optimal dose has not been established yet. A meta-analysis of BVZ for recurrent GBM in 548 patients from 15 phase II studies reported no difference in efficacy between BVZ at 5 mg/kg and higher doses (10 mg/kg every 2 weeks or 15 mg/kg every 3 weeks), and this inconsistent dose–response relationship has also been reported in other non-GBM malignancies.Citation61 Nevertheless, the efficacy and toxicity of these different doses warrants further investigation in prospective clinical trials.

The GLARIUSCitation20 and the AVAGlioCitation18 trials continued treatment until progression. Recent retrospective data on recurrent GBM have shown that BVZ discontinuation unrelated to disease progression does not appear to cause rebound recurrence or shorten PFS in patients who benefit from BVZ.Citation62 However, the optimal duration of BVZ treatment and the impact of discontinuation requires prospective evaluation in clinical trials.

Safety

Regardless of the potential benefits of BVZ in combination with standard therapy, BVZ is associated with higher risk of several potential toxicities when combined with standard radiotherapy. The most frequent toxicity was hypertension, and BVZ was also associated with an increased risk of arterial and venous thromboembolic events.Citation18Citation20 Other observed BVZ toxicities were gastrointestinal perforation, cerebral bleeding, fatigue, wound healing complications, and proteinuria.Citation11,Citation18Citation20 Evaluation for these toxicities is important in BVZ-treated patients, because early intervention may decrease morbidity and mortality risk if this treatment moves into first line treatment in the future. Additionally, treatment with BVZ can significantly alter a patient’s treatment planning, as it can require treatment cessation to avoid exacerbation of an adverse event. However, the data on the optimal toxicity management guidelines are currently limited, and are based on the experiences in recurrent GBM and a variety of non-GBM cancers.Citation63

Clinical endpoints of GBM treatment

When assessing clinical efficacy, different endpoints are used in various malignant diseases. Interpreting endpoints in GBM presents several hurdles, especially when treating with anti-angiogenic drugs. These problems are discussed in the following section.

Response evaluation

Tumor response is normally evaluated by the MacDonald Criteria and/or the Radiologic Assessment in Neuro-Oncology (RANO) criteria, which are based on a combination of tumor measurements, or estimates based on contrast-and non-enhancing tumor images and utilizing magnetic resonance imaging (MRI) scan techniques, including MRI T2-weighted and fluid-attenuated inversion recovery image sequences. In addition, corticosteroid dose and neurological status are evaluated.Citation64Citation66

Response evaluation has some inherent difficulties. Tumor enhancement can be non-specific, as it also partly represents peritumoral edema due to the leakiness of tumor vessels. Moreover, enhancement can be influenced by changes in corticosteroid dose and radiologic technique. In addition, increased enhancement can be caused by other processes that are not related to tumor progression such as treatment-related inflammation, seizure activity, post-surgical changes, ischemia, and radiation effects.Citation67Citation70

Pseudo-progression is an increase in contrast enhancement unrelated to tumor progression. Pseudo-progression is seen in 20%–30% of patients receiving RT and temozolomide, is a result of increased permeability of vasculature after RT, and can be accompanied by progressive neurological symptoms.Citation71Citation74

Other limitations include difficulty in measuring irregular-shaped tumors, inter-observer variability, lack of assessment of the non-enhancing tumor component, and limitations when assessing multifocal tumors and walls of cystic or surgical cavities.Citation75Citation77

Anti-angiogenic treatment raises new challenges, as MRI scans can sometimes show signs of response to such treatment due to normalization of tumor vasculature, not necessarily antitumor activity, and therefore represent a so-called pseudo-response. As a consequence, it is postulated that anti-angiogenic treatment might control contrast-enhancing tumors more effectively than non-enhancing tumors, which can cause problems when interpreting MRI scans.Citation24 As anti-angiogenic treatment seems to control tumor enhancement as early as 1–2 days after initiation of treatment,Citation32,Citation78 non-enhancing tumors may continue to grow and therefore, measuring only the enhancing lesion fails to correctly estimate tumor growth.Citation75,Citation76,Citation79

Difficulties assessing non-enhancing tumors using MRI can arise from the fact that increases in T2 and fluid-attenuated inversion recovery signal can arise from post-irradiation damage, decreased glucocorticoid dosage, postoperative changes, seizure activity, demyelinization, and ischemic injury, and can mistakenly be interpreted as tumor progression.Citation9

In recognition of the potential challenges with response assessment, the AVAGlio studyCitation18 used the modified response criteria,Citation80 while both the RTOG 0825Citation19 and the GLARIUSCitation20 studies used the MacDonald Criteria.Citation64 In these studies, there seems to be a strong concordance in determining response and progression by different “standardized criteria”,Citation81 but it cannot be excluded that variation in the estimate of PFS in the three randomized studies might be due to differences in interpretation of response.

Recently, baseline 18F-fluoro-D-glucose (FDG)-PET (positron emission tomography) imaging has been suggested as a feasible biomarker in recurrent GBM patients for predicting tumor control and survival following anti-angiogenic therapy with BVZ.Citation82 However, due to the high rate of glucose metabolism in normal brain tissue and in inflammatory cells, FDG has shown diagnostic limitations when used for brain tumor imaging.Citation83 In contrast, radiolabeled amino acids have a relatively low uptake in normal brain tissue and usually accumulate intensely in tumor cells. Preclinical and clinical studies have shown that 18F amino acid and nucleic analog PET tracers, eg, O-(2-18F-fluoroethyl)-L-tyrosine and [18F]-fluoro-L-thymidine, are superior to the FDG-PET in predicting treatment efficacy, and that predictability is enhanced by a combination of both PET and MRI.Citation84Citation86 Although these techniques still need to be validated, non-invasive PET tracers, in addition to MRI, may add information in response assessment, which could be useful for clinical decision making, eg, discontinuation of BVZ.

PFS and OS

When assessing the efficacy of drugs in clinical trials, PFS and OS have been the most commonly used primary endpoints.Citation1,Citation18Citation20,Citation32 OS is a straightforward endpoint that apparently offers a simple interpretation.Citation87 However, OS is likely to be influenced by the subsequent anticancer treatments the patients receive after going off-study for any reason (eg, due to disease progression or intolerable toxicities).

Treatment with BVZ in patients with GBM seems to increase median PFS, but not OS. However, evaluating PFS depends on accurate and objective means to detect progression. Further, it is influenced by pseudo-progression and pseudo-response (see the “Response evaluation” section), especially in patients receiving temozolomide in combination with radiation and anti-angiogenic treatment, and these problems may lead to inaccurate estimation of PFS.Citation88

QOL

Maintenance or increase of QOL is an important endpoint in the treatment of GBM. GBM is accompanied by neurological and cognitive impairments in almost all patients.Citation89 These impairments are related to the tumor and its position in the central nervous system, tumor-related epilepsy, and treatment related factors.Citation9,Citation89,Citation91 When treating GBM patients with targeted therapies such as anti-angiogenic agents, QOL should be taken into consideration, since treatment could delay deterioration of neurological and cognitive functions and thereby improve QOL. Although still controversial, especially in newly diagnosed GBMs, some evidence suggest that BVZ produces an improvement or preservation of neurocognitive function.Citation8

Headache, nausea, and vomiting are frequent symptoms seen in patients with GBM, which are due to increased intracranial pressure caused by peritumoral edema. Administering corticosteroids can diminish peritumoral edema, thereby alleviating the symptoms associated with increased intracranial pressure. On the other hand, corticosteroids may induce negative effects on neurocognitive function and/or QOL,Citation92Citation94 and it has been documented that corticosteroid use in primary brain tumors and metastases can lead to increases in blood glucose levels, peripheral edema, psychiatric disorders, and Cushing’s syndrome.Citation95,Citation96 However, the reduced intracranial pressure associated with corticosteroid treatment improves neurocognitive functions and QOL, despite other corticosteroid-induced adverse effects.

Studies, including the AVAGlio study, have shown that BVZ has a steroid-sparing effect, which may be a consequence of improved tumor control as well as alleviation of vasogenic brain edema. This would be expected to positively impact QOL due to lower corticosteroid dose and, therefore, less corticosteroid-related toxicity.Citation9,Citation18,Citation63,Citation81Citation85 Moreover, the steroid-sparing effect of BVZ is associated with clinical response and improved neurological symptoms in high-grade glioma, or GBM.Citation84,Citation86Citation88

It is still unclear whether BVZ given as part of primary treatment in patients with newly diagnosed GBM in fact influences QOL positively, and further evaluation of the AVAGlio,Citation18 RTOG 0825,Citation19 and the GLARIUSCitation20 studies is therefore needed.

Conclusion

BVZ given as part of primary treatment in newly diagnosed GBM patients is feasible and safe. It does prolong PFS, but not OS. It might influence QOL, but we still await critical evaluation of published data. No molecular or clinical data are available that can predict which subpopulation of patients might benefit from BVZ treatment.

Disclosure

The authors report no conflicts of interest in this work.

Reference

  • StuppRMasonWPvan den BentMJRadiotherapy plus concomitant and adjuvant temozolomide for glioblastomaN Engl J Med20053521098799615758009
  • HicklinDJEllisLMRole of the vascular endothelial growth factor pathway in tumor growth and angiogenesisJ Clin Oncol20052351011102715585754
  • PlateKHScholzADumontDJTumor angiogenesis and anti-angiogenic therapy in malignant gliomas revisitedActa Neuropathol2012124676377523143192
  • JainRKNormalization of tumor vasculature: an emerging concept in antiangiogenic therapyScience20053075706586215637262
  • BatchelorTTGerstnerEREmblemKEImproved tumor oxygenation and survival in glioblastoma patients who show increased blood perfusion after cediranib and chemoradiationProc Natl Acad Sci USA201311047190591906424190997
  • de GrootJFMandelJJUpdate on anti-angiogenic treatment for malignant gliomasCurr Oncol Rep201416438024510742
  • HanKRenMWickWProgression-free survival as a surrogate endpoint for overall survival in glioblastoma: a literature-based meta-analysis from 91 trialsNeuro Oncol201416569670624335699
  • HenrikssonRAsklundTPoulsenHSImpact of therapy on quality of life, neurocognitive function and their correlates in glioblastoma multiforme: a reviewJ Neurooncol2011104363964621468776
  • JakobsenJNHasselbalchBStockhausenMTLassenUPoulsenHSIrinotecan and bevacizumab in recurrent glioblastoma multiformeExpert Opin Pharmacother201112582583321385110
  • OlsonJJNayakLOrmondDRThe role of targeted therapies in the management of progressive glioblastoma: A systematic review and evidence-based clinical practice guidelineJ Neurooncol2014
  • ChauffertBFeuvretLBonnetainFRandomized phase II trial of irinotecan and bevacizumab as neo-adjuvant and adjuvant to temozolomide-based chemoradiation compared to temozolomide-chemoradiation for unresectable glioblastoma. Final results of the TEMAVIR study from ANOCEFAnn Oncol2014
  • HoflandKFHansenSSorensenMNeoadjuvant bevacizumab and irinotecan versus bevacizumab and temozolomide followed by concomitant chemoradiotherapy in newly diagnosed glioblastoma multiforme: A randomized phase II studyActa Oncol2014
  • LaiATranANghiemphuPLPhase II study of bevacizumab plus temozolomide during and after radiation therapy for patients with newly diagnosed glioblastoma multiformeJ Clin Oncol201129214214821135282
  • LouEPetersKBSumrallALPhase II trial of upfront bevacizumab and temozolomide for unresectable or multifocal glioblastomaCancer Med20132218519523634286
  • NarayanaAGruberDKunnakkatSA clinical trial of bevacizumab, temozolomide, and radiation for newly diagnosed glioblastomaJ Neurosurg2012116234134522035272
  • OmuroABealKGutinPPhase II study of bevacizumab, temozolomide, and hypofractionated stereotactic radiotherapy for newly diagnosed glioblastomaClin Cancer Res Epub882014
  • VredenburghJJDesjardinsAReardonDAThe addition of bevacizumab to standard radiation therapy and temozolomide followed by bevacizumab, temozolomide, and irinotecan for newly diagnosed glioblastomaClin Cancer Res201117124119412421531816
  • ChinotOLWickWMasonWBevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastomaN Engl J Med2014370870972224552318
  • GilbertMRDignamJJArmstrongTSA randomized trial of bevacizumab for newly diagnosed glioblastomaN Engl J Med2014370869970824552317
  • HerrlingerUSchäferNSteinbachJPSurvival and quality of life in the randomized, multicenter GLARIUS trial investigating bevacizumab/irinotecan versus standard temozolomide in newly diagnosed, MGMT-non-methylated glioblastoma patientsJ Clin Oncol5312014325s (suppl; abstr 2042)
  • FolkmanJTumor angiogenesis: therapeutic implicationsN Engl J Med197128521118211864938153
  • SathornsumeteeSRichJNAntiangiogenic therapy in malignant glioma: promise and challengeCurr Pharm Des200713353545355818220791
  • BergersGHanahanDModes of resistance to anti-angiogenic therapyNat Rev Cancer20088859260318650835
  • Lucio-EterovicAKPiaoYde GrootJFMediators of glioblastoma resistance and invasion during antivascular endothelial growth factor therapyClin Cancer Res200915144589459919567589
  • PiaoYLiangJHolmesLAcquired resistance to anti-VEGF therapy in glioblastoma is associated with a mesenchymal transitionClin Cancer Res201319164392440323804423
  • JainRKNormalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapyNat Med20017998798911533692
  • ChaeSSKamounWSFarrarCTAngiopoietin-2 interferes with anti-VEGFR2-induced vessel normalization and survival benefit in mice bearing gliomasClin Cancer Res201016143618362720501615
  • GoelSDudaDGXuLNormalization of the vasculature for treatment of cancer and other diseasesPhysiol Rev20119131071112121742796
  • McGeeMCHamnerJBWilliamsRFImproved intratumoral oxygenation through vascular normalization increases glioma sensitivity to ionizing radiationInt J Radiat Oncol Biol Phys20107651537154520338480
  • WinklerFKozinSVTongRTKinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, angiopoietin-1, and matrix metalloproteinasesCancer Cell20046655356315607960
  • HamerlikPLathiaJDRasmussenRAutocrine VEGF-VEGFR2-Neuropilin-1 signaling promotes glioma stem-like cell viability and tumor growthJ Exp Med2012209350752022393126
  • FriedmanHSPradosMDWenPYBevacizumab alone and in combination with irinotecan in recurrent glioblastomaJ Clin Oncol200927284733474019720927
  • MichaelsenSRChristensenIJGrunnetKClinical variables serve as prognostic factors in a model for survival from glioblastoma multiforme: an observational study of a cohort of consecutive non-selected patients from a single institutionBMC Cancer20131340224004722
  • HegiMEDiserensACGorliaTMGMT gene silencing and benefit from temozolomide in glioblastomaN Engl J Med200535210997100315758010
  • GilbertMRWangMAldapeKDDose-dense temozolomide for newly diagnosed glioblastoma: a randomized phase III clinical trialJ Clin Oncol201331324085409124101040
  • BlochOSafaeeMSunMZDisseminated progression of glioblastoma after treatment with bevacizumabClin Neurol Neurosurg201311591795180123706614
  • ChamberlainMCRadiographic patterns of relapse in glioblastomaJ Neurooncol2011101231932321052776
  • PopeWBXiaQPatonVEPatterns of progression in patients with recurrent glioblastoma treated with bevacizumabNeurology201176543243721282590
  • de GrootJFFullerGKumarAJTumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and miceNeuro Oncol201012323324220167811
  • KeunenOJohanssonMOudinAAnti-VEGF treatment reduces blood supply and increases tumor cell invasion in glioblastomaProc Natl Acad Sci USA201110893749375421321221
  • HasselbalchBEriksenJGBroholmHProspective evaluation of angiogenic, hypoxic and EGFR-related biomarkers in recurrent glioblastoma multiforme treated with cetuximab, bevacizumab and irinotecanAPMIS2010118858559420666740
  • LaffaireJDi StefanoALChinotOAn ANOCEF genomic and transcriptomic microarray study of the response to irinotecan and bevacizumab in recurrent glioblastomasBiomed Res Int2014201428281524804210
  • SathornsumeteeSCaoYMarcelloJETumor angiogenic and hypoxic profiles predict radiographic response and survival in malignant astrocytoma patients treated with bevacizumab and irinotecanJ Clin Oncol200826227127818182667
  • DeLayMJahangiriACarbonellWSMicroarray analysis verifies two distinct phenotypes of glioblastomas resistant to antiangiogenic therapyClin Cancer Res201218102930294222472177
  • Paez-RibesMAllenEHudockJAntiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasisCancer Cell200915322023119249680
  • JubbAMBrowningLCampoLExpression of vascular Notch ligands Delta-like 4 and Jagged-1 in glioblastomaHistopathology201260574074722296176
  • Lu-EmersonCSnuderlMKirkpatrickNDIncrease in tumor-associated macrophages after antiangiogenic therapy is associated with poor survival among patients with recurrent glioblastomaNeuro Oncol20131581079108723828240
  • MurdochCMuthanaMCoffeltSBLewisCEThe role of myeloid cells in the promotion of tumour angiogenesisNat Rev Cancer20088861863118633355
  • LuKVBergersGMechanisms of evasive resistance to anti-VEGF therapy in glioblastomaCNS Oncol201321496523750318
  • PiaoYLiangJHolmesLGlioblastoma resistance to anti-VEGF therapy is associated with myeloid cell infiltration, stem cell accumulation, and a mesenchymal phenotypeNeuro Oncol201214111379139222965162
  • ShojaeiFWuXZhongCBv8 regulates myeloid-cell-dependent tumour angiogenesisNature2007450717182583118064003
  • JanbazianLKaramchandaniJDasSMouse models of glioblastoma: lessons learned and questions to be answeredJ Neurooncol201411811824522719
  • JahangiriADeLMMillerLMGene expression profile identifies tyrosine kinase c-Met as a targetable mediator of antiangiogenic therapy resistanceClin Cancer Res20131971773178323307858
  • LuKVChangJPParachoniakCAVEGF inhibits tumor cell invasion and mesenchymal transition through a MET/VEGFR2 complexCancer Cell2012221213522789536
  • FrancesconeRScullySBentleyBGlioblastoma-derived tumor cells induce vasculogenic mimicry through Flk-1 protein activationJ Biol Chem201228729248212483122654102
  • YaoXPingYLiuYVascular endothelial growth factor receptor 2 (VEGFR-2) plays a key role in vasculogenic mimicry formation, neovascularization and tumor initiation by Glioma stem-like cellsPLoS One201383e5718823536763
  • LampsonLAMonoclonal antibodies in neuro-oncology: Getting past the blood-brain barrierMAbs20113215316021150307
  • PhillipsHSandmanTLiCCorrelation of molecular subtypes with survival in AVAglio (bevacizumab [Bv] and radiotherapy [RT] and temozolomide [T] for newly diagnosed glioblastoma [GB]).)J Clin Oncol2014325 (suppl; abstr 2001)
  • PhillipsHSKharbandaSChenRMolecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesisCancer Cell20069315717316530701
  • VerhaakRGHoadleyKAPurdomEIntegrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1Cancer Cell20101719811020129251
  • WongETGautamSMalchowCBevacizumab for recurrent glioblastoma multiforme: a meta-analysisJ Natl Compr Canc Netw20119440340721464145
  • AndersonMDHamzaMAHessKRPuduvalliVKImplications of bevacizumab discontinuation in adults with recurrent glioblastomaNeuro Oncol201416682382824596117
  • ArmstrongTSWenPYGilbertMRSchiffDManagement of treatment-associated toxicites of anti-angiogenic therapy in patients with brain tumorsNeuro Oncol201214101203121422307472
  • MacdonaldDRCascinoTLScholdSCJrCairncrossJGResponse criteria for phase II studies of supratentorial malignant gliomaJ Clin Oncol199087127712802358840
  • ReardonDAGalanisEDeGrootJFClinical trial end points for high-grade glioma: the evolving landscapeNeuro Oncol201113335336121310734
  • WenPYMacdonaldDRReardonDAUpdated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working groupJ Clin Oncol201028111963197220231676
  • FinnMABlumenthalDTSalzmanKLJensenRLTransient postictal MRI changes in patients with brain tumors may mimic disease progressionSurg Neurol200767324625017320628
  • HenegarMMMoranCJSilbergeldDLEarly postoperative magnetic resonance imaging following nonneoplastic cortical resectionJ Neurosurg19968421741798592218
  • KumarAJLeedsNEFullerGNMalignant gliomas: MR imaging spectrum of radiation therapy- and chemotherapy-induced necrosis of the brain after treatmentRadiology2000217237738411058631
  • UlmerSBragaTABarkerFGClinical and radiographic features of peritumoral infarction following resection of glioblastomaNeurology20066791668167017101902
  • BrandsmaDStalpersLTaalWSminiaPvan den BentMJClinical features, mechanisms, and management of pseudoprogression in malignant gliomasLancet Oncol20089545346118452856
  • ChamberlainMCGlantzMJChalmersLVanHASloanAEEarly necrosis following concurrent Temodar and radiotherapy in patients with glioblastomaJ Neurooncol2007821818316944309
  • de WitMCde BruinHGEijkenboomWSillevis SmittPAvan den BentMJImmediate post-radiotherapy changes in malignant glioma can mimic tumor progressionNeurology200463353553715304589
  • TaalWBrandsmaDde BruinHGIncidence of early pseudo-progression in a cohort of malignant glioma patients treated with chemoirradiation with temozolomideCancer2008113240541018484594
  • HensonJWUlmerSHarrisGJBrain tumor imaging in clinical trialsAJNR Am J Neuroradiol200829341942418272557
  • SorensenAGBatchelorTTWenPYZhangWTJainRKResponse criteria for gliomaNat Clin Pract Oncol200851163464418711427
  • van den BentMJVogelbaumMAWenPYMacdonaldDRChangSMEnd point assessment in gliomas: novel treatments limit usefulness of classical Macdonald’s CriteriaJ Clin Oncol200927182905290819451418
  • VredenburghJJDesjardinsAHerndonJEBevacizumab plus irinotecan in recurrent glioblastoma multiformeJ Clin Oncol200725304722472917947719
  • Hygino da CruzLCJrRodriguezIDominguesRCGasparettoELSorensenAGPseudoprogression and pseudoresponse: imaging challenges in the assessment of posttreatment gliomaAJNR Am J Neuroradiol201132111978198521393407
  • ChinotOLMacdonaldDRAbreyLEResponse assessment criteria for glioblastoma: practical adaptation and implementation in clinical trials of antiangiogenic therapyCurr Neurol Neurosci Rep201313534723529375
  • Gallego Perez-LarrayaJLahutteMPetrirenaGResponse assessment in recurrent glioblastoma treated with irinotecan-bevacizumab: comparative analysis of the Macdonald, RECIST, RANO, and RECIST + F criteriaNeuro Oncol201214566767322492961
  • ColavolpeCChinotOMetellusPFDG-PET predicts survival in recurrent high-grade gliomas treated with bevacizumab and irinotecanNeuro Oncol201214564965722379188
  • OliveroWCDulebohnSCListerJRThe use of PET in evaluating patients with primary brain tumours: is it useful?J Neurol Neurosurg Psychiatry19955822502527876865
  • Corroyer-DulmontAPeresEAPetitEDetection of glioblastoma response to temozolomide combined with bevacizumab based on muMRI and muPET imaging reveals [18F]-fluoro-L-thymidine as an early and robust predictive marker for treatment efficacyNeuro Oncol2013151415623115160
  • NedergaardMKKristoffersenKMichaelsenSRThe Use of Longitudinal 18F-FET MicroPET Imaging to Evaluate Response to Irinotecan in Orthotopic Human Glioblastoma Multiforme XenograftsPLoS One201492e10000924918622
  • GalldiksNRappMStoffelsGResponse assessment of bevacizumab in patients with recurrent malignant glioma using [18F] Fluoroethyl-L-tyrosine PET in comparison to MRIEur J Nucl Med Mol Imaging2013401223323053325
  • BrandesAAFranceschiEGorliaTAppropriate end-points for right results in the age of antiangiogenic agents: future options for phase II trials in patients with recurrent glioblastomaEur J Cancer201248689690322119352
  • NordenADDrappatzJMuzikanskyAAn exploratory survival analysis of anti-angiogenic therapy for recurrent malignant gliomaJ Neurooncol200992214915519043778
  • TaphoornMJKleinMEvaluation of cognitive functions and quality of lifeHandb Clin Neurol201210417318322230444
  • BosmaIVosMJHeimansJJThe course of neurocognitive functioning in high-grade glioma patientsNeuro Oncol200791536217018697
  • MeyersCAScheibelRSEarly detection and diagnosis of neurobehavioral disorders associated with cancer and its treatmentOncology (Williston Park)1990471151221697183
  • de VriesMAvan LitsenburgRRHuismanJEffect of dexamethasone on quality of life in children with acute lymphoblastic leukaemia: a prospective observational studyHealth Qual Life Outcomes2008610319036151
  • LupienSJGillinCJHaugerRLWorking memory is more sensitive than declarative memory to the acute effects of corticosteroids: a dose-response study in humansBehav Neurosci1999113342043010443770
  • YoungAHSahakianBJRobbinsTWCowenPJThe effects of chronic administration of hydrocortisone on cognitive function in normal male volunteersPsychopharmacology (Berl)1999145326026610494574
  • HempenCWeissEHessCFDexamethasone treatment in patients with brain metastases and primary brain tumors: do the benefits outweigh the side-effects?Support Care Cancer200210432232812029432
  • SturdzaAMillarBABanaNThe use and toxicity of steroids in the management of patients with brain metastasesSupport Care Cancer20081691041104818256860
  • BoksteinFShpigelSBlumenthalDTTreatment with bevacizumab and irinotecan for recurrent high-grade glial tumorsCancer2008112102267227318327820
  • DesjardinsAReardonDAHerndonJEBevacizumab plus irinotecan in recurrent WHO grade 3 malignant gliomasClin Cancer Res200814217068707318981004
  • GuiuSTaillibertSChinotOBevacizumab/irinotecan. An active treatment for recurrent high grade gliomas: preliminary results of an ANOCEF Multicenter StudyRev Neurol (Paris)20081646–758859418565358
  • NghiemphuPLLiuWLeeYBevacizumab and chemotherapy for recurrent glioblastoma: a single-institution experienceNeurology200972141217122219349600
  • NordenADYoungGSSetayeshKBevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrenceNeurology2008701077978718316689
  • BurgerPCDuboisPJScholdSCJrComputerized tomographic and pathologic studies of the untreated, quiescent, and recurrent glioblastoma multiformeJ Neurosurg19835821591696294260
  • ChangELAkyurekSAvalosTEvaluation of peritumoral edema in the delineation of radiotherapy clinical target volumes for glioblastomaInt J Radiat Oncol Biol Phys200768114415017306935
  • HochbergFHPruittAAssumptions in the radiotherapy of glioblastomaNeurology19803099079116252514
  • LevinVACraftsDCNormanDMCriteria for evaluating patients undergoing chemotherapy for malignant brain tumorsJ Neurosurg1977473329335894339
  • IwamotoFMAbreyLEBealKPatterns of relapse and prognosis after bevacizumab failure in recurrent glioblastomaNeurology200973151200120619822869
  • NarayanaAKellyPGolfinosJAntiangiogenic therapy using bevacizumab in recurrent high-grade glioma: impact on local control and patient survivalJ Neurosurg2009110117318018834263
  • WickADornerNSchaferNBevacizumab does not increase the risk of remote relapse in malignant gliomaAnn Neurol201169358659221446027
  • SoffiettiRTrevisanEBerteroLBevacizumab and fotemustine for recurrent glioblastoma: a phase II study of AINO (Italian Association of Neuro-Oncology)J Neurooncol2014116353354124293233
  • DesjardinsAReardonDACoanABevacizumab and daily temozolomide for recurrent glioblastomaCancer201211851302131221792866