Efficacy and safety of bevacizumab for the treatment of glioblastoma (Review)

  • Authors:
    • Zhiyun Yu
    • Gang Zhao
    • Zhonghua Zhang
    • Yunqian Li
    • Yong Chen
    • Nan Wang
    • Zhongying Zhao
    • Guifang Xie
  • View Affiliations

  • Published online on: December 16, 2015     https://doi.org/10.3892/etm.2015.2947
  • Pages: 371-380
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glioblastoma (GBM) is the most common and devastating primary malignant intracranial tumor in adults. The current first‑line treatment for patients with newly diagnosed GBM is surgical resection followed by radiotherapy plus concomitant and adjuvant temozolomide. This treatment protocol may prolong the survival period of the patient, however it is not curative and more effective therapeutic strategies are required. GBM is a type of highly vascularized tumor with increased expression levels of vascular endothelial growth factor (VEGF), which is a significant mediator of angiogenesis. Since angiogenesis is essential for tumor growth, anti‑angiogenic therapies hold potential for the treatment of GBM, and targeting VEGF has demonstrated promising results in previous studies. Bevacizumab (BEV) is a recombinant humanized monoclonal antibody that inhibits VEGF and is approved by the US Food and Drug Administration as a monotherapy treatment for patients with recurrent GBM and is associated with manageable toxicity. Previous studies have demonstrated that BEV may be an effective treatment for recurrent GBM, with prolonged progression‑free survival and overall survival, and maintained patient quality of life and functional status. The present review article briefly outlines the mechanism of action of BEV and summarizes the current literature and clinical trial research on the role of BEV for the treatment of patients with recurrent and newly diagnosed GBM.

Introduction

Glioblastoma (GBM) is the most common type of primary intracranial tumors, with an annual incidence of 5/100,000 individuals (1,2). The current standard treatment for patients with newly diagnosed GBM is surgery followed by external beam radiation and concomitant temozolomide (TMZ) chemotherapy and an additional 6 cycles of TMZ administration (3). In spite of advances in diagnosis and therapy, the prognosis is still relatively poor with a median overall survival (OS) of 14.6 months and a 5-year survival rate of 9.8% following diagnosis (4). The majority of patients with GBM experience recurrent disease, with a median time to recurrence of 7 months (5). The prognosis of recurrent GBM is severe with a median progression-free survival (PFS) and OS of 2.5 and 7.5 months (6), respectively. Therefore, there is an urgent need for more effective therapeutic strategies for the treatment of patients with GBM.

Angiogenesis is the formation of new blood vessels from existing vasculature, characterized by endothelial cell migration and proliferation. This normal physiological response occurs in wound healing and following hypoxic exposure; however, for tumor cells in an increased proliferative state, new vasculature is also required to access oxygen and facilitate metastasis (7,8). The angiogenic switch is mediated by various pro-angiogenic factors, predominantly vascular endothelial growth factor (VEGF), which are released by tumor, stromal and endothelial cells, resulting in vessel growth and tumor expansion (9,10). Previous preclinical studies investigating the use of bevacizumab (BEV) in GBM models have detected normalization of mature blood vessels, microvascular regression, and the inhibition of new blood vessels being formed in tumors (11). A previous study demonstrated that BEV is capable of inhibiting the action of VEGF on its receptor, preventing the proliferation and migration of endothelial cells, which in turn downregulates tumor vascularization and results in tumor cell hypoxia and death (12).

GBM is one of the most vascularized human tumors (1), which highly expresses VEGF, a significant mediator of angiogenesis (13). BEV is a humanized immunoglobulin (Ig) G1 monoclonal antibody that targets VEGF, which was approved by the US Food and Drug Administration (FDA) as a single agent for the treatment of recurrent GBM (14). However the European Medicines Agency (EMA) rejected this instruction due to a lack of evidence. Largely for this reason, BEV is currently used as the standard treatment for recurrent GBM in the United States, but not in Europe; although, in many countries BEV is administered for off-label use as a monotherapy or in combination with irinotecan (CPT-11), which is a topoisomerase I inhibitor. Previous studies, including the AVAglio (15) and Radiation Therapy Oncology Group (RTOG) 0825 (16) double-blinded, placebo-controlled phase III studies investigated BEV as an addition to the standard treatment of radiotherapy (RT) plus concomitant and adjuvant TMZ in patients with newly diagnosed GBM. OS did not reach significance in both trials; however PFS favored BEV administration in both.

In the present review, the mechanism of action of BEV is briefly introduced, with the focus on providing an overview and evaluation of the efficacy and safety of BEV as a monotherapy or in combination with cytotoxic chemotherapy and/or RT for the treatment of patients with recurrent and newly diagnosed GBM.

Mechanism of action and pharmacokinetics

BEV is a recombinant humanized monoclonal IgG1 antibody with a molecular weight of 149 kDa. BEV is capable of binding to and neutralizing the biological effects of VEGF, which is an important regulator of pathologic and physiologic angiogenesis (13). VEGF binds to and activates its target receptors, VEGF receptor (R)-1 and VEFGR-2, leading to their tyrosine phosphorylation and a subsequent signal transduction cascade, which activates vascular endothelial cells, pro-survival activity and elicits mitogenic signals to promote angiogenesis (17). BEV reduces tumor angiogenesis by blocking the biological activity of VEGF, thereby preventing vasogenic brain edema and tumor growth.

In the majority of previous clinical trials, BEV administration was characterized by a limited central compartment volume (Vc), low clearance (CL), and a long elimination half-life. The Vc and CL correspond to an initial half-life of 1.4 days and a terminal half-life of ~20 days (18). In another previous study, the mean steady-state volume of distribution for BEV ranged from 50–60 ml/kg (dose range, 0.1–10 mg/kg), whereas the steady-state volume did not alter with increasing dosages of BEV (19). The majority of previous clinical trials administered 5–15 mg/kg BEV every 2–4 weeks; therefore, whether higher doses of BEV produce faster clinical efficacy than low doses remains unknown. Wong et al (20) performed a meta-analysis regarding the treatment of recurrent GBM with BEV, concluding that there was no dose-response effect.

Efficacy of bevacizumab treatment for patients with recurrent glioblastoma

BEV was initially assessed in recurrent and previously treated GBM in combination with CPT-11, which is a topoisomerase I inhibitor; the response rate (RR) for the patients with GBM in an initial retrospective study was 43% (21), promoting the investigation of BEV with CPT-11 in subsequent clinical trials. The first completed, prospectively designed, single agent, phase II trial of BEV and CPT-11 for recurrent GBM was conducted by Vredenburgh et al (22,23). The patients were divided into two groups, an initial group 23) which were treated with 10 mg/kg BEV plus CPT-11 every 2 weeks, and a second group of patients 12) which were administered 15 mg/kg BEV every 21 days and CPT-11 on days 1, 8, 22 and 29. In both groups, 340–350 mg/m2 CPT-11 was administered to patients receiving enzyme-inducing anti-epileptic drug (EIAED) and 125 mg/m2 was administered to patients who were not receiving EIAEDs. The initial cohort demonstrated a RR of 63% in 23 patients with a median OS and median PFS of 9.2 and 5.3 months, respectively (22). Among all 35 patients included in the study, the PFS rate at 6 months (PFS-6) was 46% (95% CI, 32–66%), the 6-month OS rate (OS-6) was 77% (95% CI, 64–92%); whereas the median PFS and median OS were 24 weeks (95% CI, 18–36 weeks) and 42 weeks (95% CI, 35–60 weeks), respectively (23). In addition, 57% of the patients (95% CI, 39–74%) demonstrated at least a partial response (PR), and the 4-year survival rate was demonstrated to be 11% in this trial (24).

Friedman et al (25) conducted a randomized noncomparative phase II clinical trial of BEV with and without CPT-11 in 167 patients with recurrent GBM. Patients were randomized into groups of either 10 mg/kg BEV monotherapy every 2 weeks 85) or BEV in combination with CPT-11 82). The CPT-11 dose was based on the patient's anticonvulsant intake; patients taking EIAED received 340 mg/m2 and patients not taking EIAED received 125 mg/m2. Patients treated with BEV monotherapy demonstrated a RR of 28.2% (31/82 patients; 97.5% CI, 18.5–40.3%), whereas the PFS-6 was 42.6% (97.5% CI, 29.6–55.5%), and the median PFS and median OS were 4.2 months (95% CI, 2.9–5.8 months) and 9.2 months (95% CI, 8.2–10.7 months), respectively. Combination therapy of BEV and CPT-11 (25,26) demonstrated a RR of 37.8% (31/85 patients; 97.5% CI, 26.5–50.8%), whereas the PFS-6 was 50.3% (97.5% CI, 36.8–63.9%) and the median PFS and median OS were 5.6 months (95% CI, 4.4–6.2 months) and 8.7 months (95% CI, 7.8–10.9 months), respectively.

Kreisl et al (27) conducted another single-institution prospective study of BEV involving 48 patients with recurrent GBM. All patients were administered 10 mg/kg BEV monotherapy every 2 weeks until disease progression was detected. Following disease recurrence, patients were treated with BEV in combination with 340 or 125 mg/m2 CPT-11 every 2 weeks, depending on EIAED use. The RR for BEV monotherapy was 35% [1 complete response (CR); 16 PRs], whereas the PFS-6 was 29% (95% CI, 18–48%) and the OS-6 was 57%. The median PFS and median OS were 16 weeks (95% CI, 12–26 weeks) and 31 weeks (95% CI, 21–54 weeks), respectively (27).

Based on the findings of these two clinical studies (25,27), in May 2009 the FDA granted accelerated approval of single agent BEV for the treatment of patients with GBM (28).

Retrospective analyses of data from additional studies supplied more evidence for the efficacy of BEV monotherapy or combination therapy with cytotoxic or targeted agents in patients with recurrent GBM. Previous studies evaluating the efficacy of BEV monotherapy for patients with recurrent GBM have demonstrated objective RR (PR plus CR), overall survival, PFS and PFS-6 rates of 28.2–43%, 7.2–12 months, 1.0–4.2 months and 20.9–42.6%, respectively, as calculated from statistical treatment of the data (2527,2933). Furthermore, BEV plus chemotherapy combination therapy increased RR and PFS (25,27), and additional studies of BEV in combination with cytotoxic agents including carboplatin, erlotinib, etoposide, fotemustine, CPT-11 and dose-intense daily TMZ for patients with recurrent GBM also demonstrated RR, OS, PFS and PFS-6 rates of 20–67.6%, 4.3–11.5 months, 2.5–7.6 months and 25–63.7%, respectively (Table I) (17).

Table I.

Efficacy of bevacizumab alone or in combination with cytotoxic agents in recurrent glioblastoma.

Table I.

Efficacy of bevacizumab alone or in combination with cytotoxic agents in recurrent glioblastoma.

Response rate (%)
PFS
OS
Author, yearTreatment regimenPatients (n)CRPRMedian (months)PFS-6 (%)Median (months)OS-6 (%)(Ref.)
Stark-Vance et al, 2005BEV + CPT-1111  538N/A30  9N/A(21)
Vredenburgh et al, 2007BEV + CPT-1123   4.356.55.338   9.2372(22)
Vredenburgh et al, 2007BEV + CPT-113557N/A5.546   9.777(23)
Friedman et al, 2009BEV8528.2N/A4.242.6   9.2N/A(25)
Cloughesy et al, 2010BEV + CPT-118237.8N/A5.650.3   8.7N/A(26)
Kreisl et al, 2009BEV and BEV + CPT-114835*N/A3.729   7.257(27)
Chamberlain et al, 2010BEV5042N/A142   8.5N/A(29)
Raizer et al, 2010BEV50N/AN/A2.725   6.554(30)
Kreisl et al, 2011BEV30N/A43   2.9320.912N/A(31)
Nagane et al, 2012BEV2927.6N/A3.333.910.5N/A(32)
Hofer et al, 2011BEV176N/AN/AN/AN/A   8.3N/A(33)
Norden et al, 2008BEV + CT3334N/A442   8.265(41)
Gilbert et al, 2011BEV + CPT-115726.3N/AN/A37N/AN/A(42)
Gil et al, 2012BEV + CPT-119256N/A5.142   8.866(43)
Zuniga et al, 2009BEV + CPT-1137   5.462.27.663.711.578(44)
Quant et al, 2009BEV + CT35023N/AN/AN/AN/A(45)
Poulsen et al, 2009BEV + CPT-115230N/A540   6.9N/A(50)
Raval et al, 2007BEV + CPT-112047.3N/A4.225755(51)
Bokstein et al, 2008BEV + CPT-111910.536.84.225   7.055(52)
Taillibert et al, 2010BEV + CPT-11224N/AN/A4.839.4   8.3N/A(53)
Keyrouz et al, 2010BEV + CPT-113063N/A533.4   8.7N/A(54)
Pope et al, 2006BEV + CPT-11 or etoposide14050N/AN/AN/AN/A(55)
Nghiemphu et al, 2009BEV + CT44N/AN/A   4.2541  9N/A(56)
Desjardins et al, 2012BEV + TMZ3228N/A3.719   8.5N/A(57)
Verhoeff et al, 2010BEV + TMZ1520N/A2.517   4.3N/A(58)
Soffietti et al, 2011BEV + Fotemustine5448.1N/A5.244   9.1N/A(59)
Trevisan et al, 2010BEV + Fotemustine5943.4N/AN/A32   6.7N/A(60)
Hasselbach et al, 2010BEV + Cetuximab3234N/A3.830   7.2N/A(61)
Shapiro et al, 2013BEV + HFSRT24N/AN/A7.5N/A12.2N/A(62)
Sathornsumetee et al, 2010BEV + Erlotinib2448N/A4.22810N/A(63)
Niyazi et al, 2012BEV + RT20N/AN/A8N/A12.1N/A(64)
Hundsberger et al, 2013BEV + RT10N/AN/A5.7N/A   8.4N/A(65)
Gutin et al, 2009BEV + RT2050N/A7.36012.5N/A(66)

[i] *Macdonald criteria. PFS, progression-free survival; OS, overall survival; CR, complete response; PR, partial response; PFS-6, 6-month PFS rate; OS-6, 6-month OS rate; BEV, bevacizumab; CPT-11, irinotecan; N/A, not available; CT, chemotherapy; HFSRT, hypofractionated stereotactic radiotherapy; RT, radiotherapy.

Zhang et al (34) performed a meta-analysis to evaluate the efficacy and safety of BEV monotherapy 183) compared with BEV plus CPT-11 297) for the treatment of recurrent GBM. In the BEV group, the mean objective RR was 33.9% (95% CI, 18.1–52.1%), the PFS-6 was 38.8% (95% CI, 18.8–57.0%) and the median OS was 8.63 months (95% CI, 8.54–8.72 months). In the combined group, the mean objective RR was 45.8% (95% CI, 28.2–66.7%), the PFS-6 was 48.3% (95% CI, 25.4–54.3%) and the median OS was 8.91 months (95% CI, 8.69–9.13 months). The rates of discontinuation of treatment were 5.5 and 20.0%, respectively. Zhang et al concluded that patients in the BEV plus CPT-11 group demonstrated increased PFS-6 (P=0.046), objective RR (P=0.013) and discontinuation rates (P<0.001), as compared with the BEV monotherapy group. No significant difference in OS was detected between the groups (P=0.487) (34).

However, the EMA rejected the use of BEV for the treatment of patients with recurrent GBM, with one of the reasons being a lack of positive benefit-risk for BEV. Furthermore, the EMA did not consider the differences in objective RR to be noteworthy and concluded that the validity of this parameter as a surrogate endpoint for clinical benefit had not been established. In addition, the results were presented in terms of OS and PFS, which were difficult to interpret due to the lack of a randomized concurrent control (35). Furthermore, the use of contrast-enhanced magnetic resonance imaging may overestimate the RR (17); therefore, RR and PFS may not be optimal surrogate endpoints for anti-angiogenic treatment. Anti-VEGF treatment can reduce vascular permeability (17), which may also account for the radiographic improvement; however, this may not necessarily reflect tumor cell death. Therefore, the clinical relevance of these findings in predicting OS in patients with GBM following BEV monotherapy remains uncertain.

Efficacy of bevacizumab treatment for patients with newly diagnosed glioblastoma

With potential synergistic activity demonstrated by BEV in the treatment of recurrent GBM, BEV administration may also benefit patients with newly diagnosed GBM. The first phase II study investigating this was performed by Lai et al (36), who conducted an open-label, prospective, multicenter single-arm phase II study of BEV in combination with the standard treatment of RT plus concomitant and adjuvant TMZ in 70 patients with newly diagnosed GBM. All patients were treated with intravenous 10 mg/kg BEV every 2 weeks and 75 mg/m2 TMZ was administered orally daily during standard RT (2.0 Gy fractions totaling 60.0 Gy). Following completion of RT, patients were placed on a maintenance phase of TMZ (150–200 mg/m2 on days 1–5 starting every 28 days) plus 10 mg/kg BEV every 14 days until disease progression was evident or for a maximum of 24 months. The median OS was 19.6 months (95% CI, 6.1–23.3 months) and the median PFS was 13.6 months (95% CI, 11.1–16.5 months). Lai et al compared these findings with the results of the trial conducted by Stupp et al (3) (OS, 14.6 months; median PFS, 6.9 months) and by the University of California, Los Angeles (OS, 21.1 months; median PFS, 7.6 months). Lai et al concluded that patients treated with BEV and TMZ during and after RT demonstrated improved PFS without improved OS, as compared with the findings presented by Stupp et al and the findings of the University of California, Los Angeles trial (19). Additional studies are required in order to determine whether first-line administration of BEV improves survival, as compared with the use of BEV at recurrence.

Various other phase II studies have evaluated the efficacy of the addition of BEV for the treatment of patients with newly diagnosed GBM (Table II). Narayana et al (37) investigated 51 patients with newly diagnosed GBM treated with RT and concomitant TMZ alongside 10 mg/kg BEV every 2 weeks, initiated 4 weeks post-surgery. This regimen was followed by 6 cycles of adjuvant standard-dose TMZ therapy with 10 mg/kg BEV administered on days 8 and 22 of each 28-day cycle. PFS-6 and 12-month PFS (PFS-12) rates were 85.1 and 51%, respectively, whereas the OS rates at 12 months (OS-12) and 24 months (OS-24) were 85.1 and 42.5%, respectively. Furthermore, 19.6% of the patients (10/51 patients) experienced grade III/IV toxicity, and asymptomatic intracranial bleeding was observed in 5 patients; however, no treatment-related mortality was observed. Narayana et al concluded that the addition of BEV to conventional therapy in patients with newly diagnosed GBM appears to improve both PFS and OS in patients with newly diagnosed GBM, with tolerable toxicity (37).

Table II.

Efficacy of bevacizumab alone or in combination with cytotoxic agents in newly diagnosed glioblastoma.

Table II.

Efficacy of bevacizumab alone or in combination with cytotoxic agents in newly diagnosed glioblastoma.

PFS
OS
Author, yearTreatment regimenPatients (n)Median (months)PFS-6 (%)PFS-12 (%)Median (months)OS-6 (%)OS-12 (%)(Ref.)
Gilbert et al, 2014RT + TMZ + BEV32010.7804415.78767(15)
RT + TMZ3177.3543116.18665
Chinot et al, 2014RT + TMZ + BEV45810.6794016.89372.4(16)
RT + TMZ4636.2502716.79066.3
Lai et al, 2011RT + TMZ + BEV7013.688N/A19.6N/AN/A(36)
Narayana et al, 2012RT + TMZ + BEV511385.15123N/A85.1(37)
Vredenburgh et al, 2011RT + TMZ + BEV + CPT-117514.2N/A62.721.2N/A78.7(38)
Vredenburgh et al, 2012RT + TMZ + BEV + CPT-1112513.887.264N/A9482(39)
Hainsworth et al, 2012RT + TMZ + BEV + everolimus6811.3N/AN/A13.9N/AN/A(67)
Herrlinger et al, 2013RT + BEV + CPT-11N/AN/AN/AN/A16.6N/AN/A(68)
RT + TMZN/AN/AN/AN/A14.8N/AN/A

[i] PFS, progression-free survival; OS, overall survival; PFS-6, 6-month PFS rate; PFS-12, 12-month PFS rate; OS-6, 6-month OS rate; OS-12, 12-month OS rate; RT, radiotherapy; TMZ, temozolomide; BEV, bevacizumab; N/A, not available; CPT-11, irinotecan.

Vredenburgh et al (38) conducted an upfront, phase II trial in patients with newly diagnosed GBM 75), which evaluated the addition of BEV to standard RT and daily TMZ administration followed by the addition of BEV and CPT-11 to adjuvant TMZ. BEV was administered at 10 mg/kg every 2 weeks and was initiated a minimum of 4 weeks post-craniotomy. Following 2 weeks of RT, the patients began 6 to 12 cycles of 5-day TMZ with BEV and CPT-11 administration every 2 weeks. The median OS was 21.2 months (95% CI, 17.2–25.4 months), and 65% of the patients survived to 16 months (95% CI, 53.4–74.9%). The median PFS was 14.2 months (95% CI, 12–16 months). Vredenburgh et al concluded that this therapeutic regimen may improve the efficacy of treatment for patients with newly diagnosed GBM, as compared with historical controls (39). After 1 year Vredenburg et al reported on 125 patients with newly diagnosed GBM that were treated with standard external beam irradiation plus concurrent TMZ followed by adjuvant BEV, TMZ, and CPT-11 (39); the PFS at 6 months, 1 year and 2 years was 88, 64 and 16%, respectively; whereas OS was 94, 82 and 44%, respectively.

Previously, two large randomized phase III trials evaluated the role of BEV in combination with TMZ and RT in patients newly diagnosed with GBM. The first, entitled RTOG 0825 (15), was a randomized phase III double-blind, placebo-controlled trial sponsored by RTOG, including 637 patients enrolled into BEV or placebo plus standard RT and TMZ groups following surgery for GBM. During maintenance therapy, patients were treated with BEV or placebo plus TMZ until disease progression was evident or the toxicity became intolerable (Fig. 1A). In the patients who received BEV, the median OS was demonstrated to be 15.7 months, as compared with 16.1 months in the patients who received placebo. The hazard ratio for mortality in the BEV group was 1.13. The median PFS in the BEV and placebo groups was 10.7 months and 7.3 months, respectively; and the hazard ratio for progression or mortality was 0.79 (15). Another randomized phase III double-blind, placebo-controlled trial, was sponsored by Hoffman-La Roche, entitled the AVAglio (16) trial (Fig. 1B). A total of 921 patients were enrolled following surgery for GBM and were subsequently randomized into BEV or placebo groups plus standard RT and TMZ followed by TMZ and treatment maintenance with BEV or placebo until disease progression was demonstrated (Fig. 1B). OS rates at 1 year following treatment with BEV or placebo were 72.4 and 6.3% (P=0.049); whereas the 2-year follow-up OS rates were 33.9 and 30.1% (P=0.24), respectively. The median PFS rates in the BEV and placebo groups were 10.6 and 6.2 months, respectively, with a hazard ratio for progression or mortality of 0.64 (95% CI, 0.55–0.74; P<0.001) (16). Notably, both trials concluded that the addition of BEV standard treatment did not improve OS in patients with newly diagnosed GBM; however, the median PFS was increased in the BEV group, as compared with the placebo group in both trials.

Safety of bevacizumab treatment in patients with glioblastoma

Although BEV is an enticing agent for the treatment of GBM, it has well-recognized complications (40). Common and significant adverse events of all grades that have been associated with BEV monotherapy for patients with recurrent GBM include fatigue (32–63%), headache (20–36.9%), hypertension (12.5–29.8%), hemorrhage (overall, 27.4%), thromboembolic event (8–12.5%), and proteinuria (2.1–10%) (25,27,29,30). In previous studies, the most common adverse events detected following treatment with a combination of BEV and cytotoxic agents were hemorrhage (overall, 17.6–40.5%), fatigue (11.4–75.9%), hypertension (3.5–26.6%), and diarrhea (74.7%) (23,25,4145).

The safety of BEV when combined with TMZ and RT in the standard chemoradiotherapy schedule for patients with newly diagnosed GBM was evaluated in the RTOG 0825 and AVAglio studies. In the AVAglio study, adverse events of all grades were demonstrated in 98.5% of the patients who received BEV treatment, as compared with 96.0% in the placebo group. Adverse events of ≥grade 3 were demonstrated in 66.8 and 51.3% of patients, respectively. The rate of serious adverse events was 38.8% in the BEV group, as compared with 25.6% in the placebo group, and adverse events ≥grade 3 that are often associated with BEV were 32.5 vs. 15.8% (16). Similarly, the RTOG 0825 study (15) concluded that the incidence of adverse events was increased in the BEV group, as compared with the placebo group; and the most common adverse events detected were hypertension, hemorrhage, proteinuria, and thromboembolic events (Table III).

Table III.

Major adverse events following bevacizumab treatment with and without chemotherapeutics in patients with glioblastoma.

Table III.

Major adverse events following bevacizumab treatment with and without chemotherapeutics in patients with glioblastoma.

Bevacizumab alone
Bevacizumab with chemotherapeutics
Bevacizumab with temozolomide and RT
ToxicitiesAll gradesGrade ≥3All gradesGrade ≥3All gradesGrade ≥3
Fatigue32–633.6–5511.4–75.94–8.9207.4
Headache20–36.91832.90N/AN/A
Hypertension12.5–29.88.33.5–26.61.3–239.311.3
Thromboembolic event8–12.56–95–11.4414.112.6
Proteinuria2.1–10N/A2.5–5.71.315.65.4
Wound dehiscence4–62.43.6–5.51.3–5.56.93.3
Hemorrhage
  Overall27.4017.6–40.52.537.11.3
  Intracranial2.40–42.9–3.81.33.32.0
Bowel perforation2.1–92.12.52.51.71.1
Anemia10N/AN/AN/AN/AN/A
Neutropenia2.41.274–15N/AN/A
Diarrhea21.41.274.75.1–6N/AN/A

[i] Selected adverse events reported in large (≥35 patients) phase II or III studies and retrospective analyses. Bevacizumab alone (25,27,29,30). Bevacizumab with chemotherapeutics (23,25,4145) for recurrent glioblastoma. Bevacizumab plus temozolomide and RT (16) for newly diagnosed glioblastoma. RT, radiotherapy.

Hypertension, which was demonstrated to be the most common adverse event in patients treated with BEV, may lead to hemorrhage, thromboembolic event, cerebral ischemia and proteinuria. VEGF normally increases endothelial transcription of nitric oxide (NO) synthase, leading to the increased production of NO, which is a potent vasodilator. Anti-VEGF agents, including BEV, are capable of decreasing NO production, which may induce vasoconstriction and result in hypertension. At the renal level, this vasoconstriction induces sodium retention which may contribute to hypertension (46). Furthermore, anti-VEGF agents may reduce the concentration of microvascular beds, a phenomenon known as ‘rarefaction’, thus increasing systemic vascular resistance and blood pressure (46). A previous meta-analysis of clinical trials that randomized patients with numerous tumor types concluded that there was a significant dose-dependent increase in the risk of hypertension in patients with tumors following treatment with BEV (47). Standard schedules for the treatment of BEV-associated hypertension are yet to be elucidated. Some scholars have suggested that the general principles of hypertension management should be followed (40); however, this theory lacks sufficient evidence.

Proteinuria is a characteristic adverse event often exhibited in patients following treatment with anti-VEGF agents. A previous meta-analysis demonstrated that the incidence of grade III/IV proteinuria in patients treated with BEV was 2.2% (RR, 4.8) (48), and a significant dose-dependent increase in the risk of proteinuria was detected in patients with tumors who received BEV (47). The incidence of BEV-related proteinuria appears to be lower in patients with GBM, as compared with other cancers. A previous randomized noncomparative phase II trial demonstrated grade 1 proteinuria in only 4% of patients, and grade 3 proteinuria in just 1 of the 167 patients (25). Why proteinuria is less common in patients with brain tumors has yet to be examined, however it may be associated with a shorter median duration of therapy.

For patients with malignant brain tumors, intracranial hemorrhage may be a life-threatening event. Anti-VEGF agents inhibit the proliferation and survival of vascular endothelial cells, particularly in tissues with a high dependence on VEGF, leading to dysregulation of the coagulation cascade, injury to the mucosal membrane of the airway, damage to the tumor-infiltrated vascular wall as a consequence of an antitumor effect, decreased matrix deposition in the supporting layers of the vessels, and occasionally, treatment-induced thrombocytopenia. All of these events may be associated with the mechanisms of hemorrhage (15). Although the rate of hemorrhage has previously been demonstrated to be as high as 40% (25), the majority are low-grade systemic hemorrhages, including epistaxis; and life-threatening intracranial hemorrhages have been demonstrated in <3.8% of patients treated with BEV (Table III).

Thromboembolic events, including venous thromboembolism and arterial thromboembolism, are also common in GBM treated with anti-VEGF agents. However, due to the variable and often high rates of venous thromboembolic events demonstrated in patients with GBM (41,49), it is often difficult to determine whether the reported incidence of venous thromboembolic events exceeds the anticipated rate normally associated with the disease. Conversely, as arterial thromboses are uncommon in patients with GBM, arterial thromboembolism is considered to be directly associated with anti-angiogenic therapy, and patients who develop arterial thrombosis should cease anti-angiogenic treatment. Treatment of arterial thrombosis should be guided by the disease process, recognizing that the optimal management of stroke, myocardial infarction and peripheral vascular occlusion may be distinct (41).

In addition to common toxicity, other common adverse events detected following BEV treatment in patients with recurrent or newly diagnosed GBM include fatigue, diarrhea, headache and impaired wound healing. The majority of these adverse events appear to reflect the destruction of VEGF in normal organs and tissues induced by the on-target, class-specific actions of anti-VEGF agents. These adverse events must be monitored in order to guarantee the basic quality of life of patients treated with BEV.

Discussion

Whether it is used as a single-agent or in combination with other cytotoxic agents, BEV has been demonstrated to be an effective therapy with tolerable toxicity for the treatment of patients with GBM. As compared with historical controls of patients with recurrent GBM, BEV-related treatment prolonged PFS and OS (25,27). Double-blinded, placebo-controlled phase III studies concluded that the addition of BEV to the first-line treatment improved PFS in patients with newly diagnosed GBM (15,16). Hypertension and proteinuria were characteristic adverse events in the course of treatment. Various life-threatening adverse events, including intracranial hemorrhage and thromboembolic events were demonstrated in patients with GBM; however, the fatal event rate was low and these events have also been associated with GBM itself. However, problems remain regarding the role of BEV in the treatment of GBM.

Firstly, BEV may be used as monotherapy or in combination with chemotherapy and/or RT in the treatment of patients with recurrent GBM. Whether combination therapy with BEV is superior to BEV as a single agent in the treatment of patients with recurrent GBM remains uncertain. If combination therapy with BEV proved superior to BEV monotherapy, the optimal therapeutic partner has yet to be identified.

Secondly, there are no clear standards for the optimal dose and duration of BEV treatment for patients with recurrent GBM. The majority of clinical trials have used 5–15 mg/kg BEV every 2–4 weeks; however it is yet to be elucidated whether higher doses of BEV improve clinical efficacy. Furthermore, it remains unclear whether BEV dosage should be adjusted according to the patient's age, the degree of glioma malignancy, the progress of the disease throughout the process of treatment, or whether the original treatment should be changed or terminated according to the patient's toxicity tolerance.

Thirdly, further research is required into how to manage the adverse events associated with BEV treatment. Toxicity is inevi›, however reducing toxicity may improve the quality of life of the patients. Whether the management of adverse events associated with BEV should or should not follow the general principles is still uncertain, and further studies are required.

In conclusion, the main issue with BEV therapy is the lack of biomarkers and genetic models to identify patients who may benefit from BEV treatment (17). It is hoped that further studies investigating biomarkers and genetic patterns will identify patients who may benefit from treatment with BEV or other anti-angiogenic agents, and these studies may also suggest other treatable cellular targets that may be critical to the advancement of treatment for patients with GBM (17). Therefore, more randomized controlled trials are required for patients with GBM in order to provide definitive answers on the optimal therapeutic partner, dose and length of treatment.

References

1 

Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW and Kleihues P: The 2007 WHO classification of tumors of the central nervous system. Acta Neuropathol. 114:97–109. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Wen PY and Kesari S: Malignant gliomas in adults. N Engl J Med. 359:492–507. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al: Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 10:459–466. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Wen PY and DeAngelis LM: Chemotherapy for low-grade gliomas: Emerging consensus on its benefits. Neurology. 68:1762–1763. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Wong ET, Hess KR, Gleason MJ, Jaeckle KA, Kyritsis AP, Prados MD, Levin VA and Yung WK: Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J Clin Oncol. 17:2572–2578. 1999.PubMed/NCBI

7 

Folkman J: What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst. 82:4–6. 1990. View Article : Google Scholar : PubMed/NCBI

8 

Hicklin DJ and Ellis LM: Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol. 23:1011–1027. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Folkman J: Tumor angiogenesis: T herapeutic implications. N Engl J Med. 285:1182–1186. 1971. View Article : Google Scholar : PubMed/NCBI

10 

Hanahan D and Folkman J: Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 86:353–364. 1996. View Article : Google Scholar : PubMed/NCBI

11 

Siemann DW and Shi W: Targeting the tumor blood vessel network to enhance the efficacy of radiation therapy. Semin Radiat Oncol. 13:53–61. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Miletic H, Niclou SP, Johansson M and Bjerkvig R: Anti-VEGF therapies for malignant glioma, Treatment effects and escape mechanisms. Expert Opin Ther Targets. 13:455–468. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Ferrara N, Gerber HP and LeCouter J: The biology of VEGF and its receptors. Nat Med. 9:669–676. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Beal K, Abrey LE and Gutin PH: Antiangiogenic agents in the treatment of recurrent or newly diagnosed glioblastoma, Analysis of single-agent and combined modality approaches. Radiat Oncol. 6:22011. View Article : Google Scholar : PubMed/NCBI

15 

Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, Colman H, Chakravarti A, Pugh S, Won M, et al: A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 370:699–708. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, Carpentier AF, Hoang-Xuan K, Kavan P, Cernea D, et al: Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 370:709–722. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Gil-Gil MJ, Mesia C, Rey M and Bruna J: Bevacizumab for the treatment of glioblastoma. Clin Med Insights Oncol. 7:123–135. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Lu JF, Bruno R, Eppler S, Novotny W, Lum B and Gaudreault J: Clinical pharmacokinetics of bevacizumab in patients with solid tumors. Cancer Chemother Pharmacol. 62:779–786. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Specenier P: Bevacizumab in glioblastoma multiforme. Expert Rev Anticancer Ther. 12:9–18. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Wong ET, Gautam S, Malchow C, Lun M, Pan E and Brem S: Bevacizumab for recurrent glioblastoma multiforme, A meta-analysis. J Natl Compr Canc Netw. 9:403–407. 2011.PubMed/NCBI

21 

Stark-Vance V: Bevacizumab and CPT-11 in the treatment of relapsed malignant glioma. Neuro Oncol. 7:3692005.

22 

Vredenburgh JJ, Desjardins A, Herndon JE II, Dowell JM, Reardon DA, Quinn JA, Rich JN, Sathornsumetee S, Gururangan S, Wagner M, et al: Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 13:1253–1259. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Vredenburgh JJ, Desjardins A, Herndon JE II, Marcello J, Reardon DA, Quinn JA, Rich JN, Sathornsumetee S, Gururangan S, Sampson J, et al: Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 25:4722–4729. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Desjardins A, VJ, Reardon DA, Herndon JE, Marcello J, Peters K, Gururangan S, Sathornsumetee S, Rich JN and Friedman HS: Long-term survival from the initial trial of bevacizumab and irinotecan. J Clin Oncol. 28(15 Suppl): 1912010.PubMed/NCBI

25 

Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE, Yung WK, Paleologos N, Nicholas MK, Jensen R, et al: Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 27:4733–4740. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Cloughesy T, VJ, Day B, Das A and Friedman HS: The BRAIN Investigators: Updated safety and survival of patients with relapsed glioblastoma treated with bevacizumab in the BRAIN study. J Clin Oncol. 28(15 Suppl): 181s2010.

27 

Kreisl TN, Kim L, Moore K, Duic P, Royce C, Stroud I, Garren N, Mackey M, Butman JA, Camphausen K, et al: Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol. 27:740–745. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Cohen MH, Shen YL, Keegan P and Pazdur R: FDA drug approval summary, Bevacizumab (Avastin) as treatment of recurrent glioblastoma multiforme. Oncologist. 14:1131–1138. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Chamberlain MC and Johnston SK: Salvage therapy with single agent bevacizumab for recurrent glioblastoma. J Neurooncol. 96:259–269. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Raizer JJ, Grimm S, Chamberlain MC, Nicholas MK, Chandler JP, Muro K, Dubner S, Rademaker AW, Renfrow J and Bredel M: A phase 2 trial of single-agent bevacizumab given in an every-3-week schedule for patients with recurrent high-grade gliomas. Cancer. 116:5297–5305. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Kreisl TN, Zhang W, Odia Y, Shih JH, Butman JA, Hammoud D, Iwamoto FM, Sul J and Fine HA: A phase II trial of single-agent bevacizumab in patients with recurrent anaplastic glioma. Neuro Oncol. 13:1143–1150. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Nagane M, Nishikawa R, Narita Y, Kobayashi H, Takano S, Shinoura N, Aoki T, Sugiyama K, Kuratsu J, Muragaki Y, et al: Phase II study of single-agent bevacizumab in Japanese patients with recurrent malignant glioma. Jpn J Clin Oncol. 42:887–895. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Hofer S, Elandt K, Greil R, Hottinger AF, Huber U, Lemke D, Marosi C, Ochsenbein A, Pichler J, Roelcke U, et al: Clinical outcome with bevacizumab in patients with recurrent high-grade glioma treated outside clinical trials. Acta Oncol. 50:630–635. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Zhang G, Huang S and Wang Z: A meta-analysis of bevacizumab alone and in combination with irinotecan in the treatment of patients with recurrent glioblastoma multiforme. J Clin Neurosci. 19:1636–1640. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Balañá C, Etxaniz O, Bugés C and Martínez A: Approval denied by the european medicines agency (EMA) for bevacizumab in the treatment of high-grade glioma recurrence A good idea or a grave error? Clin Transl Oncol. 13:209–210. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Lai A, Tran A, Nghiemphu PL, Pope WB, Solis OE, Selch M, Filka E, Yong WH, Mischel PS, Liau LM, et al: Phase II study of bevacizumab plus temozolomide during and after radiation therapy for patients with newly diagnosed glioblastoma multiforme. J Clin Oncol. 29:142–148. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Narayana A, Gruber D, Kunnakkat S, Golfinos JG, Parker E, Raza S, Zagzag D, Eagan P and Gruber ML: A clinical trial of bevacizumab, temozolomide and radiation for newly diagnosed glioblastoma. J Neurosurg. 116:341–345. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Vredenburgh JJ, Desjardins A, Reardon DA, Peters KB, Herndon JE II, Marcello J, Kirkpatrick JP, Sampson JH, Bailey L, Threatt S, et al: The addition of bevacizumab to standard radiation therapy and temozolomide followed by bevacizumab, temozolomide and irinotecan for newly diagnosed glioblastoma. Clin Cancer Res. 17:4119–4124. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Vredenburgh JJ, Desjardins A, Kirkpatrick JP, Reardon DA, Peters KB, Herndon JE II, Marcello J, Bailey L, Threatt S, Sampson J, et al: Addition of bevacizumab to standard radiation therapy and daily temozolomide is associated with minimal toxicity in newly diagnosed glioblastoma multiforme. Int J Radiat Oncol Biol Phys. 82:58–66. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Armstrong TS, Wen PY, Gilbert MR and Schiff D: Management of treatment-associated toxicites of anti-angiogenic therapy in patients with brain tumors. Neuro Oncol. 14:1203–1214. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Norden AD, Young GS, Setayesh K, Muzikansky A, Klufas R, Ross GL, Ciampa AS, Ebbeling LG, Levy B, Drappatz J, et al: Bevacizumab for recurrent malignant gliomas: Efficacy, toxicity and patterns of recurrence. Neurology. 70:779–787. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Gilbert MR, Wang M, Aldape K, Lassman A, Sorenson AG, Mikkelson T, Groves M, Wener-Wasik M, Regine W and Mehta M: RTOG 0625: A phase II study of bevacizumab with irinotecan in recurrent glioblastoma (GBM). J Clin Oncol. 27(Suppl 15): 89s2009.

43 

Gil MJ, de Las Peñas R, Reynés G, Balañá C, Peréz-Segura P, García-Velasco A, Mesia C, Gallego O, Fernández-Chacón C, Martínez-García M, et al: Bevacizumab plus irinotecan in recurrent malignant glioma shows high overall survival in a multicenter retrospective pooled series of the Spanish neuro-oncology research group (GEINO). Anticancer Drugs. 23:659–665. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Zuniga RM, Torcuator R, Jain R, Anderson J, Doyle T, Ellika S, Schultz L and Mikkelsen T: Efficacy, safety and patterns of response and recurrence in patients with recurrent high-grade gliomas treated with bevacizumab plus irinotecan. J Neurooncol. 91:329–336. 2009. View Article : Google Scholar : PubMed/NCBI

45 

Quant EC, Norden AD, Drappatz J, Muzikansky A, Doherty L, Lafrankie D, Ciampa A, Kesari S and Wen PY: Role of a second chemotherapy in recurrent malignant glioma patients who progress on bevacizumab. Neuro Oncol. 11:550–555. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Gurevich F and Perazella MA: Renal effects of anti-angiogenesis therapy: Update for the internist. Am J Med. 122:322–328. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Zhu X, Wu S, Dahut WL and Parikh CR: Risks of proteinuria and hypertension with bevacizumab, an antibody against vascular endothelial growth factor, Systematic review and meta-analysis. Am J Kidney Dis. 49:186–193. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Wu S, Kim C, Baer L and Zhu X: Bevacizumab increases risk for severe proteinuria in cancer patients. J Am Soc Nephrol. 21:1381–1389. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Rahmathulla G, Hovey EJ, Hashemi-Sadraei N and Ahluwalia MS: Bevacizumab in high-grade gliomas, A review of its uses, toxicity assessment, and future treatment challenges. Onco Targets Ther. 6:371–389. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Poulsen HS, Grunnet K, Sorensen M, Olsen P, Hasselbalch B, Nelausen K, Kosteljanetz M and Lassen U: Bevacizumab plus irinotecan in the treatment patients with progressive recurrent malignant brain tumors. Acta Oncol. 48:52–58. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Raval SHS and Dorsett L: Bevacizumab and irinotecan in patients (pts) with recurrent glioblastoma multiforme (GBM). J Clin Oncol. 25(Suppl 18): 20782007.PubMed/NCBI

52 

Bokstein F, Shpigel S and Blumenthal DT: Treatment with bevacizumab and irinotecan for recurrent high-grade glial tumors. Cancer. 112:2267–2273. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Taillibert S, Hoang-Xuan K, Barrie M, Guiu S, Chauffert B, Cartalat-Carel S, Taillandier L, Fabbro M, Laigre M, Guillamo JS, et al: ANOCEF group: Bevacizumab (B) with irinotecan (I) in recurrent glioblastoma (GBM): A national retrospective cohort of the ANOCEF (Association Des Neuro-Oncologues D'Expression Française) group. Neuro Oncol. 12((Suppl 4)): iv52–iv53. 2010.

54 

Keyrouz VF, Elias E, Chahine GY, Comair YG, Dimassi H and Kamar FG: Updated results of a phase II trial of bevacizumab and irinotecan in relapsed high-grade glioma. Neuro Oncol. 12(Suppl 3): iii472010.

55 

Pope WB, Lai A, Nghiemphu P, Mischel P and Cloughesy TF: MRI in patients with high-grade gliomas treated with bevacizumab and chemotherapy. Neurology. 66:1258–1260. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Nghiemphu PL, Liu W, Lee Y, Than T, Graham C, Lai A, Green RM, Pope WB, Liau LM, Mischel PS, et al: Bevacizumab and chemotherapy for recurrent glioblastoma: A single-institution experience. Neurology. 72:1217–1222. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Desjardins A, Reardon DA, Coan A, Marcello J, Herndon JE II, Bailey L, Peters KB, Friedman HS and Vredenburgh JJ: Bevacizumab and daily temozolomide for recurrent glioblastoma. Cancer. 118:1302–1312. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Verhoeff JJ, Lavini C, van Linde ME, Stalpers LJ, Majoie CB, Reijneveld JC, van Furth WR and Richel DJ: Bevacizumab and dose-intense temozolomide in recurrent high-grade glioma. Ann Oncol. 21:1723–1727. 2010. View Article : Google Scholar : PubMed/NCBI

59 

Soffietti R, Trevisan E, Ruda R, Bertero L, Bosa C, Fabrini MG and Lolli I: Phase II trial of bevacizumab with fotemustine in recurrent glioblastoma, Final results of a multicenter study of AINO (Italian Association of Neuro-oncology). J Clin Oncol. 29:2011.PubMed/NCBI

60 

Trevisan E, Ruda R, Picco E, Crasto Greco S, Caroli M, Fabrini A, Scotti V, Lolli I, Guarneri D and Soffietti R: Bevacizumab and fotemustine as salvage therapy in recurrent glioblastoma, A phase II multicenter Italian study. Neuro Oncol. 12((Suppl 3)): iii49–iii50. 2010.

61 

Hasselbalch B, Lassen U, Hansen S, Holmberg M, Sørensen M, Kosteljanetz M, Broholm H, Stockhausen MT and Poulsen HS: Cetuximab, bevacizumab, and irinotecan for patients with primary glioblastoma and progression after radiation therapy and temozolomide, A phase II trial. Neuro Oncol. 12:508–516. 2010.PubMed/NCBI

62 

Shapiro LQ, Beal K, Goenka A, Karimi S, Iwamoto FM, Yamada Y, Zhang Z, Lassman AB, Abrey LE and Gutin PH: Patterns of failure after concurrent bevacizumab and hypofractionated stereotactic radiation therapy for recurrent high-grade glioma. Int J Radiat Oncol Biol Phys. 85:636–642. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Sathornsumetee S, Desjardins A, Vredenburgh JJ, McLendon RE, Marcello J, Herndon JE, Mathe A, Hamilton M, Rich JN, Norfleet JA, et al: Phase II trial of bevacizumab and erlotinib in patients with recurrent malignant glioma. Neuro Oncol. 12:1300–1310. 2010.PubMed/NCBI

64 

Niyazi M, Ganswindt U, Schwarz SB, Kreth FW, Tonn JC and Geisler J: laF ougère C, Ertl L, Linn J, Siefert A and Belka C: Irradiation and bevacizumab in high-grade glioma retreatment settings. Int J Radiat Oncol Biol Phys. 82:67–76. 2012. View Article : Google Scholar : PubMed/NCBI

65 

Hundsberger T, Brügge D, Putora PM, Weder P, Weber J and Plasswilm L: Re-irradiation with and without bevacizumab as salvage therapy for recurrent or progressive high-grade gliomas. J Neurooncology. 112:133–139. 2013. View Article : Google Scholar

66 

Gutin PH, Iwamoto FM, Beal K, Mohile NA, Karimi S, Hou BL, Lymberis S, Yamada Y, Chang J and Abrey LE: Safety and efficacy of bevacizumab with hypofractionated stereotactic irradiation for recurrent malignant gliomas. Int J Radiat Oncol Biol Phys. 75:156–163. 2009. View Article : Google Scholar : PubMed/NCBI

67 

Hainsworth JD, Shih KC, Shepard GC, Tillinghast GW, Brinker BT and Spigel DR: Phase II study of concurrent radiation therapy, temozolomide and bevacizumab followed by bevacizumab/everolimus as first-line treatment for patients with glioblastoma. Clin Adv Hematol Oncol. 10:240–246. 2012.PubMed/NCBI

68 

Herrlinger U, Schafer N, Steinbach JP, Weyerbrock A, Hau P, Goldbrunner R, Friedrich F, Stockhammer F, Ringel F, Braun C, et al: Bevacizumab, irinotecan and radiotherapy versus standard temozolomide and radiotherapy in newly diagnosed, MGMT non-methylated glioblastoma patients: Updated results from the randomized multicenter GLARIUS trial. European J Cancer. 49:S7742013.

Related Articles

Journal Cover

February-2016
Volume 11 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu Z, Zhao G, Zhang Z, Li Y, Chen Y, Wang N, Zhao Z and Xie G: Efficacy and safety of bevacizumab for the treatment of glioblastoma (Review). Exp Ther Med 11: 371-380, 2016
APA
Yu, Z., Zhao, G., Zhang, Z., Li, Y., Chen, Y., Wang, N. ... Xie, G. (2016). Efficacy and safety of bevacizumab for the treatment of glioblastoma (Review). Experimental and Therapeutic Medicine, 11, 371-380. https://doi.org/10.3892/etm.2015.2947
MLA
Yu, Z., Zhao, G., Zhang, Z., Li, Y., Chen, Y., Wang, N., Zhao, Z., Xie, G."Efficacy and safety of bevacizumab for the treatment of glioblastoma (Review)". Experimental and Therapeutic Medicine 11.2 (2016): 371-380.
Chicago
Yu, Z., Zhao, G., Zhang, Z., Li, Y., Chen, Y., Wang, N., Zhao, Z., Xie, G."Efficacy and safety of bevacizumab for the treatment of glioblastoma (Review)". Experimental and Therapeutic Medicine 11, no. 2 (2016): 371-380. https://doi.org/10.3892/etm.2015.2947