Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jun 15.
Published in final edited form as: Cancer. 2020 Mar 10;126(12):2821–2828. doi: 10.1002/cncr.32811

NRG/RTOG 1122: phase II double-blinded, placebo-controlled study of bevacizumab with/without trebananib in patients with recurrent glioblastoma or gliosarcoma

Eudocia Q Lee 1, Peixin Zhang 2, Patrick Y Wen 1, Elizabeth R Gerstner 3, David A Reardon 1, Kenneth D Aldape 4, John F deGroot 5, Edward Pan 6, Jeffrey J Raizer 7, Lyndon J Kim 8, Steven J Chmura 9, H Ian Robins 10, Jennifer M Connelly 11, James D Battiste 12, John L Villano 13, Naveed Wagle 14, Ryan T Merrell 15, Merideth M Wendland 16, Minesh P Mehta 17
PMCID: PMC7245544  NIHMSID: NIHMS1582953  PMID: 32154928

Abstract

Background:

Targeting vascular endothelial growth factor (VEGF) alone does not improve overall survival in recurrent glioblastoma (rGBM). The angiopoiein (Ang)-TIE2 system may play a role in tumor survival under VEGF inhibition. We conducted a phase II, double-blinded, placebo-controlled trial of bevacizumab + trebananib (a novel Fc fusion protein that sequesters Ang1/Ang2) over bevacizumab alone in rGBM.

Methods:

Patients 18 years or older with a KPS ≥ 70 and GBM or variants in first or second relapse were randomized to bevacizumab 10 mg/kg every 2 weeks + trebananib 15 mg/kg every week or bevacizumab + placebo. The primary endpoint was 6-month progression-free survival (6m PFS).

Results:

After an initial 6-patient lead-in cohort confirmed the safety of combining bevacizumab and trebananib, 115 eligible patients were randomized to the control (n=58) or experimental treatment (n=57). In the control arm, 6m PFS was 41.1%, median survival time (MST) was 11.5 months [95% CI: 8.4-14.2], median PFS was 4.8 months [95% CI: 3.8-7.1], and RR was 5.9%. In the experimental arm, 6m PFS was 22.6%, MST was 7.5 months [95% CI 6.8-10.1], median PFS was 4.2 months [95% CI 3.7-5.6], and RR was 4.2%. The rate of severe toxicities was not significantly different between arms.

Conclusion:

The combination of bevacizumab and trebananib was well tolerated but did not significantly improve 6m PFS rate, PFS, or OS for patients with rGBM over bevacizumab alone. The shorter PFS in the experimental arm with a HR=1.51 (p=0.04) suggests that the addition of trebananib to bevacizumab is detrimental.

Trial Registration:

ClinicalTrials.gov identifier NCT01609790

Keywords: Glioblastoma, angiogenesis, bevacizumab, angiopoietin, trebananib

Precis:

In this phase II, double-blinded, placebo-controlled trial in patients with recurrent GBM through NRG oncology, the combination of bevacizumab plus trebananib (an angiopoietin inhibitor) did not significantly improve 6m PFS rate, PFS, or OS for patients with recurrent GBM as compared to bevacizumab plus placebo. The Ang1 blocking effects of trebananib may somehow counteract or negate the anti-tumor effects of Ang2-VEGF blockade.

Introduction

Bevacizumab, a monoclonal antibody against vascular endothelial growth factor (VEGF), is frequently used to treat patients with recurrent glioblastoma (GBM) and received FDA approval for this indication.1, 2 Although randomized, phase III trials in newly diagnosed GBM suggest an improvement in progression-free survival (PFS), the addition of bevacizumab to standard therapy is not associated with an improvement in overall survival (OS) compared to standard therapy alone.3, 4 Indeed, GBMs often develop resistance to bevacizumab treatment within months of starting therapy.1, 5 Targeting VEGF alone may not be sufficient for durable responses. Intrinsic and acquired resistance may be due to the redundancy of angiogenesis stimulators.6, 7

Angiopoietins (Ang) may play a role in mediating resistance to anti-VEGF therapy in GBM. In a phase II clinical trial of cediranib [a pan-VEGF receptor (VEGFR) inhibitor] in recurrent GBM, progression on cediranib correlated with increases in the soluble form of the Ang-binding Tie2 receptor.8, 9 In a U87 glioma xenograft model, dual inhibition of VEGFR and Ang2 inhibited tumor growth, prolonged vessel normalization compared to VEGFR inhibition alone, and improved survival through reprogramming of tumor-associated macrophages toward an antitumor phenotype.10, 11 In addition, a subset of GBM patients on anti-VEGF therapy develops a non-enhancing tumor pattern of progression, believed to represent a more invasive phenotype.5, 12, 13 Upregulation of Ang2 correlates with the invasiveness of gliomas.14, 15 Blockade of Ang2 may decrease the invasiveness of gliomas and abrogate an escape mechanism for anti-VEGF therapy. Data from a colon cancer xenograft model suggest that combined Ang1/Ang2 inhibition confers better tumor suppression than Ang2 inhibition alone.16 Furthermore, in the same models, Ang1/Ang2 inhibition plus bevacizumab mediated better tumor suppression than Ang2 inhibition plus bevacizumab. For these reasons, dual inhibition of the VEGF and angiopoietin-TIE2 signaling pathways is an attractive therapeutic strategy in the recurrent GBM population.

Trebananib (AMG 386) is a novel peptide-Fc fusion protein that sequesters Ang1 and Ang2, thereby preventing their interaction with Tie2 and inhibiting tumor endothelial cell proliferation and tumor growth. We performed a phase II, double-blinded, placebo-controlled trial in patients with recurrent GBM comparing bevacizumab plus trebananib versus bevacizumab plus placebo.

Patients and Methods

Patients

Eligibility criteria included age ≥18 years at registration with a histologic diagnosis of GBM or variants (gliosarcoma, glioblastoma with oligodendroglial features, giant cell glioblastoma) in first or second relapse with unequivocal radiographic evidence for tumor progression ≤14 days prior to registration. Patients with a secondary GBM were also eligible as long as GBM had been histologically proven. For patients with a recent tumor resection for progressive disease, registration on study could not occur any sooner than 28 days from surgery; Additional eligibility criteria included supratentorial disease only; Karnofsky performance status (KPS) ≥70; and adequate renal, hepatic, and bone marrow function.

No prior treatment with VEFG inhibitors including bevacizumab or with angiopoietin-TIE2 inhibitors was allowed. Exclusion criteria also included MRI evidence of recent intracranial hemorrhage, history of bleeding diathesis or coagulopathy, clinically significant cardiovascular disease including myocardial infarction and stroke within 180 days prior to registration, and uncontrolled hypertension. Therapeutic or prophylactic therapy with aspirin, a low-molecular weight heparin, or a Factor Xa inhibitor was allowed, but not warfarin.

All patients provided written informed consent. The study was approved by the institutional review board or the equivalent panel at each study center before patient enrollment. The study was registered at clinicaltrials.gov (NCT01609790).

Study Design, Treatment, and Endpoints

For the phase II portion, under the permuted block design,17 patients were stratified by age (<50 vs. ≥50), KPS (70-80 vs. 90-100), and recent resection (Yes vs. No/Biopsy), and then randomly assigned to receive trebananib or placebo with a 1:1 allocation ratio. All patients received bevacizumab at a dose of 10 mg/kg every two weeks. Trebananib (or placebo) was administered intravenously at a dose of 15 mg/kg every week until disease progression or severe treatment-related toxicity. At the time of tumor progression, patients were unblinded. Those who received bevacizumab monotherapy were allowed to cross over and to receive treatment with bevacizumab and trebananib, provided that they still met eligibility criteria.

The primary endpoint was 6-month progression-free survival rate (6m PFS) and was confirmed by central review (EQL, ERG). Secondary endpoints included PFS, OS, radiographic response (RR), and safety. Toxic effects were graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.0. Radiographic assessments were performed by the local investigator and were based on the Response Assessment in Neuro-Oncology (RANO) criteria.18 Additional details regarding study design can be found in the supplement.

Statistical Methods

The primary endpoint was 6m PFS. Based on the March 2009 FDA briefing19, 6m PFS for bevacizumab monotherapy in recurrent GBM patients is 36%. We hypothesized that the addition of trebananib to bevacizumab in bevacizumab-naïve patients would result in a 6m PFS rate of 55%. Assuming exponential distribution for PFS time, these rates corresponded to a median PFS time of 4.1 and 7 months for the control and experimental arm, respectively, with a hazard ratio (HR) of 0.59. With a one-sided significance level at 0.15, a total of 114 patients (57 per arm) would yield an 85% power to detect the projected effect size on 6m PFS, based on a two-sample test on proportions. To guard against up to a 10% ineligible rate, 127 patients were required to be accrued for the phase II portion of the study.

An interim futility analysis on 6m PFS was designed to be performed when 50% of the total required eligible patients had a minimum of 6-month follow-up. The analysis followed the principle of intent-to-treat, with all the eligible cases included in the treatment arm to which they were randomized regardless of what treatment the patients actually received. The decision rule was that if the 6m PFS rate for the experimental arm was worse than that for the control arm by any amount, the study would be stopped and reported early.

PFS was measured from the date of randomization to the date of first progression or death, whichever came first, or otherwise, the last follow-up date on which the patient was reported alive without experiencing disease progression. OS was measured from the date of randomization to the date of death, or otherwise, the last follow-up date on which the patient was reported alive. OS and PFS rates by treatment arm were estimated using the Kaplan-Meier method. The HRs for the treatment effects on OS and PFS were estimated using the Cox proportional hazard model and tested using the log-rank test. Multivariate analyses with the Cox proportional hazard model were performed with the stratification factors and other patient pretreatment characteristics as covariates. Differences in the rate of the reported severe toxicities and RR between treatment arms were tested using the Chi-square test. For all the secondary endpoints, two-sided tests with a significance level of 0.05 were used in the analyses.

The efficacy (as measured by OS, PFS, RR and 6m PFS) of the salvage treatment for patients who crossed over from the control arm to receive the experimental drug after disease progression was investigated as exploratory endpoints. For this group of patients, OS and PFS were measured from the date of initial progression (before cross-over). OS and PFS rates were similarly estimated using the Kaplan-Meier method. The incidence rates of RR and the reported severe toxicities were also calculated.

Results

Patients

One hundred and thirty patients were enrolled across 41 centers in the United States through NRG oncology and randomized on the phase II portion of the study between March 2013 and September 2014 (Figure 1; CONSORT diagram), of whom 15 (11.5%) were subsequently found to be ineligible. In total, 58 and 57 eligible patients randomized to the control and experimental arms, respectively, were included in the analyses. Table 1 shows the distributions of pretreatment characteristics by treatment arm. The distributions of the stratification factors were balanced between the two treatment arms.

Figure 1.

Figure 1.

CONSORT diagram. Enrollment onto the safety run-in portion of the study began on June 4, 2012. It temporarily closed on October 2, 2012, when the target accrual of 7 patients for the safety run-in component was met. Enrollment onto the randomized portion of the study began on March 13, 2013, and accrual was competed on September 2, 2014.

Table 1.

Patient and tumor characteristics at baseline for all eligible patients.

Bevacizumab + Placebo (n=58) Bevacizumab + Trebananib (n=57)
Patient or Tumor Characteristic n % n %
Age (years)
 Median 58 57
 Min - Max 22 - 79 30 - 80
 < 50 17 29.3 14 24.6
 ≥50 41 70.7 43 75.4
Gender
 Male 36 62.1 32 56.1
 Female 22 37.9 25 43.9
Race
 Asian 2 3.4 0 0.0
 Black or African American 3 5.2 2 3.5
 White 53 91.4 53 93.0
 Unknown or not reported 0 0.0 2 3.5
Ethnicity
 Hispanic or Latino 3 5.2 2 3.5
 Not Hispanic or Latino 55 94.8 54 94.7
 Unknown 0 0.0 1 1.8
Karnofsky Performance Status
 70-80 31 53.4 34 59.6
 90-100 27 46.6 23 40.4
Neurologic Function
 No symptoms 12 20.7 11 19.3
 Minor symptoms 32 55.2 27 47.4
 Moderate symptoms (fully active) 10 17.2 16 28.1
 Moderate symptoms (required assistance) 4 6.9 3 5.3
Surgery (Initial Brain Tumor)
 Biopsy only 8 13.8 3 5.3
 Subtotal resection 19 32.8 12 21.1
 Gross total resection 30 51.7 42 73.7
 Other 1 1.7 0 0.0
Recent resection
 No/biopsy only 30 51.7 30 52.6
 Yes 28 48.3 27 47.4
Histologic Tumor Type
 Glioblastoma 53 91.4 53 93.0
 Gliosarcoma 2 3.4 2 3.5
 Glioblastoma with Oligodendroglial Features 2 3.4 1 1.8
 Giant Cell Glioblastoma 0 0.0 1 1.8
 Other 1 1.7 0 0.0

Outcomes

The interim futility analysis was performed in March 2015. Out of the 27 and 30 eligible patients from the experimental and control arms, respectively, 7 (25.9%) and 11 (36.7%) were progression-free at 6 months after randomization. Of note, the total accrual to the study had already been reached and all patients had had 6-month follow-up by the time of this analysis. The report of this analysis was reviewed by the NRG Oncology Data Monitoring Committee, and it was recommended to unblind protocol treatment and to report the final analyses based on the data from all the randomized and eligible patients.

One hundred and nine (94.8%) patients were evaluable for 6m PFS (Table 2). Twelve of the 53 (22.6%) in the experimental arm were progression-free at 6 months after randomization, as compared with 23 of the 56 (41.1%) in the control arm (one-sided p-value=0.98).

Table 2.

Study Outcomes

Outcome Randomized Cohort Cross-Over Cohort
Bevacizumab + Placebo Bevacizumab + Trebananib P-value Bevacizumab + Trebananib
6m PFS (n, %) 23/56 (41.1%) 12/53 (22.6%) 0.98 1/22 (4.5%)

OS (median, 95% CI) 11.5 months (8.4, 14.2) 7.5 months (6.8, 10.1) 0.09 4.9 months (3.8, 8.4)

PFS (median, 95% CI) 4.8 months (3.8, 7.1) 4.2 months (3.7, 5.6) 0.04 2.3 months (2.1, 3.2)

RR for evaluable patients 0.99*
CR 0 0 0
PR 3/51 (5.9%) 2/48 (4.2%) 0
SD 16/51 (31.4%) 9/48 (18.8%) 0
PD 32/51 (62.7%) 37/48 (77.1%) 9/9 (100%)
*

p-value by the Fisher’s exact test for PR vs SD/PD by treatment arms is 0.99

Abbreviations:

6m PFS: 6-month progression-free survival

CR: complete response

OS: overall survival

PD: progressive disease

PFS: progression free survival

PR: partial response

RR: radiographic response

SD: stable disease

The median follow-up time for the patients who were still alive at the time of the analysis was 15.0 months (2.5-26.4 months). The median survival time (MST) was 11.5 months (95% CI: 8.4-14.2 months) for the control arm and 7.5 months (95% CI: 6.8-10.1 months) for the experimental arm, with an HR=1.46 (95% CI: 0.95-2.27; p-value=0.09) (Figure 2A). The median PFS time was 4.8 months (95% CI: 3.8-7.1 months) for the control arm and 4.2 months (95% CI: 3.7-5.6 months) for the experimental arm, with an HR=1.51 (95% CI: 1.02-2.24; p-value=0.04) (Figure 2B). After adjusting for the stratification factors and other patient pretreatment characteristics, the adjusted HR of the treatment effect on OS was 1.46 (95% CI: 0.94-2.26) with a p-value=0.09 and that on PFS was 1.50 (95% CI: 1.01-2.23) with a p-value=0.05.

Figure 2.

Figure 2.

Figure 2.

Overall survival (A) and progression-free survival (B) by treatment arm.

Table 2 also contains the best RR by RANO criteria. Overall, 99 (86.1%) patients were evaluable. For the 51 patients in the control arm, 3 (5.9%) achieved a partial response (PR). For the 48 patients in the experimental arm, 2 (4.2%) achieved PR. No patient in either treatment arm achieved a complete response (CR). No significant difference was found between the two treatment arms.

Safety

Information for adverse events (AEs) without regard to attribution is presented by treatment arm for all the randomized and eligible patients who received protocol treatment (Supplementary Table 1). Although the proportion of patients with grade 3+ AEs was numerically higher for the control arm, 80.7% versus 64.9% for the experimental arm, this difference did not reach statistical significance (p-value=0.06). Similarly, the proportion of patients with grade 3+ AEs attributed to study treatment (36.8% of patients in the control arm vs. 35.1% in the experimental arm) was not significantly different (p-value=0.85). Overall, there were 2 patients (3.5%) in the control arm, and 5 (8.8%) in the experimental arm with reported grade 5 AEs, all of which were deemed unrelated to protocol treatment.

Cross-Over Study

A total of 25 patients crossed over from the control arm to receive open-label trebananib and bevacizumab after disease progression (Table 2). Twenty-two (88.0%) of them were evaluable for 6m PFS, and only 1 (4.5%) was progression-free at 6 months after initial disease progression on bevacizumab. Nine (36.0%) patients were evaluable for the best RR, all with progressive disease (PD) based on the RANO criteria. MST from the date of initial progression was 4.9 months (95% CI: 3.8-8.4 months), and median PFS was 2.3 months (95% CI: 2.1-3.2 months). No grade 5 AE was reported in the cross-over cohort, and 1 (4.0%) patient experienced grade 4 sepsis.

Pharmacokinetics

Four of six patients in cohort 1 and 17 of 25 in the cross-over arm participated in the optional PK and anti-trebananib antibody studies. The peak and trough concentrations of trebananib on Days 1 and 8 of Cycles 2 were comparable to those from previous studies.20 Minimal variability of trebananib concentrations were observed between Days 1 and 8 of cycle 2, suggesting the steady state pharmacokinetics were reached.

Discussion

The combination of trebananib and bevacizumab did not significantly improve the 6m PFS rate for patients with recurrent GBM in first or second relapse as compared to bevacizumab alone. Indeed, median PFS was longer in the control arm than the experimental arm. In comparison to other randomized studies comparing bevacizumab alone with in combination with another systemic agent (such as carboplatin,21 dasatinib,22 or onartuzumab23) in recurrent GBM, our study similarly fails to demonstrate a survival advantage to combination therapy. We also examined the benefit of adding trebananib to bevacizumab in patients who progressed on bevacizumab monotherapy. Survival times were similar to prior studies examining the role of combining bevacizumab with a systemic agent after progression on bevacizumab.1

The best RR was similar for the two treatment arms (around 5.0%), but lower than the historical benchmarks from prior bevacizumab studies (35% in Kreisl et al.1 and 46.4-65.8% in Friedman et al.2). This is partly explained by differences in RR assessment methods, with the historical benchmark studies utilizing Macdonald criteria24 and our study utilizing RANO criteria18. When Friedman et al. was reassessed by independent reviewers using RANO criteria, RR dropped to 33.1%.25 The even lower RR seen in our study could reflect a heightened understanding that decreased contrast enhancement on bevacizumab may not reflect true anti-tumor responses.18 In addition, RR assessment was performed by the local investigator, some of whom were not neuro-oncologists or neuro-radiologists and may have been more apt to call progression earlier than investigators in prior benchmark studies.

The MST of 11.5 months in the control arm was numerically but not statistically significantly longer than the experimental arm (7.5 months). Although PFS was numerically similar (4.8 months in the control arm versus 4.2 months in the experimental arm), PFS was significantly longer in the control arm (p=0.04). Except for the protocol specified salvage treatment (placebo arm participants crossing-over to experimental arm) and non-protocol surgery (6 patients who progressed on the placebo arm underwent non-protocol surgery as compared with 0 on the experimental arm), there was no significant difference in reported non-protocol treatments given after disease progression between the two arms (Supplementary Table 2). When comparing patients in the control arm that crossed-over (N=25) to receive the experimental treatment versus those who did not cross-over (N=33), the patients who did not cross-over lived numerically longer (with a MST of 12.1 months versus 9.9 months). These data suggest that adding trebananib to bevacizumab could be detrimental compared to bevacizumab alone. In a separate study of trebananib with or without bevacizumab, there was no clear benefit from either trebananib alone or in combination with bevacizumab.26 In the trebananib + bevacizumab arm of that study, outcomes were similar to our study with 6m PFS 24.3%, median OS 9.5 months, and median PFS 3.6 months.

It is unclear why patients receiving bevacizumab alone fared better than patients receiving bevacizumab + trebananib. One potential reason relates to the complex interactions between Ang1 and Ang2. Ang2 can act as a Tie2 antagonist or partial agonist depending on cellular context.27 Therefore, depending on the balance of Ang1 and Ang2, targeting the Ang-Tie2 pathway has the potential to promote or inhibit tumor growth.27 Several studies examining the effects of Ang1 or Ang2 blockade suggest that Ang2 may play a more important role in tumor angiogenesis.27 In addition, tumor models of several cancers suggest that Ang1 overexpression reduces tumor growth.28 It is possible that the Ang1 blocking effects of trebananib may somehow counteract or negate the anti-tumor effects of Ang2-VEGF blockade.

Another possible explanation for the lack of benefit from combined therapy is upregulation of alternative proangiogenic pathways. Other mechanisms that have been implicated in angiogenesis resistance include upregulation of alternative proangiogenic pathways such as basic fibroblast growth factor (bFGF) pathway, hepatocyte growth factor/cMET pathway, and SDF-1α; recruitment of bone marrow-derived proangioenic cells; and increased pericyte coverage of tumor vessels leading to vessel co-option.29, 30 It is possible that co-inhibition of the VEGF and angiopoietin/TIE2 pathways is still insufficient to prevent angiogenesis escape.

In summary, the combination of bevacizumab and trebananib failed to demonstrate improvements in 6m PFS, PFS, OS, or RR compared to bevacizumab alone. Careful consideration should be given to future trials of angiopoietin inhibitors in combination with bevacizumab given the possible detrimental effects of Ang1 in this study. Dual inhibitors of Ang2 and VEGFR are in development and may have therapeutic potential for glioblastoma.

Supplementary Material

Supp TableS1-2

Acknowledgments:

We would like to thank the NRG staff, particularly Minhee Won, Suzanne Baldwin, and Kathryn Okrent, for their help with this study.

Funding: This project supported by U10CA180868 (NRG Oncology Operations), U10CA180822 (NRG Oncology SDMC), UG1CA189867 (NCORP) from the National Cancer Institute (NCI) and Amgen

Conflicts of Interest:

Aldape, Battiste, Chmura, Connelly, deGroot Gerstner, Kim, Merrell, Pan, Raizer, Robins, Villano, Wagle, Wendland, nothing to disclose. Lee reports consultant Eli Lilly, royalties from Up to Date, Inc. Mehta reports personal fees Insys, Remedy, IBA, Varian, Celgene, Abbvie, Astra-Zeneca, Tocagen, Blue Earth Diagnostics, Board of Directors Oncoceutics. Reardon reports grants Acerta Pharmaceuticals, Incyte, Midatech, Omniox, Tragara, grants/personal fees Agenus, Celldex, EMD Serono, Inovio, personal fees Abbvie, Advantagene, Amgen, Bayer, Bristol-Myers Squibb, DelMar, Genentech/Roche, Merck, Merck KGaA, Monteris, Novocure, Oncorus, Oxigene, Regeneron, Stemline, Taiho Oncology. Dr. Wen reports Grant/Research/Clinical Trial Support Lilly USA, Agios, Astra Zeneca, Beigene, Eli Lily, ImmunocellularTherapeutics, Kazia, Kadmon, Karyopharm, Merck, Novartis, Oncoceutics and Vascular Biogenics. Speaker’s Bureau Merck. Consultant Roche, Taiho Oncology, Novartis, and Agios Pharmaceuticals. Advisory Boards Merck, Puma, Abbvie, Astra Zeneca, Eli Lilly, Genentech/Roche, GW Pharmaceuticals, Immunomic Therapeutics, Kadmon, Vascular Biogenics, Ziopharm, Taiho Oncology. DSMB Monteris, Tocagen. Zhang employee of Jazz Pharmaceuticals December 2018.

References

  • 1.Kreisl TN, Kim L, Moore K, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol. 2009;27: 740–745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009;27: 4733–4740. [DOI] [PubMed] [Google Scholar]
  • 3.Chinot OL, Nishikawa R, Mason W, et al. Upfront bevacizumab may extend survival for glioblastoma patients who do not receive second-line therapy: an exploratory analysis of AVAglio. Neuro Oncol. 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Gilbert MR, Sulman EP, Mehta MP. Bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370: 2048–2049. [DOI] [PubMed] [Google Scholar]
  • 5.Norden AD, Young GS, Setayesh K, et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70: 779–787. [DOI] [PubMed] [Google Scholar]
  • 6.Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008;8: 592–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kerbel RS. Tumor angiogenesis. N Engl J Med. 2008;358: 2039–2049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Batchelor TT, Duda DG, di Tomaso E, et al. Phase II study of cediranib, an oral pan-vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma. J Clin Oncol. 2010;28: 2817–2823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Batchelor TT, Sorensen AG, di Tomaso E, et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007;11: 83–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kloepper J, Riedemann L, Amoozgar Z, et al. Ang-2/VEGF bispecific antibody reprograms macrophages and resident microglia to anti-tumor phenotype and prolongs glioblastoma survival. Proc Natl Acad Sci U S A. 2016;113: 4476–4481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Peterson TE, Kirkpatrick ND, Huang Y, et al. Dual inhibition of Ang-2 and VEGF receptors normalizes tumor vasculature and prolongs survival in glioblastoma by altering macrophages. Proc Natl Acad Sci U S A. 2016;113: 4470–4475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Iwamoto FM, Abrey LE, Beal K, et al. Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma. Neurology. 2009;73: 1200–1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Chamberlain MC. Radiographic patterns of relapse in glioblastoma. J Neurooncol (in press). 2010. [DOI] [PubMed] [Google Scholar]
  • 14.Guo P, Imanishi Y, Cackowski FC, et al. Up-regulation of angiopoietin-2, matrix metalloprotease-2, membrane type 1 metalloprotease, and laminin 5 gamma 2 correlates with the invasiveness of human glioma. Am J Pathol. 2005;166: 877–890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hu B, Jarzynka MJ, Guo P, Imanishi Y, Schlaepfer DD, Cheng SY. Angiopoietin 2 induces glioma cell invasion by stimulating matrix metalloprotease 2 expression through the alphavbeta1 integrin and focal adhesion kinase signaling pathway. Cancer Res. 2006;66: 775–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Coxon A, Bready J, Min H, et al. Context-dependent role of angiopoietin-1 inhibition in the suppression of angiogenesis and tumor growth: implications for AMG 386, an angiopoietin-1/2-neutralizing peptibody. Mol Cancer Ther. 2010;9: 2641–2651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zelen M The randomization and stratification of patients to clinical trials. J Chronic Dis. 1974;27: 365–375. [DOI] [PubMed] [Google Scholar]
  • 18.Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010;28: 1963–1972. [DOI] [PubMed] [Google Scholar]
  • 19.Cohen MH, Shen YL, Keegan P, Pazdur R. FDA drug approval summary: bevacizumab (Avastin) as treatment of recurrent glioblastoma multiforme. Oncologist. 2009;14: 1131–1138. [DOI] [PubMed] [Google Scholar]
  • 20.Lu JF, Rasmussen E, Karlan BY, et al. Exposure-response relationship of AMG 386 in combination with weekly paclitaxel in recurrent ovarian cancer and its implication for dose selection. Cancer Chemother Pharmacol. 2012;69: 1135–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Field KM, Simes J, Nowak AK, et al. Randomized phase 2 study of carboplatin and bevacizumab in recurrent glioblastoma. Neuro Oncol. 2015;17: 1504–1513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Galanis E, Anderson SK, Anastasiadis P, et al. NCCTG N0872 (Alliance): A randomized placebo-controlled phase II trial of bevacizumab plus dasatinib in patients with recurrent glioblastoma (GBM). ASCO Meeting Abstracts. 2015;33: 2004. [Google Scholar]
  • 23.Cloughesy TF, Finocchiaro G, Belda C, et al. Onartuzumab plus bevacizumab versus placebo plus bevacizumab in recurrent glioblastoma (GBM): HGF and MGMT biomarker data. ASCO Meeting Abstracts. 2015;33: 2015. [Google Scholar]
  • 24.Macdonald DR, Cascino TL, Schold SC Jr., Cairncross JG. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8: 1277–1280. [DOI] [PubMed] [Google Scholar]
  • 25.Huang RY, Rahman R, Ballman KV, et al. The Impact of T2/FLAIR Evaluation per RANO Criteria on Response Assessment of Recurrent Glioblastoma Patients Treated with Bevacizumab. Clin Cancer Res. 2016;22: 575–581. [DOI] [PubMed] [Google Scholar]
  • 26.Reardon DA, Lassman AB, Schiff D, et al. Phase 2 and biomarker study of trebananib, an angiopoietin-blocking peptibody, with and without bevacizumab for patients with recurrent glioblastoma. Cancer. 2018;124: 1438–1448. [DOI] [PubMed] [Google Scholar]
  • 27.Cascone T, Heymach JV. Targeting the angiopoietin/Tie2 pathway: cutting tumor vessels with a double-edged sword? J Clin Oncol. 2012;30: 441–444. [DOI] [PubMed] [Google Scholar]
  • 28.Augustin HG, Koh GY, Thurston G, Alitalo K. Control of vascular morphogenesis and homeostasis through the angiopoietin-Tie system. Nat Rev Mol Cell Biol. 2009;10: 165–177. [DOI] [PubMed] [Google Scholar]
  • 29.Lu-Emerson C, Duda DG, Emblem KE, et al. Lessons from anti-vascular endothelial growth factor and anti-vascular endothelial growth factor receptor trials in patients with glioblastoma. J Clin Oncol. 2015;33: 1197–1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gacche RN. Compensatory angiogenesis and tumor refractoriness. Oncogenesis. 2015;4: e153. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp TableS1-2

RESOURCES