Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2025 Apr 25;15(4):1874–1901. doi: 10.62347/RNUE7193

Treatment mechanism and research progress of bevacizumab for glioblastoma

Xinliang Liu 1,2,*, Zhigang Chen 3,*, Pengwei Yan 2, Tao Yang 4, Dan Zong 2, Wenjie Guo 2, Xia He 2,5
PMCID: PMC12070100  PMID: 40371151

Abstract

Hypervascularization is a notable pathological hallmark of glioblastoma (GBM). Bevacizumab (Bev) remains the sole antiangiogenic agent approved by the U.S. Food and Drug Administration (FDA) for GBM treatment. The approval for this indication was supported by several phase II studies demonstrating that Bev significantly improved progression-free survival and the best imaging response in patients with recurrent GBM. Three large phase III randomized controlled trials reported that Bev did not significantly extend overall survival (OS). Nevertheless, Bev has been shown to delay the deterioration of patients’ quality of life by postponing tumor progression. This review synthesizes findings from recent investigations exploring Bev in combination with targeted therapies, immunotherapy, or reirradiation. Additionally, this review discusses dosing regimens, administration, treatment failure patterns, third-line therapeutic applications, and prognostic markers of Bev. By synthesizing current evidence, this review aims to inform clinical decision-making for neuro-oncology clinicians.

Keywords: Bevacizumab, glioblastoma, chemoradiotherapy, targeted therapy, immunotherapy, dosage and route of administration, patterns of treatment failure, third-line therapy, prognostic markers

Introduction

Glioblastoma (GBM) is characterized by elevated incidence and recurrence rates alongside substantial mortality. GBM accounts for 50.9% of adult malignant central nervous system tumors, exhibiting an increasing prevalence with a 6-month progression-free survival (PFS) rate of 15% and a 5-year survival rate of 6.7% [1]. Furthermore, GBM patients demonstrate severe deterioration in health-related quality of life (QoL), characterized by progressive neurocognitive decline attributed to advanced age (median age: 65), rapid disease progression, or toxicity from treatment drugs. These factors underscore critical unmet therapeutic demands for GBM.

Bevacizumab (Bev; Avastin®), a monoclonal IgG1 antibody that targets vascular endothelial growth factor (VEGF), has opened a new chapter in GBM therapeutics. Building on efficacy observed in metastatic colorectal cancer, Stark-Vance et al. pioneered the application of Bev for glioma therapy in 2005 [2,3]. A study enrolling 21 patients with recurrent malignant glioma (10/21 with high-grade glioma) demonstrated a 42.9% objective response rate (ORR) with Bev-irinotecan (Iri) combination therapy [2]. Phase II clinical studies subsequently validated Bev’s efficacy as monotherapy for recurrent GBM (rGBM), yielding ORR and 6-month progression-free survival (PFS6) rates of 28.2%-57% and 29%-46%, respectively [4-6]. This evidence led to the U.S. FDA’s accelerated approval of Bev for rGBM in 2009 [7].

The dual objectives of GBM therapy are survival prolongation and preservation or enhancement of QoL. GBM management has transitioned from traditional surgical resection combined with the Stupp protocol to multimodal approaches incorporating tumor-treating fields, targeted agents, and immunotherapies. Bev-based clinical strategies for rGBM initially focused on monotherapy and chemotherapeutic combinations (e.g., alkylating agents, Iri, and carboplatin), whereas recent investigations explore its integration into frontline settings, including administration during or prior to concurrent chemoradiotherapy or as a presurgical intervention. Three pivotal phase III randomized controlled trials (RCTs) failed to establish significant overall survival (OS) benefits with Bev [8-10]. However, Bev may mitigate QoL deterioration through delayed tumor progression [5,11,12]. This review critically evaluates emerging combinatorial strategies integrating Bev with targeted therapies, immunotherapies, or reirradiation. Furthermore, this review discusses dosing regimens, administration routes, failure patterns, third-line applications, and prognostic markers of bev, which have great value in clinical practice (Figure 1).

Figure 1.

Figure 1

Graphical abstract of the review article. Hypoxia-induced dysregulation of angiogenic factors promotes pathological vascular remodeling and the formation of an immunosuppressive TME in GBM. Bev, a VEGF-A-targeting monoclonal antibody, is utilized in clinical practice as part of combination therapies to disrupt these pathways. This review further examines four critical dimensions of Bev in clinical practice. SCLGC, stem cell-like glioma cell; VEGF, vascular endothelial growth factor; bFGF, basic fibroblast growth factor; TGF-β, transforming growth factor-β; TME, tumor microenvironment; TSP-1, thrombin-sensitive protein-1; INF-alpha, interferon-alpha; PDGF, platelet-derived growth factor; EGF, epidermal growth factor; PlGF, placental growth factor; VEGFR, VEGF receptor. Created in BioRender. Yang, T. (2025) https://BioRender.com/avq3e42.

This synthesis of pivotal Bev-associated clinical trials in GBM seeks to inform evidence-based decision-making for neuro-oncology practitioners.

Mechanism

Hypervascularization activity, characterized by irregular vascular morphology, inefficient transport, and pericyte deficiency, represents a critical pathological hallmark of GBM [13]. Hypoxia-induced dysregulation of angiogenesis-related factors in the tumor microenvironment (TME) manifests as upregulated pro-angiogenic factors - including vascular endothelial growth factors [VEGFs], basic fibroblast growth factors [bFGFs], and transforming growth factor-β [TGF-β]) - coupled with diminished expression of anti-angiogenic factors such as thrombin-sensitive protein-1 (TSP-1) and interferon (INF)-alpha, thereby perpetuating the activation of the tumor angiogenesis switch [14].

VEGF plays a pivotal role in both physiological and pathophysiological processes: it maintains endothelial cell survival, proliferation, and migration; promotes glioma cell growth via autocrine signaling; enhances vascular permeability, also known as vascular permeability factor; and contributes to the immunosuppressive TME [15,16].

Bev exerts therapeutic effects by neutralizing VEGFs (primarily VEGF-A), thereby blocking its interaction with receptors (Flt-1 and KDR) and inhibiting downstream signaling [17]. Bev mainly targets genomically stable endothelial cells, indirectly suppressing tumor progression rather than directly killing tumor cells, which confers a lower propensity for drug resistance. Furthermore, Bev disrupts the proangiogenic effect of stem cell-like glioma cells (SCLGCs), inhibiting transplanted tumor growth in experimental models [18].

Both endothelial cells and glioma cells can express VEGF and its receptors. In GBM, VEGF expression is regulated by two primary pathways: hypoxia-dependent mechanisms mediated by hypoxia-inducible factor-1 (HIF-1) [19] and hypoxia-independent mechanisms involving epidermal growth factor receptor (EGFR) activation [20,21]. SCLGCs continuously secrete substantial VEGF in vitro, amplifying the proangiogenic function of endothelial cells - a process exacerbated by hypoxia [18,22]. Notably, VEGFR expression has been detected in GBM cells. Joensuu et al. reported that 39% of GBM patients exhibit amplification of VEGFR2 [23]. Quantitative analyses across 12 brain tumor types revealed significantly elevated VEGFR levels in GBM [24]. Preclinical studies confirm that VEGF-VEGFR pathway blockade suppresses glioma cell growth in immunodeficient rat models [25].

Progressive and recurrent glioblastoma (rGBM)

Bev monotherapy

Bev has been recommended as the preferred treatment for rGBM by the National Comprehensive Cancer Network (NCCN) guidelines in the U.S. [26]. Table 1 summarizes key clinical trial outcomes. The noncomparative phase II AVAREG trial evaluated Bev against fotemustine (FTM) in rGBM patients, randomizing participants at a 2:1 ratio (Bev vs. FTM), with the primary endpoint being the 6-month OS rate [12]. No significant differences were observed in the 6-month OS rate or median OS (mOS) between the two groups [12]. A similar conclusion was reached in the Japanese JO22506 study [27].

Table 1.

Bev alone or in combination with cytotoxic chemotherapy for the treatment of rGBM

Ref. Study Design No. of patients Study arms Bev Dose (mg/kg) ORR (%) mOS (months) mPFS (months) PFS6 (%)
Stark-Vance 2005 [2] NA GBM: 11 Bev + Iri 5 43 NA NA NA
Other: 10
Vredenburgh 2007 [6] II 35 Bev + Iri 10 57 9.2 4.2 46
Bev + Iri 15
Bokstein 2008 [97] NA GBM: 17 Bev + Iri 5 47.3 7 4.2 25
2005-2007 AO: 2
AOA: 1
Kreisl 2009 NCI 06-C-0064E [5] II, comparative 48 Bev→Bev + Iri 10 Levin: 71 7.75 4 29
2006-2007 Macdonald: 35
Friedman 2009 BRAIN [4] II, multicenter, open-label, randomized, noncomparative 167 Bev + Iri 10 28.2 8.7 5.6 NA
2006-2007 Bev 9.2 4.2
Nghiemphu 2009 [11] Retrospective 44 Bev 5 NA 9.01* 4.25* 41
2005-2006 79 Other 6.11 1.82 18
Raizer 2010 [96] II GBM: 50 Bev 15 24.5 6.5 NA 25
Taal 2014 BELOB [36] II, multicenter, randomized 47 LOM 10 5 8.0 1.0 13
2009-2011 51 LOM90 + Bev 34 12.0 4.0 41
47 Bev 38 8.0 3.0 16
Field 2015 CABARET [38] II, multicenter, randomized, open-label 60 Bev + Carboplatin 10 14 6.9 3.5 15
2010-2012 62 Bev 6 7.5 3.5 18
Brandes 2016 AVAREG [12] II, Noncomparative, multicenter, randomized 91 Bev 10 29 7.3 3.38 19.6
2011-2012 FTM 9 8.7 3.45 10.7
Wick 2017 EORTC 26101 [10] III 288 LOM + Bev 10 13.9 9.1 4.2 28.4
2011-2014 149 LOM 41.5 8.6 1.5 16.8
Gilbert 2017 RTOG 0625 [146] II, randomized 60 Bev + dose-dense TMZ 10 19 9.4 4.7 39
2007 57 Bev + Iri 28 7.7 4.1 38.6
Desjardins 2019 [147] Retrospective, single-institution, real-world study 74 Bev 10 NA 11.1 6.4 24.3
2009-2012
Cloughesy 2020 TOCA 5 [148] II/III, open-label, multicenter, randomized 201 Toca 511 + Toca FC 10 NA 11.10 NA NA
rHGG 202 SOC (Bev/TMZ/LOM) 12.22
2015-2018
Detti 2021 [149] Retrospective, rHGG 92 (GBM: 71) Bev 10/15 36.1 9 6.9 58
2009-2019
Friedman 2023 [33] II, multicenter, open-label, Noncomparative Bev 10 28.2 9.2 4.2 42.6
Bev + Iri 37.8 8.7 5.6 50.3
Smolenschi 2023 [150] Retrospective, real-world study, single-institution 202 Bev 10/15 42 23.7 6.8 NA
rGBM
2006-2016
Witte 2024 [151] Retrospective 134 HCQ + BEV + aRCT NA NA 23.92 NA NA
rGBM BEV + aRCT 9.63*
2006-2016
Lee 2024 [152] Nationwide population-based study 450 Bev NA NA 22.60 NA NA
rGBM 396 Bev + Iri 20.44
2008-2021

PFS12;

AOA, Anaplastic oligoastrocytoma; AO, Anaplastic oligodendroglioma; PFS6, 6-month progression-free survival; PFS, progression-free survival; OS, overall survival; GBM, glioblastoma multiforme; q, every; TMZ, temozolomide; Carbo, carboplatin; LOM/CNNU, carmustine; FTM, fotemustine; NA, not available; SOC, standard of care; Toca 511, vocimagene amiretrorepvec; Toca FC, flucytosine; HCQ, chloroquine; aRCT, adjuvant-radiochemotherapy;

*

P < 0.05.

However, the use of Bev in rGBM remains contentious. Unlike the FDA, the European Medicines Agency has not approved Bev for rGBM treatment [28].

First, although Bev improves the ORR and median PFS (mPFS) in rGBM patients, it fails to extend OS [4-6]. This discrepancy suggests that surrogate endpoint improvements do not translate into an OS benefit. Ballman et al. reported a 90% concordance between PFS6 and 12-month OS in their analysis of phase II trials [29]. However, their analysis did not include clinical trials related to Bev, leaving its generalizability to Bev-treated cohorts uncertain.

Second, Bev complicates neuro-oncological response assessment. Traditional imaging assessments rely mainly on contrast-enhanced MRI based on the MacDonald criteria, but these can be influenced by various factors, such as radiation damage, MRI equipment parameters, and steroid use [30]. Bev’s ability to stabilize the blood-brain barrier (BBB) and reduce contrast leakage may interfere with comparisons of pre- and posttreatment enhanced MR images. As a result, a reduction in contrast enhancement may mask an actual increase in tumor burden, inducing “pseudoresponse”. To address this issue, the Response Assessment in Neuro-Oncology (RANO) criteria were developed, which incorporate T2-weighted imaging/fluid-attenuated inversion recovery (T2/FLAIR) to evaluate nonenhancing lesions for a more accurate and comprehensive evaluation of patient efficacy [31]. Additionally, the timing of assessments is crucial. Early MRI scans post-Bev initiation (e.g., within 24 hours) often show reduced enhancement unrelated to antitumor efficacy [5]. Interestingly, patients with “partial response” on day 4 or 28 scans exhibit prolonged PFS [5]. These patients may have greater sensitivity to Bev treatment.

Finally, several clinical trials have design flaws. The NCI 06-C-0064E study relied on historical controls for their interpretation [5], while the AVF3708g study employed a noncomparative design despite randomization [4].

In conclusion, despite the aforementioned controversies, two points are clear: First, owing to differing mechanisms of action, the two drugs exhibit different toxicity profiles, with Bev primarily causing adverse events such as bleeding, thrombosis, hypertension, and proteinuria, whereas alkylating agents (such as FTM, lomustine [LOM], and TMZ) mainly lead to hematological toxicity, such as thrombocytopenia and neutropenia [12,32]. A recent study showed that thrombocytopenia can lead to dose adjustments or discontinuation of CCNU, thereby decreasing survival in rGBM patients [32]. Second, Bev can significantly improve QoL and effectively decrease the use of corticosteroids by reducing BBB permeability and alleviating cerebral edema [5,11,12,33]. Therefore, in a context where treatment options are limited, Bev may provide an alternative treatment option for rGBM patients.

With chemotherapy (CT)

Theoretically, Bev combined with CT may exhibit a synergistic effect [34]. Bev induces vascular normalization, which enhances drug distribution uniformity and increases intratumoral drug perfusion. Furthermore, unlike TMZ, the first-line CT agent for GBM, CT agents such as Iri and carboplatin, do not have cross-resistance and demonstrate proven antitumor efficacy in other malignancies [35].

Main clinical trial outcomes are summarized in Table 1. In 2014, the first randomized controlled trial of Bev for rGBM - the phase II BELOB study - was conducted by Taal et al., in which 145 rGBM patients were randomized to three arms: CCNU monotherapy, Bev monotherapy, or CCNU+Bev combination therapy [36]. The 9-month OS rates were reported as 43%, 38%, and 59%, respectively, with the combination group showing superior survival compared to the Bev alone group [36]. However, Bev monotherapy achieved a higher ORR (38% vs. 5%) and longer mPFS (3 vs. 1 month) than CCNU monotherapy. No crossover Bev administration occurred among the 3 groups [36].

To validate the BELOB findings, the phase III EORTC 26101 trial, led by Wick et al., enrolled 437 rGBM patients randomized 2:1 to receive Bev+CCNU or CCNU alone [10]. Although combination therapy significantly prolonged PFS (4.2 vs. 1.5 months), no mOS benefit was observed. The addition of Bev exhibited higher rates of grade 3-5 adverse events (AEs) (63.6% vs. 38.1%), and it did not improve patients’ neurocognitive function; in fact, their overall health status scores were worse [10]. Consequently, the BELOB results were not replicated in this larger trial. Furthermore, a cost-utility analysis by Chen et al. further concluded that the Bev+CCNU combination regimen lacked cost effectiveness in the EORTC 26101 study [37].

The randomized phase II CABARET study compared Bev+carboplatin to Bev monotherapy in rGBM, revealing no significant differences in mPFS, mOS, or ORR [38]. Notably, the ORR (6%-14%) was markedly lower than historical controls, potentially attributable to the mRANO criteria (incorporating T2/FLAIR assessments) and approximately one-third of the enrolled patients with ≥2 prior recurrences [38].

In summary, while Bev+CT combinations prolong PFS, they concurrently increase toxicity without conferring OS benefits. Large phase III trials remain scarce due to the low incidence of GBM. Additionally, heterogeneity in trial design, eligibility criteria, prior therapies, and response assessment protocols limits cross-trial comparability.

With targeted therapy

The limited efficacy of Bev monotherapy in rGBM may be attributed to redundant angiogenesis signaling pathways or compensatory dysregulation of downstream signaling molecules of VEGF/VEGFR signaling molecules, such as epidermal growth factor receptor (EGFR) overexpression or gene amplification (observed in ~50% of cases), platelet-derived growth factor receptor (PDGFR) overexpression (75%), and aberrant mesenchymal-epithelial transition factor pathways activity [39,40]. Consequently, recent therapeutic strategies have focused on combining Bev with multiple targeted agents to overcome resistance.

Several phase I/II trials have explored the effects of Bev combined with targeted therapies for rGBM, including three randomized controlled phase II studies (see Table 2). Among these studies, only one study demonstrated a survival benefit [41]. This single-arm phase II study reported that TVB-2640 (a fatty acid synthase inhibitor) combined with Bev significantly improved mPFS (4.6 vs. 3 months) and PFS6 (31.4% vs. 16%) compared to Bev monotherapy in the historical BELOB cohort [41]. However, no significant OS advantage was observed.

Table 2.

Bev in combination with targeted therapy for the treatment of rGBM

Ref. Study Design NO. patients Study arms ORR (%) mOS (months) mPFS (months) PFS6 (%)
Puduvalli 2020 [153] II, multicenter, randomized, bayesian adaptiv 49 Vorinostat + Bev NA 7.8 3.7 25
41 Bev 9.3 3.9 28
Lee 2020 NRG/RTOG 1122 [154] II, randomized, double-blinded, placebo-controlled study 57 Trebananib + Bev 4.2 7.5 4.8 22.6
58 Trebananib + Pla 5.9 11.5 4.2* 41.4
Kelly 2023 [41] II, single-institution, open-label 25 TVB-2640 + Bev 56 8.9 4.6 31.4►►,*
McCrea 2021 [155] I 13 Cetuximab + Bev 30 7.2 NA NA
2013-2018
Brenner 2021 [136] II, open-label, Single group 33 Evofosfamide + Bev 9 4.6 53 days 31
2015-2017
Galanis 2022 N1174 [156] I/II 52 TRC105 + Bev 13 9.7 2.9 25
49 Bev 16 7.4 3.2 30
Zhao 2024 [42] II, Single group, rHGG 18 Anlotinib 28 15 4.2 NA
2020-2022 7 Anlotinib + Bev 43 9.8 8
Cloughesy 2017 GO27819 [39] II, double-blinded, randomized controlled, multicenter 64 Onartuzumab + Bev 22.2 8.8 3.9 29.7
65 Pla + Bev 23.7 12.6 2.9 26.2
Cardona 2021 [157] Retrospective, EGFR amplification and EGFRvIII mutation rGBM 15 osimertinib + Bev 13.3 9.0 5.1 46.7

Note: Pla, placebo;

4-month PFS rate;

►►

control from the Bev monotherapy group in the BELOB study;

When peritumoral edema develops, patients will receive interim treatment with Bev;

TRC105, anti-CD105 antibody;

*

P < 0.05.

Additionally, a phase II trial involving 25 recurrent high-grade glioma (rHGG) patients evaluated anlotinib (a multitarget tyrosine kinase inhibitor targeting VEGFR and PDGFR, etc.) alone or combined with Bev (initiated upon peritumoral edema occurred) [42]. The combination failed to enhance efficacy but significantly increased treatment-related adverse events (TRAEs) [42].

In summary, as molecular profiling of GBM advances, targeted therapy has emerged as a hot topic of intensive research. While double-blind phase II RCTs provide level II evidence [43], no published phase III studies have yet validated the effectiveness of Bev-targeted therapy combinations. Thus, the use of Bev combined with targeted agents for rGBM remains investigational and is not recommended outside clinical trials.

With immunotherapy (IT)

Patients with rGBM often derive limited benefit from IT alone, partially due in part to the VEGF-driven immunosuppressive TME [44]. Bev can reduce the use of corticosteroids, which decrease effector immune cell levels [45]. In addition, Bev synergistically enhances the antitumor activity of IT, as evidenced by preclinical and clinical data from other cancer types [46].

Currently, the application of Bev-IT combinations in rGBM remains exploratory. The findings from the main studies are compiled in Table 3. The phase III CheckMate 143 study compared nivolumab (Nivo, a PD-1 monoclonal antibody) monotherapy to Bev monotherapy in rGBM, revealing superior PFS and ORR for Bev, though OS and serious adverse events rates did not differ significantly [47]. In a phase II study Nayak et al., 80 rGBM patients randomized to pembrolizumab (Pemb, a PD-1 monoclonal antibody) ± Bev demonstrated improved mPFS (4.1 vs. 1.4 months) and ORR (20% vs. 0%) with combination therapy, yet no OS advantage (8.8 vs. 10.3 months) [48]. Similarly, durvalumab (anti-PD-L1) ± Bev failed to meet efficacy expectations in another phase II trial (mOS: 6.7-9.3 months) [45].

Table 3.

Summary of published clinical studies of Bev in combination with immunotherapy for the treatment of rGBM

Ref. Study Design NO. patients Study arms ORR (%) mOS (mos) mPFS (mos) PFS6 (%)
Wang 2024 [51] IIa, single center, single-arm, open-label 32 Tislelizumab + low-dose Bev 25 22.3 4.0 NA
2021-2023
Guo 2024 [50] II, single center, open-label 32 Tislelizumab + low-dose Bev 56.3 14.3 8.2 NA
2022-2023
Chiu 2023 [49] I, open-label, nonrandomized 5 Avelumab + Bev NA 13 3 16.5
2018-2019 7 LITT + avelumab + Bev 13.5 2.5
Bota 2022 [158] Prospective 21 SITOIGANAP + GM-CSF + cyclophosphamide + Bev + Nivo or Pemb NA 19.63 9.14 76.19
2018-2021
Sahebjam 2021 [72] I, single-arm, open-label 24 (Bev-naïve cohort) HFSRT + Pemb + Bev 83 13.4 7.92 66.67
2015-2019 8 (Bev-resistant cohort) 62 9.3 6.54
Nayak 2021 [48] II, randomized, multicenter, open-label, Bev-naïve 50 Pemb + Bev 20 8.8 4.1 26.0
2015-2016 30 Pemb 0 10.3 1.43 6.7
Ahluwalia 2021 [159] II, randomized 90 Nivo + standard-dose Bev (10 mg/kg) NA 41.1 NA NA
2018-2020 Nivo + low-dose Bev (3 mg/kg) 37.7
Brenner 2020 [54] I/II, 72 (all) VB-111 dose escalation 63 315 days 55 NA
rGBM VB-111 monotherapy 53 223 days* 60*
2011-2015 VB-111→(VB-111 + Bev) 21 414 days 90
VB-111 + Bev 20 141.5 days** 63
Cloughesy 2020 (GLOBE) [53] III, randomized 1:1 128 VB-111 + Bev 27.3 6.8 3.4 22.7
2015-2017 128 Bev 21.9 7.9 3.7 28.9
Reardon 2020 (CheckMate 143) [47] III, randomized 1:1 184 Nivo vs. Bev 7.8 9.8 1.5 15.7
2014-2015 185 23.1 10.0 3.5** 29.6
Nayak 2022 [45] II, multicenter, nonrandomized A: 40 (unmethylated MGMT promotor) Durvalumab + standard RT 10.3 15.1 4.6 41.1
2015-2017 B: 31 (bevacizumab naïve) Durvalumab 12.9 6.7 3.0 19.4
B2: 33 (bevacizumab naïve) Durvalumab + t standard-dose Bev 9.1 8.7 3.7 15.2
B3: 33 (bevacizumab naïve) Durvalumab + low-dose Bev 9.1 9.3 3.7 17.2
C: 22 (bevacizumab refractory) Durvalumab + t standard-dose Bev 0 4.5 1.9 0
Wang 2024 [51] IIa, single center 32 Tislelizumab + Bev 25 22.3 4 NA
rGBM
2021-2023

Note: LITT, laser interstitial thermal therapy; Nivo, nivolumab; Pemb, pembrolizumab; SITOIGANAP, tumor cells and lysates; RT, radiation therapy;

1-year OS rate;

*

P < 0.05;

**

P < 0.001;

Tislelizumab, an anti-PD-1 antibody.

Contrastingly, recent phase I/II studies have indicated that Bev combined with avelumab (a PD-L1 monoclonal antibody) or toripalimab (a PD-1 monoclonal antibody) can improve survival, with a median OS of 13-22.3 months [49-51]. Meanwhile, Yang et al. reported a case of rGBM with lung metastasis achieving 11-month PFS and 27-month OS. This patient received Bev combined with Pemb due to PD-L1 overexpression [52]. In addition to the limited number of case reports and differences in local drug penetration concentrations, further research is warranted to investigate whether extracranial metastatic sites and intracranial recurrence sites exhibit distinct TMEs and tumor cell biological characteristics [52].

Novel approaches, such as viruses or vaccines, have also been explored. The phase III GLOBE trial evaluated VB-111 (an anticancer viral therapy that induces endothelial cell apoptosis and activates antitumor immunity) ± Bev, showing no mOS improvement but increased grade 3-5 adverse events with combination therapy (67% vs. 40%) [53]. A study by Brenner et al. yielded similar results in the same year [54]. Additionally, Brenner et al. found a significant prolongation of survival in the subgroup of VB-111 monotherapy upon continued in combination with Bev after recurrence [54], suggesting timing-dependent immune activation. Similarly, neoadjuvant/adjuvant Pemb improves survival versus adjuvant-only regimens, potentially via enhanced tumor-specific T cell priming [55]. Reardon et al.’s phase II trial of rindopepimut (a vaccine targeting EGFRvIII) ± Bev reported comparable PFS6 but a higher 24-month OS rate than the control group did (20% vs. 3%) and corticosteroids discontinued rates (33% vs. 0%) in the combination arm [56].

In summary, the conclusions drawn from various studies differ, which may be related to the characteristics of the drugs studied, the baseline characteristics of the enrolled patients, and so on. Currently, there is no conclusive high-level evidence supporting the use of Bev combined with IT drugs in the treatment of rGBM patients. However, this combination therapy shows great promise and has become a hot area of current research (see Table 4).

Table 4.

Summary of ongoing clinical studies of Bev in combination with immunotherapy for the treatment of rGBM (data from https://clinicaltrials.gov)

NCT Study Start Enrollment Immunological drug Phase Interventions Primary endpoint
NCT04116658 2020-07-13 100 EO2401, Nivo I/II EO240 OS
EO2401 + Nivo
EO2401 + Nivo + Bev
NCT06061809 2024-08-07 20 N-803, PD-L1 t-haNK II N-803 + PD-L1 t-haNK + Bev TEAEs, SAEs
NCT04277221 2019-09-19 118 ADCTA III ADCTA vs. Bev OS
NCT06047379 2023-11-01 134 Ipi, Pemb and Nivo I/II NEO212 + Ipi MTD, PFS6
NEO212 + Pemb
NEO212 + Nivo
NEO212 + regorafenib NEO212 + carboplatin + paclitaxel
NEO212+ FOLFIRI + Bev
NEO212
NCT05502991 2022-12-11 60 Sintilimab II Sintilimab + Bev 12/18-month OS rate

Note: TEAEs, treatment-emergent adverse events; SAEs, serious adverse events; ADCTA, autologous dendritic cell/tumor antigen; NEO212, TMZ + perillyl alcohol; Ipi, ipilimumab; FOLFIRI, 5-fluorouracil + leucovorin + Iri.

With reirradiation

Reirradiation (re-RT) is an important therapeutic modality for rGBM. The mechanisms underlying the combination of Bev with re-RT are multifactorial. Preclinical studies have demonstrated that ionizing radiation markedly elevates VEGF levels in tumors, while Bev enhances intratumoral oxygenation and disrupts the vascular niche harboring cancer stem cells (CSCs) [57]. Clinical evidence further indicates that Bev mitigates the risk of radiation necrosis (RN) and cerebral edema [58].

When Bev is combined with re-RT techniques - including HSRT, SRS, and GK - mOS ranges from 10.1 to 13.9 months, and mPFS spans 5-11 months (Table 5). The NRG Oncology/RTOG1205 trial, the first multicenter phase II study to utilize modern RT techniques in rGBM, evaluated the efficacy of integrating re-RT with Bev [59]. Between 2012 and 2016, 170 patients were randomized 1:1 to Bev+re-RT or Bev monotherapy. No significant difference in mOS was observed (10.1 vs. 9.7 months); however, the combination group exhibited superior mPFS (7.1 vs. 3.8 months) and PFS6 rates (54.3% vs. 29.1%) [59]. Acute grade 3+ TRAEs occurred in only 4.8% of the combination group [59]. Notably, baseline imbalances were observed: the combination group included more patients with ≥2 more recurrences (25.6% vs. 13.1%), a poor prognostic factor, but also a higher proportion with O6-methylguanine DNA methyltransferase (MGMT) promoter methylation (20.9% vs. 14.3%), a favorable prognostic factor [59]. Prolonged PFS may delay declines in QoL and neurocognitive function [59]. Multivariate analysis identified Karnofsky performance status (KPS) - not age - as a significant OS prognosticator, supporting this regimen’s applicability to elderly patients with KPS scores ≥ 70%. A retrospective study also reported that SRS+Bev is a positive prognostic marker for HGG [60]. The ongoing phase II HSCK-005 trial (NCT05611645) is investigating whether HFSRT combined with Bev enhances PFS6 in rGBM.

Table 5.

Bev in combination with re-RT for the treatment of rGBM

Ref. Study design No. patients Treatment mOS (months) mPFS (months) PFS6 (%) Median GTV (cc) (Range) RT dose (Gy/Fractions)
Tsien 2023 [59] II, andomized 170 BEV + re-RT 10.1 7.1 54.3 18 (0.5-208) 35 Gy/10 f
2012-2016 BEV 9.7 3.8* 29.1*
She 2022 [160] Retrospective 26 Bev + re-RT 13.6 8.0 65.4 Median PTV size (ml) (range): 114.8 (11.9-360.1) 30-35 Gy/10 f (for the small GTV)
2019-2021 40-45 Gy/20-27 f (for the large GTV)
Arvold 2017 [65] Retrospective 67 re-RT 10.7 4.8 NA NA 30 Gy/5 f (36%), 35 Gy/10 f (21%), 40 Gy/15 f (15%), 18-20 Gy/1 f (15%)
2010-2014 117 Bev NA NA
Palmer 2018 [61] Retrospective 118 (GBM: 87; AA: 31): Bev → HFSRT 13.3 NA NA 44.8 cm3 35 Gy/10 f
2006-2013 50 FSRT → Bev 13.9 17.04 cm3
68
Zhang 2024 [161] Retrospective 19 Bev 6.5 5.0 NA 15.9 ± 11.5 NA
2012-2022 57 Bev + GK 11.5 7.7 7.6 ± 11.6
19 GK 7.9* 4.9* 13.2 ± 16.1
Mantica 2023 [162] II 16 Bev + SRS 11.73 NA 65.2 NA NA
2015-2017 SRS 8.74* 33.2*
Morris 2019 [163] Retrospective 45 Bev + GK 13.3 5.2 NA 2.2 cm3 (0.1-25.2 cm3) 17.0 Gy
2009-2015
Abbassy 2018 [164] I 9 Bev + SRS 13 7.5 NA 4.4 cm3 (2.1-8.8) 18 Gy
5.13 cm3 (4.38-6.47) 20 Gy
4.64 cm3 (2.54-6.62) 22 Gy

Note: HFSRT, hypofractionated stereotactic radiotherapy; SRS, stereotactic radiosurgery; GK, Gamma Knife;

*

P < 0.05.

Bev administration timing relative to re-RT - neoadjuvant (neoBev), concurrent (concBev), or adjuvant (adjBev) - has been explored. A large retrospective study by Palmer et al. found no survival difference in rHGG patients receiving Bev before or after re-RT [61]. Conversely, Cuneo et al. reported that adjBev post-SRS significantly improved mOS (5.2 vs. 2.1 months) and 1-year OS compared to no Bev [62]. A retrospective multicenter study of 482 rHGG patients (1997-2023), presented at the 2024 American Society of Clinical Oncology annual meeting, found adjBev unrelated to survival outcomes, whereas concBev correlated with poorer OS but lower neurotoxicity [63]. Thus, optimal Bev-re-RT sequencing remains undefined.

Consensus is similarly lacking on optimal re-RT target volume and dosing with Bev. In re-RT monotherapy, comparable survival was observed with 25 Gy/5 f and 35 Gy/5 f [64]. There was no difference in OS or PFS when comparing local re-RT to systemic treatment with Bev [65]. However, Kulinich et al. identified Bev as a survival enhancer in HFSRT-treated patients [66]. Recently, the GLIAA study prospectively compared 18F-fluoroethyltyrosine (FET) positron emission tomography (PET) and T1Gd MRI-guided re-RT in rGBM, revealing no differences in survival, local control rates, or RN rates [67]. Additionally, a large-scale retrospective study reported that large-volume re-RT (median PTV: 135 cm3) has been deemed feasible for refractory rGBM [68].

Radiation-related toxicity remains a key concern. As GBM typically recurs near surgical margins, re-RT target often overlap initial RT fields. RN - the most severe late radiation complication - is pathologically characterized by brain tissue necrosis. The exact mechanisms underlying RN are still unclear, but they may be related to the loss of pericytes and vascular smooth muscle cells [69]. Clinically, RN usually presents with the re-emergence of symptoms and a decline in neurological function, while imaging reveals enhancing lesions indistinguishable from tumor progression. A recent retrospective analysis revealed that apparent diffusion coefficient values exhibit significant discriminative capability [70]. Additionally, most clinical studies do not report the incidence of RN. The phase II CHROME trial protocol recently proposed chlorophyllin for RN management in diffuse gliomas [71], though its efficacy versus corticosteroids, Bev, or surgery warrants validation.

In summary, the ideal Bev-re-RT regimen remains uncertain, with most evidence derived from retrospective studies prone to selection bias. Incorporating IT into the Bev-re-RT strategies represents a promising frontier. A phase I trial of HFSRT combined with pembrolizumab (Pemb) and Bev in 32 rHGG patients demonstrated safety profile but no survival benefits [72]. Ongoing trials (e.g., NCT03743662, NCT06160206) aim to address these gaps.

Newly diagnosed glioblastoma (nGBM)

In rGBM, Bev demonstrates potent tumor-shrinking activity and significantly prolongs PFS, while its combination with RT or TMZ has been demonstrated to be safe. However, the inclusion of Bev in standard first-line therapy for nGBM remains controversial.

With the Stupp regimen

Several phase II studies have reported that the mOS for standard therapy combined with Bev is approximately 19.6-23 months, with a mPFS of approximately 13-14.2 months (Table 6). While PFS was significantly prolonged compared to historical or contemporary controls, OS improvements were observed only relative to historical controls [73]. These findings suggest the survival status of nGBM patients treated with the same first-line regimen has improved compared with that in the EORTC-NCIC era, potentially attributable to advancements in patient management [9,74]. However, cross-trial comparisons are confounded by variability in OS definitions - for instance, Vredenburgh et al. calculated OS from enrollment [73], whereas Lai et al. used the date of diagnosis as the starting point [74].

Table 6.

Bev in combination with chemoradiotherapy for the treatment of nGBM

Ref. Study design No. of patients Study arms mOS (months) mPFS (months)
Vredenburgh 2011 [73] II, single group 75 BEV + RT + TMZ→TMZ + Bev + Iri 21.2 14.2
2006-2008
Lai 2011 [74] II, single group 70 I: BEV + RT + TMZ→TMZ + Bev 19.6 13.6
2006-2008 C1 (UCLA): RT + TMZ→TMZ 21.1 7.6
C2 (EORTC-NCIC): RT + TMZ→TMZ 14.6 6.9
Chinot 2014 AVAglio [8] III, randomized 921 I: Bev + RT + TMZ→TMZ + Bev 16.8 10.6
2009-2011 C: RT + TMZ→TMZ 16.7 6.2*
Gilbert 2014 RTOG 0825 [9] III, randomized 637 I: BEV + RT + TMZ→TMZ + Bev 15.7 10.7
2009-2011 C: RT + TMZ + placebo→TMZ 16.1 7.3
Herrlinger 2016 GLARIUS [165] II, open-label, randomized, MGMTmet 122 I: BEV + RT→Bev + Iri 16.6 5.99
60 C: RT + TMZ→TMZ 17.5 9.7*
Reyes-Botero 2018 ATAG [85] II, single group, older than 70 years, KPS < 70% 66 BEV + TMZ 5.98 3.83
Nagane 2022 [166] II, multicenter 90 BEV + RT + TMZ→TMZ + Bev→Bev 25 14.9
Omuro 2014 MSKCC 08-126 [90] II, 40 HFSRT (36 Gy/6 f) + TMZ + Bev→TMZ + Bev 19 10
Ney 2015 [89] II, 2010-2013 30 Hypo-IMRT (60 Gy/10 f) + Bev + TMZ 16.3 14.3
Carlson 2015 [93] II, comparative 26 Hypo-IMRT (60 Gy/10 f) + TMZ 16.3 9.4
30 Hypo-IMRT (60 Gy/10 f) + TMZ + Bev 16.3 12.8
Wirsching 2018 ARTE [76] II, noncomparative, multicenter, older than 65 years 50 Arm A: Hypo-IMRT (40 Gy/15 f) + Bev 12.1 7.4
2013-2015 25 Arm B: Hypo-IMRT (40 Gy/15) 12.2 4.8*
Matsuda 2018 [167] Retrospective, older than 75 years 18 HFRT (45 Gy/15 f) + TMZ/Bev 20 2.5
2014-2017
Ohno 2019 [88] Retrospective, older than 75 years 20 HFRT (45 Gy/15 f) + TMZ 12.9 8.5
2007-2018 10 HFRT (45 Gy/15 f) + TMZ/Bev 14.6 10.4
Kanamori 2025 [168] II, 2015-2018 49 CNNU + RT + TMZ + Bec 24.8 11.8

Note: UCLA, University of California, Los Angeles; HFSRT, hypofractionated stereotactic radiotherapy; hypo-IMRT, hypofractionated-intensity modulated radiotherapy; HFRT, hypofractionated radiotherapy;

*

P < 0.05.

Two phase III studies (AVAglio [8], RTOG 0825 [9]) subsequently evaluated Bev’s addition to first-line standard therapy, yielding divergent outcomes. Although both studies failed to demonstrate OS benefits, AVAglio reported prolonged PFS (3-4 months) and delayed deterioration in health-related QoL, neurocognitive function, and corticosteroid dependence [8,9]. Conversely, the RTOG 0825 study found accelerated QoL and neurocognitive decline in the Bev arm [75], a trend echoed in the ARTE trial [76]. These discrepancies may reflect differences in response criteria (e.g., the AVAglio study’s modified MacDonald criteria [8]), crossover effects (48% of control-group patients received post-progression Bev), or imbalances in treatment cycles (17-27% higher 6-cycles TMZ completion rates in Bev arms) [8,9]. The incidence of adverse events was similar in both studies, but the percentage of patients who discontinued treatment due to toxicity or complications was approximately 15% greater in the experimental group than in the control group [8,9]. Notably, subgroup analysis from the AVAglio study hinted at OS/PFS benefits in patients not receiving post-progression therapy [77], though clinical implementation remains impractical due to challenges in pretreatment identification.

On the basis of the results of the AVAglio study, Japan approved the use of Bev for treating nGBM in 2013 [78]. Motoo et al. subsequently conducted postmarketing surveillance of Bev in the Japanese GBM population [8,78]. The analysis replicated the AVAglio trial’s safety profile (≥ grade 3 AE rates: 15.1%) and reported a 1-year OS rate of 78% at 18-month follow-up, while the mOS had not yet been reached [78]. However, can the reported survival improvement be entirely attributed to Bev? Can the results from this population be effectively extrapolated to other ethnicities?

Recently, some researchers have suggested the use of Bev before chemoradiotherapy or even before surgery (Table 7). The theoretical basis is that Bev can shrink tumors and prune blood vessels, thereby reducing surgical difficulty and improving the R0 resection rate; Bev can alleviate patients’ brain edema, improve their physical condition, and enhance surgical tolerance [79]. Several small-sample phase II studies have shown that this approach has no advantage in terms of ORR, PFS, or OS and may negatively affect the completion of subsequent treatments due to toxicity [79-81]. The TEMAVIR study further revealed lower concurrent chemoradiation completion rates in the Bev group (58% vs. 82%) [81]. Thus, for large, unresectable nGBM, the benefits of neoadjuvant Bev are limited.

Table 7.

Bev before chemoradiotherapy for the treatment of nGBM

Ref. Study design No. of patients Study arms ORR (%) mOS (months) mPFS (months)
Chauffert 2014 TEMAVIR [81] II, noncomparative, randomized, unresectable 60 BEV + Iri→RT + TMZ + Bev→Bev + Iri NA 11.1 7.1
2009-2011 60 RT + TMZ→TMZ 11.1 5.2
van Linde 2015 [79] Single group 19 BEV + RT + TMZ→TMZ NA 16 9.6
Balana 2016 GENOM 009 [80] II, randomized, unresectable 44 BEV + TMZ→RT + TMZ + Bev→TMZ 22.9 10.6 4.8
2009-2013 43 TMZ→RT + TMZ→TMZ 6.7* 7.7 2.2
Tanaka 2024 [169] I/II 15 Bev + TMZ→Surgery→RT + TMZ→TMZ NA 16.5 9.5
2017-2021
*

P < 0.05;

NA, not available.

In conclusion, the use of Bev in nGBM patients is not recommended due to its high incremental cost-utility ratio. One of the future strategies is to identify a beneficial population, such as the proneuronal subtype linked to OS benefits in AVAglio post hoc analyses [82].

With hypofractionated radiotherapy (HFRT)

HFRT represents a viable therapeutic option for elderly or frail nGBM patients [83]. A systematic review indicates comparable OS between HFRT and standard fractionated in HGG, particularly for GBM patients aged ≥60 years [84]. Bev has also demonstrated potential survival benefits in this population [5,11,74,85]. Notably, TMZ monotherapy or TMZ+HFRT remains preferred for elderly GBM patients with MGMT promoter methylation [86,87].

Compared with conventional fractionation, HFRT offers advantages such as shortened treatment duration, improved patient compliance, and mitigation of tumor cell repopulation via reducing the repair of sublethal damage. However, its application is limited by the elevated risk of RN and delayed neurotoxicity. Bev may theoretically reduce RN and brain edema. Therefore, Bev is expected to improve the tolerance of nGBM patients to hypofractionated radiotherapy while increasing the biological effective dose (BED).

Clinical investigations of HFRT+Bev in elderly GBM patients are summarized in Table 6. A phase II ARTE study compared the HFST ± Bev in elderly or frail nGBM patients with unmethylated MGMT promoter [76]. The results indicated that the addition of Bev extended PFS by approximately 3 months but did not improve OS, a finding similar to that in younger adult nGBM patient populations [76]. Additionally, a retrospective study compared HFRT combined with TMZ or TMZ+Bev in nGBM patients aged ≥75 years and reported that adding Bev increased a nonsignificant 2-month OS [88]. Interestingly, subgroup analysis in this study showed MGMT status showed no prognostic impact [88], contradicting the conclusions of the CAN-NCIC-CE6 study [87]. However, all three studies included surgically treated patients with acceptable tolerance, leaving the utility of these regimens in inoperable elderly GBM patients unclear.

In younger adult patients with nGBM, several studies have investigated Bev+HFRT (Table 6). This section focuses on RN and recurrence patterns reported in these studies. First, higher PTV volumes and escalated radiation doses in HFRT are associated with an increased risk of RN. A phase II study with a median PTV1 volume of 131.1 cm3 (BED = 96) reported RN in 50% of patients, prompting early study termination [89]. Conversely, the MSKCC 08-126 study, which restricted tumor volume to ≤60 cc (BED = 57.6), observed no RN cases [90]. Second, RN has been linked to extended survival outcomes, with patients experiencing RN demonstrating a median OS advantage of approximately 3 months compared to non-RN cohorts [89,91,92]. However, long-term survivors with RN exhibited diminished QoL. Third, Bev seems to have a limited role in RN prevention [89,93], and its use correlates with a shift in failure patterns from local to distant intracranial recurrence (see below) [91-93].

In summary, Bev+HFST did not achieve the expected survival outcomes, and its efficacy in elderly GBM patients is similar to that in younger adult patients. Prospective studies contradict retrospective analyses, as the former do not support Bev’s clinical utility. Current evidence remains confined to phase II studies.

Optimization of Bev dosing regimens and administration strategies

The optimal dosing regimen for Bev in GBM remains undefined. Preclinical glioma mouse models demonstrate that both low and high Bev doses induce vascular regression, with the latter also inhibiting the activity and growth of glioma cells [22]. However, high doses of Bev may exacerbate hypoxia and promote immunosuppressive myeloid cell infiltration.

Interestingly, clinical studies suggest no dose-dependent antitumor efficacy for Bev [94,95]. Most trials have adopted regimens of 5-15 mg/kg every 2-3 weeks. A phase II single-arm study first explored 15 mg/kg Bev every 3 weeks, achieving a PFS6 rate of 25% and mOS of 25.6 weeks [96]. Bokstein et al. reported comparable efficacy with reduced toxicity using low-dose Bev (5 mg/kg biweekly) in HGG patients with poor performance status (mean KPS = 65%) [97]. An early meta-analysis of rGBM studies found no dose-response effect, with similar outcomes between high- (10-15 mg/kg) and low-dose (5 mg/kg) regimens [97,98]. However, it remains unclear whether higher doses lead to a faster onset of action [98]. The phase II VAMANA study recently reported an mOS of 6.1 months for ultralow-dose Bev (1.5 mg/kg triweekly) combined with CCNU in rGBM [99]. In fact, nonrandomized controlled phase II trials were classified as Class III evidence [43].

Enhancing Bev’s intracranial delivery is critical. Rubenstein et al. proposed that antiangiogenic drugs target abnormal blood vessels and are therefore not limited by the blood-brain barrier (BBB) [100]. Indeed, intravenous administration often fails to achieve therapeutic intracranial concentrations in GBM patients due to the BBB and systemic dose-limiting toxicity. Selective intra-arterial drug infusion - direct drug delivery to tumor-feeding arteries - elevates local drug exposure by 3-5.5-fold [101,102]. Patel et al. combined hyperosmotic BBB disruption with superselective intra-arterial brain infusion (SIACI)-administered Bev in nGBM patients, achieving a mPFS of 11.5 months (95% CI 7.7-25.9 months) and mOS of 23.1 months, with no Grade ≥3 TRAEs [103]. Admittedly, this new treatment technique is more precise and shows improved efficacy [103]. However, the study has several limitations, including the absence of a control group and a small sample size (n = 31) [103]. SIACI faces challenges related to complex hemodynamics and transient drug retention [104], and the technique relies on the operator’s skill level, which has a long training period, making widespread adoption difficult.

Notably, the application of novel nanotechnology-based carriers in in vitro models has been reported to suppress angiogenesis and reduce tumor volume, demonstrating promising therapeutic efficacy [105,106]. Moreover, the nanocapsules can be administered intranasally [107]. Recently, an exosome-based Bev delivery platform engineered by Chu et al. demonstrated enhanced BBB penetrability [108].

In conclusion, the distinct pharmacokinetic and pharmacodynamic profiles of targeted therapies compared to traditional chemotherapy necessitate comprehensive dose-response studies for Bev in GBM, providing important evidence for the use of low-dose Bev in patients with poor performance status or in cost-constrained therapeutic settings. The development of advanced drug delivery systems specifically targeting GBM remains insufficient, whereas novel biomimetic materials persist as a focal point in current research efforts.

Treatment failure patterns associated with Bev

The duration of response to Bev in GBM is short, with 40-60% of patients experiencing recurrence within six months, predominantly exhibiting local progression [109,110]. Treatment failure may be driven by phosphofructokinase-1, muscle isoform, mesenchymal transition, and aberrant activation of the cell adhesion molecule pathway [111-113]. Through microarray analysis, DeLay et al. classified Bev-resistant GBM (BRG) MRI enhancement patterns into two subtypes: enhanced (62%) and nonenhanced type (34.2%) [114,115]. BRG patients have an mOS of 2.5 months (range: 1-4.5 months) [96,116].

The impact of Bev on recurrence patterns - specifically, diffuse vs. nondiffuse invasion and multifocal vs. focal growth - remains controversial. Lucio and Rubenstein reported that long-term use of Bev enhances GBM invasiveness, characterized by multifocal satellite lesions and finger-like tumor cell formations [100,117]. The formation of these structures helps hijack existing host blood vessels [100]. Norden et al. observed higher rates of diffuse and metastatic progression in the Bev-treated patients compared to controls (30% vs. 21%) [109,110], potentially mediated by CXCL12/CXCR4 axis activation and MET/VEGFR2 complex formation [118,119]. Conversely, post hoc analyses of the GLARIUS and AVAglio trials found no association between Bev and multifocal or diffuse recurrence [120,121]. Furthermore, the prognostic significance of diffuse progression is unclear. Pope et al. reported similar survival outcomes for patients with local-to-diffuse versus local-to-local progression [122]. Another study indicated that T2 diffuse progression is associated with prolonged survival [123]. Prognostic factors in Bev-treated GBM will be discussed in detail later.

Bev as a third-line therapeutic option for Bev-naïve or resistant rGBM

Currently, there is no standard third-line therapy established for rGBM. Given the limited treatment options, whether Bev can be used to delay or continue treatment for secondary or later relapses remains exploratory. Franceschi et al. retrospectively analyzed 168 GBM patients receiving third-line treatment, revealing a 2-month survival benefit with Bev compared to chemotherapy (P = 0.014) [124]. In most third-line Bev studies, first recurrence (second-line) treatments primarily include CT (such as nitrosoureas) or Bev.

Post-progression options after second-line Bev include continued Bev, CT, and re-RT. First, two randomized phase II trials - CABARET and TAMIGA - evaluated whether Bev continuation after progression during Bev treatment improved outcomes, with no significant survival or QoL benefits compared to non-Bev therapies [116,125]. Schaub et al. reported similar PFS between third- and second-line Bev treatment [126]. Second, retrospective studies noted that Bev combined with nitrosourea increased toxicity without survival benefits [127]. Finally, re-RT combined with Bev demonstrated good tolerability and superior survival (mOS: 4.8-8.8 months) compared to historical controls in rHGG after second-line Bev failure [128-132]. These findings suggest that Bev+re-RT may outperform BEV-based CT in survival and toxicity. Can the addition of IT to re-RT combined with Bev further reverse Bev resistance? A recent phase II study reported improved survival with re-RT plus Pemb and Bev in Bev-refractory patients [133].

On the other hand, two retrospective studies support single-agent Bev feasibility in GBM patients progressing after second-line nitrosoureas (such as CCNU or FTM), with mOS of 6-7.5 months and PFS6 rates of 13-21.5% [134,135].

In summary, for patients progressing after second-line CT or Bev treatment, single-agent Bev or Bev plus re-RT may be viable options. However, due to short survival, many patients only receive first- or second-line treatments, driving current research to optimize interventions for newly diagnosed patients or first recurrence GBM. As OS improves, identifying an optimal subsequent therapy plan will become increasingly important. Recent efforts explore novel agents, such as evofosfamide [136], carotuximab [137], and base excision repair inhibitor TRC102 [138], for Bev-refractory GBM.

Prognostic markers for Bev

In the context of limited survival benefits in the general GBM population, the identification of patient subgroups likely to benefit from Bev is critical.

Genetic, molecular, imaging, and clinical markers associated with Bev response in GBM have been identified in recent studies (Table 8). For instance, Takei et al. demonstrated that reduced FOXM1 expression predicts prolonged survival in Bev-treated GBM patients [139]. Clinical factors such as hypertension have also been linked to favorable responses [140], while Hiller-Vallina et al. identified sexualbiased necroinflammation as a novel positive predictor [141]. Conversely, negative correlations have been reported with soluble CD146 secretion [142], elevated antibody-dependent cellular cytotoxicity (ADCC) activity [143], and c-Met/VEGFR2 overexpression [144], underscoring the multifactorial nature of resistance mechanisms in Bev-treated GBM.

Table 8.

Prognostic biomarkers of BEV

Ref. Prognostic factors Positive/Negative
Fu 2021 [82,112] Low ITGAM expression, proneural subtype Positive
Takei 2022 [139] FOXM1 low-expression Positive
Scheer 2023 [140] Hypertension Positive
Strauss 2025 [170]
Kessler 2023 [171] Neurofibromin 1 mutations Positive
Kim and Breda-Yepes 2023 [172,173] vessel size index and relative cerebral blood volume↑ Negative
Hiller-Vallina 2024 [141] Sexual-biased necroinflammation Positive
Joshkon 2022 [142] high soluble CD146 secretion Negative
Lallemand 2020 [143] ADCC activity↑ Negative
Carvalho 2021 [144] c-Met/VEGFR2 overexpression Negative
Ellingson 2023 [174] time to tumor regrowth, depth of response Positive
Nagane 2022 [166] macrophage or microglia activation Positive
Jiguet-Jiglaire 2022 [175] matrix metalloproteinase 9 Negative

However, the clinical translation of predictive biomarkers is hindered by several challenges. First, study design biases, high examination costs, invasive sample collection, and stringent regulatory approvals limit their practical application [145]. Second, the extent to which predictive biomarkers demonstrate reproducible and independent predictive utility across diverse cohorts and clinical contexts represents a pivotal consideration. Finally, future research should prioritize the integration of technological advancements, including multi-omics approaches and artificial intelligence, alongside fostering interdisciplinary collaborations and standardized protocols to expedite the translation of biomarkers from experimental research to clinical practice.

In brief, the development of personalized treatment paradigms guided by prognostic markers is recognized as an inevitable trajectory in the era of precision medicine.

Summary and outlook

GBM is the most common primary malignant brain tumor, known for its rapid progression and poor prognosis, with limited effective treatment options. Bev is one of only three options approved by the FDA for GBM treatment in the past two decades, leading to a rapid increase in related research. This paper provides a comprehensive review of recent advancements in BEV research, with a focused discussion on topics pertinent to clinical decision-making.

However, the clinical outcomes of Bev in GBM have been disappointing, with several unresolved challenges. First, the mechanisms underlying Bev’s limited efficacy are not fully understood. Second, improvements in short-term efficacy metrics - such as the ORR, PFS6 rates, and PFS - have not translated into OS benefits. Additionally, the optimal sequencing or combination of Bev with other therapies for second- or third-line therapy remains unclear. Finally, clinically applicable prognostic markers to guide Bev use in GBM are still lacking.

Recent advances, such as regorafenib’s survival benefits in rGBM, highlight the potential of antiangiogenic therapies. The exploration of novel agents or multimodal treatment approaches, such as multitarget antiangiogenic agents, phytochemicals, or combination strategies, warrants further investigation. Given the constraints imposed by BBB, enhancing drug delivery efficiency to the brain tumor has emerged as a critical focus in advancing GBM therapeutics development. Concurrently, the identification of patient subgroups likely to benefit from Bev through prognostic biomarker-driven stratification has emerged as a key area of investigation in current research. The Global Brain Tumor Adaptive Clinical Trial System (GBM AGILE, NCT03970447) has pioneered an innovative framework for clinical trial design, significantly streamlining the assessment of emerging therapies.

Acknowledgements

The Graphical Abstract in this review was created with Biorender (https://www.biorender.com/). This paper was supported by [National Natural Science Foundation of China] under Grant [number: 82172804].

Disclosure of conflict of interest

None.

References

  • 1.Ostrom QT, Price M, Neff C, Cioffi G, Waite KA, Kruchko C, Barnholtz-Sloan JS. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2016-2020. Neuro Oncol. 2023;25:iv1–iv99. doi: 10.1093/neuonc/noad149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Stark-Vance V. Bevacizumab and CPT-11 in the treatment of relapsed malignant glioma. Neuro Oncology. 2005;7:369. [Google Scholar]
  • 3.Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E, Ferrara N, Fyfe G, Rogers B, Ross R, Kabbinavar F. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335–2342. doi: 10.1056/NEJMoa032691. [DOI] [PubMed] [Google Scholar]
  • 4.Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE, Yung WKA, Paleologos N, Nicholas MK, Jensen R, Vredenburgh J, Huang J, Zheng M, Cloughesy T. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J. Clin. Oncol. 2009;27:4733–4740. doi: 10.1200/JCO.2008.19.8721. [DOI] [PubMed] [Google Scholar]
  • 5.Kreisl TN, Kim L, Moore K, Duic P, Royce C, Stroud I, Garren N, Mackey M, Butman JA, Camphausen K, Park J, Albert PS, Fine HA. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. JCO. 2009;27:740–745. doi: 10.1200/JCO.2008.16.3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Vredenburgh JJ, Desjardins A, Herndon JE, Marcello J, Reardon DA, Quinn JA, Rich JN, Sathornsumetee S, Gururangan S, Sampson J, Wagner M, Bailey L, Bigner DD, Friedman AH, Friedman HS. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J. Clin. Oncol. 2007;25:4722–4729. doi: 10.1200/JCO.2007.12.2440. [DOI] [PubMed] [Google Scholar]
  • 7.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: 10.1634/theoncologist.2009-0121. [DOI] [PubMed] [Google Scholar]
  • 8.Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, Carpentier AF, Hoang-Xuan K, Kavan P, Cernea D, Brandes AA, Hilton M, Abrey L, Cloughesy T. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370:709–722. doi: 10.1056/NEJMoa1308345. [DOI] [PubMed] [Google Scholar]
  • 9.Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, Colman H, Chakravarti A, Pugh S, Won M, Jeraj R, Brown PD, Jaeckle KA, Schiff D, Stieber VW, Brachman DG, Werner-Wasik M, Tremont-Lukats IW, Sulman EP, Aldape KD, Curran WJ, Mehta MP. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370:699–708. doi: 10.1056/NEJMoa1308573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wick W, Gorlia T, Bendszus M, Taphoorn M, Sahm F, Harting I, Brandes AA, Taal W, Domont J, Idbaih A, Campone M, Clement PM, Stupp R, Fabbro M, Le Rhun E, Dubois F, Weller M, Von Deimling A, Golfinopoulos V, Bromberg JC, Platten M, Klein M, Van Den Bent MJ. Lomustine and bevacizumab in progressive glioblastoma. N Engl J Med. 2017;377:1954–1963. doi: 10.1056/NEJMoa1707358. [DOI] [PubMed] [Google Scholar]
  • 11.Nghiemphu PL, Liu W, Lee Y, Than T, Graham C, Lai A, Green RM, Pope WB, Liau LM, Mischel PS, Nelson SF, Elashoff R, Cloughesy TF. Bevacizumab and chemotherapy for recurrent glioblastoma: a single-institution experience. Neurology. 2009;72:1217–1222. doi: 10.1212/01.wnl.0000345668.03039.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Brandes AA, Finocchiaro G, Zagonel V, Reni M, Caserta C, Fabi A, Clavarezza M, Maiello E, Eoli M, Lombardi G, Monteforte M, Proietti E, Agati R, Eusebi V, Franceschi E. AVAREG: a phase II, randomized, noncomparative study of fotemustine or bevacizumab for patients with recurrent glioblastoma. NEUONC. 2016;18:1304–1312. doi: 10.1093/neuonc/now035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Tamma R, Ingravallo G, Annese T, d’Amati A, Lorusso L, Ribatti D. Tumor microenvironment and microvascular density in human glioblastoma. Cells. 2022;12:11. doi: 10.3390/cells12010011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Groblewska M, Mroczko B. Pro- and antiangiogenic factors in gliomas: implications for novel therapeutic possibilities. IJMS. 2021;22:6126. doi: 10.3390/ijms22116126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Michaelsen SR, Staberg M, Pedersen H, Jensen KE, Majewski W, Broholm H, Nedergaard MK, Meulengracht C, Urup T, Villingshøj M, Lukacova S, Skjøth-Rasmussen J, Brennum J, Kjær A, Lassen U, Stockhausen MT, Poulsen HS, Hamerlik P. VEGF-C sustains VEGFR2 activation under bevacizumab therapy and promotes glioblastoma maintenance. Neuro Oncol. 2018;20:1462–1474. doi: 10.1093/neuonc/noy103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tamura R, Tanaka T, Akasaki Y, Murayama Y, Yoshida K, Sasaki H. The role of vascular endothelial growth factor in the hypoxic and immunosuppressive tumor microenvironment: perspectives for therapeutic implications. Med Oncol. 2019;37:2. doi: 10.1007/s12032-019-1329-2. [DOI] [PubMed] [Google Scholar]
  • 17.Gerber HP, Ferrara N. Pharmacology and pharmacodynamics of bevacizumab as monotherapy or in combination with cytotoxic therapy in preclinical studies. Cancer Res. 2005;65:671–680. [PubMed] [Google Scholar]
  • 18.Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB, Shi Q, McLendon RE, Bigner DD, Rich JN. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 2006;66:7843–7848. doi: 10.1158/0008-5472.CAN-06-1010. [DOI] [PubMed] [Google Scholar]
  • 19.Kaur B, Khwaja FW, Severson EA, Matheny SL, Brat DJ, Van Meir EG. Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro Oncol. 2005;7:134–153. doi: 10.1215/S1152851704001115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Pore N, Liu S, Haas-Kogan DA, O’Rourke DM, Maity A. PTEN mutation and epidermal growth factor receptor activation regulate vascular endothelial growth factor (VEGF) mRNA expression in human glioblastoma cells by transactivating the proximal VEGF promoter. Cancer Res. 2003;63:236–241. [PubMed] [Google Scholar]
  • 21.Reardon DA, Turner S, Peters KB, Desjardins A, Gururangan S, Sampson JH, McLendon RE, Herndon JE, Jones LW, Kirkpatrick JP, Friedman AH, Vredenburgh JJ, Bigner DD, Friedman HS. A review of VEGF/VEGFR-targeted therapeutics for recurrent glioblastoma. J Natl Compr Canc Netw. 2011;9:414–427. doi: 10.6004/jnccn.2011.0038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.García-Romero N, Palacín-Aliana I, Madurga R, Carrión-Navarro J, Esteban-Rubio S, Jiménez B, Collazo A, Pérez-Rodríguez F, Ortiz de Mendivil A, Fernández-Carballal C, García-Duque S, Diamantopoulos-Fernández J, Belda-Iniesta C, Prat-Acín R, Sánchez-Gómez P, Calvo E, Ayuso-Sacido A. Bevacizumab dose adjustment to improve clinical outcomes of glioblastoma. BMC Med. 2020;18:142. doi: 10.1186/s12916-020-01610-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Joensuu H, Puputti M, Sihto H, Tynninen O, Nupponen NN. Amplification of genes encoding KIT, PDGFRalpha and VEGFR2 receptor tyrosine kinases is frequent in glioblastoma multiforme. J Pathol. 2005;207:224–231. doi: 10.1002/path.1823. [DOI] [PubMed] [Google Scholar]
  • 24.Huang H, Held-Feindt J, Buhl R, Mehdorn HM, Mentlein R. Expression of VEGF and its receptors in different brain tumors. Neurol Res. 2005;27:371–377. doi: 10.1179/016164105X39833. [DOI] [PubMed] [Google Scholar]
  • 25.Stefanik DF, Fellows WK, Rizkalla LR, Rizkalla WM, Stefanik PP, Deleo AB, Welch WC. Monoclonal antibodies to vascular endothelial growth factor (VEGF) and the VEGF receptor, FLT-1, inhibit the growth of C6 glioma in a mouse xenograft. J Neurooncol. 2001;55:91–100. doi: 10.1023/a:1013329832067. [DOI] [PubMed] [Google Scholar]
  • 26.Horbinski C, Nabors LB, Portnow J, Baehring J, Bhatia A, Bloch O, Brem S, Butowski N, Cannon DM, Chao S, Chheda MG, Fabiano AJ, Forsyth P, Gigilio P, Hattangadi-Gluth J, Holdhoff M, Junck L, Kaley T, Merrell R, Mrugala MM, Nagpal S, Nedzi LA, Nevel K, Nghiemphu PL, Parney I, Patel TR, Peters K, Puduvalli VK, Rockhill J, Rusthoven C, Shonka N, Swinnen LJ, Weiss S, Wen PY, Willmarth NE, Bergman MA, Darlow S. NCCN guidelines® insights: central nervous system cancers, version 2.2022. J Natl Compr Canc Netw. 2023;21:12–20. doi: 10.6004/jnccn.2023.0002. [DOI] [PubMed] [Google Scholar]
  • 27.Nagane M, Nishikawa R, Narita Y, Kobayashi H, Takano S, Shinoura N, Aoki T, Sugiyama K, Kuratsu J, Muragaki Y, Sawamura Y, Matsutani M. Phase II study of single-agent bevacizumab in Japanese patients with recurrent malignant glioma. Jpn J Clin Oncol. 2012;42:887–895. doi: 10.1093/jjco/hys121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Weller M, Van Den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, Bendszus M, Balana C, Chinot O, Dirven L, French P, Hegi ME, Jakola AS, Platten M, Roth P, Rudà R, Short S, Smits M, Taphoorn MJB, Von Deimling A, Westphal M, Soffietti R, Reifenberger G, Wick W. EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol. 2021;18:170–186. doi: 10.1038/s41571-020-00447-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ballman KV, Buckner JC, Brown PD, Giannini C, Flynn PJ, LaPlant BR, Jaeckle KA. The relationship between six-month progression-free survival and 12-month overall survival end points for phase II trials in patients with glioblastoma multiforme. Neuro Oncol. 2007;9:29–38. doi: 10.1215/15228517-2006-025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Jaspan T, Morgan PS, Warmuth-Metz M, Sanchez Aliaga E, Warren D, Calmon R, Grill J, Hargrave D, Garcia J, Zahlmann G. Response assessment in pediatric neuro-oncology: implementation and expansion of the RANO criteria in a randomized phase II trial of pediatric patients with newly diagnosed high-grade gliomas. AJNR Am J Neuroradiol. 2016;37:1581–1587. doi: 10.3174/ajnr.A4782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wen PY, van den Bent M, Youssef G, Cloughesy TF, Ellingson BM, Weller M, Galanis E, Barboriak DP, de Groot J, Gilbert MR, Huang R, Lassman AB, Mehta M, Molinaro AM, Preusser M, Rahman R, Shankar LK, Stupp R, Villanueva-Meyer JE, Wick W, Macdonald DR, Reardon DA, Vogelbaum MA, Chang SM. RANO 2.0: update to the response assessment in neuro-oncology criteria for high- and low-grade gliomas in adults. J. Clin. Oncol. 2023;41:5187–5199. doi: 10.1200/JCO.23.01059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Le Rhun E, Oppong FB, van den Bent M, Wick W, Brandes AA, Taphoorn MJ, Platten M, Idbaih A, Clement PM, Preusser M, Golfinopoulos V, Gorlia T, Weller M. Thrombocytopenia limits the feasibility of salvage lomustine chemotherapy in recurrent glioblastoma: a secondary analysis of EORTC 26101. Eur J Cancer. 2023;178:13–22. doi: 10.1016/j.ejca.2022.10.006. [DOI] [PubMed] [Google Scholar]
  • 33.Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE, Yung WKA, Paleologos N, Nicholas MK, Jensen R, Vredenburgh J, Huang J, Zheng M, Cloughesy T. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J. Clin. Oncol. 2023;41:4945–4952. doi: 10.1200/JCO.22.02772. [DOI] [PubMed] [Google Scholar]
  • 34.Dickson PV, Hamner JB, Sims TL, Fraga CH, Ng CYC, Rajasekeran S, Hagedorn NL, McCarville MB, Stewart CF, Davidoff AM. Bevacizumab-induced transient remodeling of the vasculature in neuroblastoma xenografts results in improved delivery and efficacy of systemically administered chemotherapy. Clin Cancer Res. 2007;13:3942–3950. doi: 10.1158/1078-0432.CCR-07-0278. [DOI] [PubMed] [Google Scholar]
  • 35.Chamberlain MC. Bevacizumab plus irinotecan in recurrent glioblastoma. J. Clin. Oncol. 2008;26:1012–1013. doi: 10.1200/JCO.2007.15.1605. author reply 1013. [DOI] [PubMed] [Google Scholar]
  • 36.Taal W, Oosterkamp HM, Walenkamp AME, Dubbink HJ, Beerepoot LV, Hanse MCJ, Buter J, Honkoop AH, Boerman D, De Vos FYF, Dinjens WNM, Enting RH, Taphoorn MJB, Van Den Berkmortel FWPJ, Jansen RLH, Brandsma D, Bromberg JEC, Van Heuvel I, Vernhout RM, Van Der Holt B, Van Den Bent MJ. Single-agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): a randomised controlled phase 2 trial. Lancet Oncol. 2014;15:943–953. doi: 10.1016/S1470-2045(14)70314-6. [DOI] [PubMed] [Google Scholar]
  • 37.Chen Z, Tian F, Zhang Y. Cost-effectiveness analysis of bevacizumab combined with lomustine in the treatment of progressive glioblastoma using a Markov model simulation analysis. Front Public Health. 2024;12:1410355. doi: 10.3389/fpubh.2024.1410355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Field KM, Simes J, Nowak AK, Cher L, Wheeler H, Hovey EJ, Brown CS, Barnes EH, Sawkins K, Livingstone A, Freilich R, Phal PM, Fitt G CABARET/COGNO investigators. Rosenthal MA. Randomized phase 2 study of carboplatin and bevacizumab in recurrent glioblastoma. Neuro-Oncology. 2015;17:1504–1513. doi: 10.1093/neuonc/nov104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Cloughesy T, Finocchiaro G, Belda-Iniesta C, Recht L, Brandes AA, Pineda E, Mikkelsen T, Chinot OL, Balana C, Macdonald DR, Westphal M, Hopkins K, Weller M, Bais C, Sandmann T, Bruey JM, Koeppen H, Liu B, Verret W, Phan SC, Shames DS. Randomized, double-blind, placebo-controlled, multicenter phase II study of onartuzumab plus bevacizumab versus placebo plus bevacizumab in patients with recurrent glioblastoma: efficacy, safety, and hepatocyte growth factor and O6-methylguanine-DNA methyltransferase biomarker analyses. J. Clin. Oncol. 2017;35:343–351. doi: 10.1200/JCO.2015.64.7685. [DOI] [PubMed] [Google Scholar]
  • 40.Cruz Da Silva E, Mercier MC, Etienne-Selloum N, Dontenwill M, Choulier L. A systematic review of glioblastoma-targeted therapies in phases II, III, IV clinical trials. Cancers (Basel) 2021;13:1795. doi: 10.3390/cancers13081795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kelly W, Diaz Duque AE, Michalek J, Konkel B, Caflisch L, Chen Y, Pathuri SC, Madhusudanannair-Kunnuparampil V, Floyd J, Brenner A. Phase II investigation of TVB-2640 (Denifanstat) with bevacizumab in patients with first relapse high-grade astrocytoma. Clin Cancer Res. 2023;29:2419–2425. doi: 10.1158/1078-0432.CCR-22-2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zhao S, Zhang M, Zhang Q, Wu J, Dai H. Anlotinib alone or in combination with bevacizumab in the treatment of recurrent high-grade glioma: a prospective single-arm, open-label phase II trial. BMC Cancer. 2024;24:6. doi: 10.1186/s12885-023-11776-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Winograd E, Germano I, Wen P, Olson JJ, Ormond DR. Congress of neurological surgeons systematic review and evidence-based guidelines update on the role of targeted therapies and immunotherapies in the management of progressive glioblastoma. J Neurooncol. 2022;158:265–321. doi: 10.1007/s11060-021-03876-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Motz GT, Santoro SP, Wang LP, Garrabrant T, Lastra RR, Hagemann IS, Lal P, Feldman MD, Benencia F, Coukos G. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat Med. 2014;20:607–615. doi: 10.1038/nm.3541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Nayak L, Standifer N, Dietrich J, Clarke JL, Dunn GP, Lim M, Cloughesy T, Gan HK, Flagg E, George E, Gaffey S, Hayden J, Holcroft C, Wen PY, Macri M, Park AJ, Ricciardi T, Ryan A, Schwarzenberger P, Venhaus R, Reyes M de L, Durham NM, Creasy T, Huang RY, Kaley T, Reardon DA. Circulating immune cell and outcome analysis from the phase II study of PD-L1 blockade with durvalumab for newly diagnosed and recurrent glioblastoma. Clin Cancer Res. 2022;28:2567–2578. doi: 10.1158/1078-0432.CCR-21-4064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wang F, Jin Y, Wang M, Luo HY, Fang WJ, Wang YN, Chen YX, Huang RJ, Guan WL, Li JB, Li YH, Wang FH, Hu XH, Zhang YQ, Qiu MZ, Liu LL, Wang ZX, Ren C, Wang DS, Zhang DS, Wang ZQ, Liao WT, Tian L, Zhao Q, Xu RH. Combined anti-PD-1, HDAC inhibitor and anti-VEGF for MSS/pMMR colorectal cancer: a randomized phase 2 trial. Nat Med. 2024;30:1035–1043. doi: 10.1038/s41591-024-02813-1. [DOI] [PubMed] [Google Scholar]
  • 47.Reardon DA, Brandes AA, Omuro A, Mulholland P, Lim M, Wick A, Baehring J, Ahluwalia MS, Roth P, Bähr O, Phuphanich S, Sepulveda JM, De Souza P, Sahebjam S, Carleton M, Tatsuoka K, Taitt C, Zwirtes R, Sampson J, Weller M. Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: the CheckMate 143 phase 3 randomized clinical trial. JAMA Oncol. 2020;6:1003–1010. doi: 10.1001/jamaoncol.2020.1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Nayak L, Molinaro AM, Peters K, Clarke JL, Jordan JT, de Groot J, Nghiemphu L, Kaley T, Colman H, McCluskey C, Gaffey S, Smith TR, Cote DJ, Severgnini M, Yearley JH, Zhao Q, Blumenschein WM, Duda DG, Muzikansky A, Jain RK, Wen PY, Reardon DA. Randomized phase II and biomarker study of pembrolizumab plus bevacizumab versus pembrolizumab alone for patients with recurrent glioblastoma. Clin Cancer Res. 2021;27:1048–1057. doi: 10.1158/1078-0432.CCR-20-2500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Chiu D, Qi J, Thin TH, Garcia-Barros M, Lee B, Hahn M, Mandeli J, Belani P, Nael K, Rashidipour O, Ghatan S, Hadjipanayis CG, Yong RL, Germano IM, Brody R, Tsankova NM, Gnjatic S, Kim-Schulze S, Hormigo A. A phase I trial of VEGF-A inhibition combined with PD-L1 blockade for recurrent glioblastoma. Cancer Res Commun. 2023;3:130–139. doi: 10.1158/2767-9764.CRC-22-0420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Guo G, Zhang Z, Zhang J, Wang D, Xu S, Liu G, Gao Y, Mei J, Yan Z, Zhao R, Wang M, Li T, Bu X. Predicting recurrent glioblastoma clinical outcome to immune checkpoint inhibition and low-dose bevacizumab with tumor in situ fluid circulating tumor DNA analysis. Cancer Immunol Immunother. 2024;73:193. doi: 10.1007/s00262-024-03774-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wang D, Zhang J, Bu C, Liu G, Guo G, Zhang Z, Lv G, Sheng Z, Yan Z, Gao Y, Wang M, Liu G, Zhao R, Li T, Ma C, Bu X. Dynamics of tumor in situ fluid circulating tumor DNA in recurrent glioblastomas forecasts treatment efficacy of immune checkpoint blockade coupled with low-dose bevacizumab. J Cancer Res Clin Oncol. 2024;150:466. doi: 10.1007/s00432-024-05997-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Yang G, Fang Y, Zhou M, Li W, Dong D, Chen J, Da Y, Wang K, Li X, Zhang X, Ma T, Shen G. Case report: the effective response to pembrolizumab in combination with bevacizumab in the treatment of a recurrent glioblastoma with multiple extracranial metastases. Front Oncol. 2022;12:948933. doi: 10.3389/fonc.2022.948933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Cloughesy TF, Brenner A, de Groot JF, Butowski NA, Zach L, Campian JL, Ellingson BM, Freedman LS, Cohen YC, Lowenton-Spier N, Rachmilewitz Minei T, Fain Shmueli S GLOBE Study Investigators. Wen PY. A randomized controlled phase III study of VB-111 combined with bevacizumab vs bevacizumab monotherapy in patients with recurrent glioblastoma (GLOBE) Neuro Oncol. 2020;22:705–717. doi: 10.1093/neuonc/noz232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Brenner AJ, Peters KB, Vredenburgh J, Bokstein F, Blumenthal DT, Yust-Katz S, Peretz I, Oberman B, Freedman LS, Ellingson BM, Cloughesy TF, Sher N, Cohen YC, Lowenton-Spier N, Rachmilewitz Minei T, Yakov N, Mendel I, Breitbart E, Wen PY. Safety and efficacy of VB-111, an anticancer gene therapy, in patients with recurrent glioblastoma: results of a phase I/II study. Neuro Oncol. 2020;22:694–704. doi: 10.1093/neuonc/noz231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Cloughesy TF, Mochizuki AY, Orpilla JR, Hugo W, Lee AH, Davidson TB, Wang AC, Ellingson BM, Rytlewski JA, Sanders CM, Kawaguchi ES, Du L, Li G, Yong WH, Gaffey SC, Cohen AL, Mellinghoff IK, Lee EQ, Reardon DA, O’Brien BJ, Butowski NA, Nghiemphu PL, Clarke JL, Arrillaga-Romany IC, Colman H, Kaley TJ, de Groot JF, Liau LM, Wen PY, Prins RM. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat Med. 2019;25:477–486. doi: 10.1038/s41591-018-0337-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Reardon DA, Desjardins A, Vredenburgh JJ, O’Rourke DM, Tran DD, Fink KL, Nabors LB, Li G, Bota DA, Lukas RV, Ashby LS, Duic JP, Mrugala MM, Cruickshank S, Vitale L, He Y, Green JA, Yellin MJ, Turner CD, Keler T, Davis TA, Sampson JH ReACT trial investigators. Rindopepimut with bevacizumab for patients with relapsed EGFRvIII-expressing glioblastoma (ReACT): Results of a double-blind randomized phase II trial. Clin Cancer Res. 2020;26:1586–1594. doi: 10.1158/1078-0432.CCR-18-1140. [DOI] [PubMed] [Google Scholar]
  • 57.Fu M, Zhou Z, Huang X, Chen Z, Zhang L, Zhang J, Hua W, Mao Y. Use of bevacizumab in recurrent glioblastoma: a scoping review and evidence map. BMC Cancer. 2023;23:544. doi: 10.1186/s12885-023-11043-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Fleischmann DF, Gajdi L, Corradini S, Schönecker S, Marschner S, Bodensohn R, Hofmaier J, Garny S, Forbrig R, Thon N, Belka C, Niyazi M. Re-irradiation treatment regimens for patients with recurrent glioma - evaluation of the optimal dose and best concurrent therapy. Radiother Oncol. 2024;199:110437. doi: 10.1016/j.radonc.2024.110437. [DOI] [PubMed] [Google Scholar]
  • 59.Tsien CI, Pugh SL, Dicker AP, Raizer JJ, Matuszak MM, Lallana EC, Huang J, Algan O, Deb N, Portelance L, Villano JL, Hamm JT, Oh KS, Ali AN, Kim MM, Lindhorst SM, Mehta MP. NRG oncology/RTOG1205: a randomized phase II trial of concurrent bevacizumab and reirradiation versus bevacizumab alone as treatment for recurrent glioblastoma. J. Clin. Oncol. 2023;41:1285–1295. doi: 10.1200/JCO.22.00164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Guan Y, Xiong J, Pan M, Shi W, Li J, Zhu H, Gong X, Li C, Mei G, Liu X, Pan L, Dai J, Wang Y, Wang E, Wang X. Safety and efficacy of Hypofractionated stereotactic radiosurgery for high-grade Gliomas at first recurrence: a single-center experience. BMC cancer. 2021;21:123. doi: 10.1186/s12885-021-07856-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Palmer JD, Bhamidipati D, Song A, Eldredge-Hindy HB, Siglin J, Dan TD, Champ CE, Zhang I, Bar-Ad V, Kim L, Glass J, Evans JJ, Andrews DW, Werner-Wasik M, Shi W. Bevacizumab and re-irradiation for recurrent high grade gliomas: does sequence matter? J Neurooncol. 2018;140:623–628. doi: 10.1007/s11060-018-2989-z. [DOI] [PubMed] [Google Scholar]
  • 62.Cuneo KC, Vredenburgh JJ, Sampson JH, Reardon DA, Desjardins A, Peters KB, Friedman HS, Willett CG, Kirkpatrick JP. Safety and efficacy of stereotactic radiosurgery and adjuvant bevacizumab in patients with recurrent malignant gliomas. Int J Radiat Oncol Biol Phys. 2012;82:2018–2024. doi: 10.1016/j.ijrobp.2010.12.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Helis CA, Zarabi H, Russell G, LeCompte M, Liu W, Hattangadi-Gluth J, Halasz LM, Soltys SG, Braunstein SE, Wang TJC, Shi W, Shen C, Mignano J, Masters A, Kleinberg L, Huang J, Hopper A, Barbour AB, Jeyapalan SA, Chan MD. Multi-institutional study of reirradiation for recurrent high grade glioma. J. Clin. Oncol. 2024:2063–2063. [Google Scholar]
  • 64.Chen ATC, Serante AR, Ayres AS, Tonaki JO, Moreno RA, Shih H, Gattás GS, Lopez RVM, Dos Santos de Jesus GR, de Carvalho IT, Marotta RC, Marta GN, Feher O, Neto HS, Ribeiro ISN, Vasconcelos KGMDC, Figueiredo EG, Weltman E. Prospective randomized phase 2 trial of hypofractionated stereotactic radiation therapy of 25 gy in 5 fractions compared with 35 gy in 5 fractions in the reirradiation of recurrent glioblastoma. Int J Radiat Oncol Biol Phys. 2024;119:1122–1132. doi: 10.1016/j.ijrobp.2024.01.013. [DOI] [PubMed] [Google Scholar]
  • 65.Arvold ND, Shi DD, Aizer AA, Norden AD, Reardon DA, Lee EQ, Nayak L, Dunn IF, Golby AJ, Johnson MD, Claus EB, Chiocca EA, Ligon KL, Wen PY, Alexander BM. Salvage re-irradiation for recurrent high-grade glioma and comparison to bevacizumab alone. J Neurooncol. 2017;135:581–591. doi: 10.1007/s11060-017-2611-9. [DOI] [PubMed] [Google Scholar]
  • 66.Kulinich DP, Sheppard JP, Nguyen T, Kondajji AM, Unterberger A, Duong C, Enomoto A, Patel K, Yang I. Radiotherapy versus combination radiotherapy-bevacizumab for the treatment of recurrent high-grade glioma: a systematic review. Acta Neurochir (Wien) 2021;163:1921–1934. doi: 10.1007/s00701-021-04794-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Popp I, Weber WA, Graf E, Mix M, Wiehle R, Nestle U, Schimek-Jasch T, Niyazi M, Belka C, Paulsen F, Eckert F, Eble MJ, König L, Giordano FA, Sperk E, Momm F, Spehl I, Combs SE, Bernhardt D, Grosu AL. Re-irradiation in recurrent glioblastoma: PET- or MRI-based? Results of a prospective randomized clinical trial. J. Clin. Oncol. 2024;42:2021–2021. [Google Scholar]
  • 68.Tong E, Horsley P, Wheeler H, Wong M, Venkatesha V, Chan J, Kastelan M, Back M. Hypofractionated re-irradiation with bevacizumab for relapsed chemorefractory glioblastoma after prior high dose radiotherapy: a feasible option for patients with large-volume relapse. J Neurooncol. 2024;168:69–76. doi: 10.1007/s11060-024-04643-0. [DOI] [PubMed] [Google Scholar]
  • 69.Lee ST, Seo Y, Bae JY, Chu K, Kim JW, Choi SH, Kim TM, Kim IH, Park SH, Park CK. Loss of pericytes in radiation necrosis after glioblastoma treatments. Mol Neurobiol. 2018;55:4918–4926. doi: 10.1007/s12035-017-0695-z. [DOI] [PubMed] [Google Scholar]
  • 70.Khalaj K, Jacobs MA, Zhu JJ, Esquenazi Y, Hsu S, Tandon N, Akhbardeh A, Zhang X, Riascos R, Kamali A. The use of apparent diffusion coefficient values for differentiating bevacizumab-related cytotoxicity from tumor recurrence and radiation necrosis in glioblastoma. Cancers (Basel) 2024;16:2440. doi: 10.3390/cancers16132440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Dasgupta A, Sawant S, Chatterjee A, Gota V, Sahu A, Choudhari A, Bhattacharya K, Puranik A, Dev I, Moiyadi A, Shetty P, Singh V, Menon N, Epari S, Sahay A, Shah A, Bano N, Shaikh F, Jirage A, Gupta T. Study protocol of a prospective phase 2 study of chlorophyllin for the management of brain radionecrosis in patients with diffuse glioma (CHROME) Cancer Med. 2025;14:e70657. doi: 10.1002/cam4.70657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Sahebjam S, Forsyth PA, Tran ND, Arrington JA, Macaulay R, Etame AB, Walko CM, Boyle T, Peguero EN, Jaglal M, Mokhtari S, Enderling H, Raghunand N, Gatewood T, Long W, Dzierzeski JL, Evernden B, Robinson T, Wicklund MC, Kim S, Thompson ZJ, Chen DT, Chinnaiyan P, Yu HM. Hypofractionated stereotactic re-irradiation with pembrolizumab and bevacizumab in patients with recurrent high-grade gliomas: results from a phase I study. Neuro Oncol. 2021;23:677–686. doi: 10.1093/neuonc/noaa260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Vredenburgh JJ, Desjardins A, Reardon DA, Peters KB, Herndon JE, Marcello J, Kirkpatrick JP, Sampson JH, Bailey L, Threatt S, Friedman AH, Bigner DD, Friedman HS. The addition of bevacizumab to standard radiation therapy and temozolomide followed by bevacizumab, temozolomide, and irinotecan for newly diagnosed glioblastoma. Clin Cancer Res. 2011;17:4119–4124. doi: 10.1158/1078-0432.CCR-11-0120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lai A, Tran A, Nghiemphu PL, Pope WB, Solis OE, Selch M, Filka E, Yong WH, Mischel PS, Liau LM, Phuphanich S, Black K, Peak S, Green RM, Spier CE, Kolevska T, Polikoff J, Fehrenbacher L, Elashoff R, Cloughesy T. Phase II study of bevacizumab plus temozolomide during and after radiation therapy for patients with newly diagnosed glioblastoma multiforme. J. Clin. Oncol. 2011;29:142–148. doi: 10.1200/JCO.2010.30.2729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wefel JS, Armstrong TS, Pugh SL, Gilbert MR, Wendland MM, Brachman DG, Roof KS, Brown PD, Crocker IR, Robins HI, Hunter G, Won M, Mehta MP. Neurocognitive, symptom, and health-related quality of life outcomes of a randomized trial of bevacizumab for newly diagnosed glioblastoma (NRG/RTOG 0825) Neuro Oncol. 2021;23:1125–1138. doi: 10.1093/neuonc/noab011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wirsching HG, Tabatabai G, Roelcke U, Hottinger AF, Jörger F, Schmid A, Plasswilm L, Schrimpf D, Mancao C, Capper D, Conen K, Hundsberger T, Caparrotti F, von Moos R, Riklin C, Felsberg J, Roth P, Jones DTW, Pfister S, Rushing EJ, Abrey L, Reifenberger G, Held L, von Deimling A, Ochsenbein A, Weller M. Bevacizumab plus hypofractionated radiotherapy versus radiotherapy alone in elderly patients with glioblastoma: the randomized, open-label, phase II ARTE trial. Ann Oncol. 2018;29:1423–1430. doi: 10.1093/annonc/mdy120. [DOI] [PubMed] [Google Scholar]
  • 77.Chinot OL, Nishikawa R, Mason W, Henriksson R, Saran F, Cloughesy T, Garcia J, Revil C, Abrey L, Wick W. Upfront bevacizumab may extend survival for glioblastoma patients who do not receive second-line therapy: an exploratory analysis of AVAglio. Neuro Oncol. 2016;18:1313–1318. doi: 10.1093/neuonc/now046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Motoo N, Hayashi Y, Shimizu A, Ura M, Nishikawa R. Safety and effectiveness of bevacizumab in Japanese patients with malignant glioma: a post-marketing surveillance study. Jpn J Clin Oncol. 2019;49:1016–1023. doi: 10.1093/jjco/hyz125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.van Linde ME, Verhoeff JJC, Richel DJ, van Furth WR, Reijneveld JC, Verheul HMW, Stalpers LJA. Bevacizumab in combination with radiotherapy and temozolomide for patients with newly diagnosed glioblastoma multiforme. Oncologist. 2015;20:107–108. doi: 10.1634/theoncologist.2014-0418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Balana C, De Las Penas R, Sepúlveda JM, Gil-Gil MJ, Luque R, Gallego O, Carrato C, Sanz C, Reynes G, Herrero A, Ramirez JL, Pérez-Segura P, Berrocal A, Vieitez JM, Garcia A, Vazquez-Estevez S, Peralta S, Fernandez I, Henriquez I, Martinez-Garcia M, De la Cruz JJ, Capellades J, Giner P, Villà S. Bevacizumab and temozolomide versus temozolomide alone as neoadjuvant treatment in unresected glioblastoma: the GENOM 009 randomized phase II trial. J Neurooncol. 2016;127:569–579. doi: 10.1007/s11060-016-2065-5. [DOI] [PubMed] [Google Scholar]
  • 81.Chauffert B, Feuvret L, Bonnetain F, Taillandier L, Frappaz D, Taillia H, Schott R, Honnorat J, Fabbro M, Tennevet I, Ghiringhelli F, Guillamo JS, Durando X, Castera D, Frenay M, Campello C, Dalban C, Skrzypski J, Chinot O. Randomized phase II trial of irinotecan and bevacizumab as neo-adjuvant and adjuvant to temozolomide-based chemoradiation compared with temozolomide-chemoradiation for unresectable glioblastoma: final results of the TEMAVIR study from ANOCEF†. Ann Oncol. 2014;25:1442–1447. doi: 10.1093/annonc/mdu148. [DOI] [PubMed] [Google Scholar]
  • 82.Sandmann T, Bourgon R, Garcia J, Li C, Cloughesy T, Chinot OL, Wick W, Nishikawa R, Mason W, Henriksson R, Saran F, Lai A, Moore N, Kharbanda S, Peale F, Hegde P, Abrey LE, Phillips HS, Bais C. Patients with proneural glioblastoma may derive overall survival benefit from the addition of bevacizumab to first-line radiotherapy and temozolomide: retrospective analysis of the AVAglio trial. JCO. 2015;33:2735–2744. doi: 10.1200/JCO.2015.61.5005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Chang EL, Yi W, Allen PK, Levin VA, Sawaya RE, Maor MH. Hypofractionated radiotherapy for elderly or younger low-performance status glioblastoma patients: outcome and prognostic factors. Int J Radiat Oncol Biol Phys. 2003;56:519–28. doi: 10.1016/s0360-3016(02)04522-4. [DOI] [PubMed] [Google Scholar]
  • 84.Khan L, Soliman H, Sahgal A, Perry J, Xu W, Tsao MN. External beam radiation dose escalation for high grade glioma. Cochrane Database Syst Rev. 2020;5:CD011475. doi: 10.1002/14651858.CD011475.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Reyes-Botero G, Cartalat-Carel S, Chinot OL, Barrie M, Taillandier L, Beauchesne P, Catry-Thomas I, Barrière J, Guillamo JS, Fabbro M, Frappaz D, Benouaich-Amiel A, Le Rhun E, Campello C, Tennevet I, Ghiringhelli F, Tanguy ML, Mokhtari K, Honnorat J, Delattre JY. Temozolomide plus bevacizumab in elderly patients with newly diagnosed glioblastoma and poor performance status: an ANOCEF phase II trial (ATAG) Oncologist. 2018;23:524–e44. doi: 10.1634/theoncologist.2017-0689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Wick W, Platten M, Meisner C, Felsberg J, Tabatabai G, Simon M, Nikkhah G, Papsdorf K, Steinbach JP, Sabel M, Combs SE, Vesper J, Braun C, Meixensberger J, Ketter R, Mayer-Steinacker R, Reifenberger G, Weller M NOA-08 Study Group of Neuro-oncology Working Group (NOA) of German Cancer Society. Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol. 2012;13:707–15. doi: 10.1016/S1470-2045(12)70164-X. [DOI] [PubMed] [Google Scholar]
  • 87.Perry JR, Laperriere N, O’Callaghan CJ, Brandes AA, Menten J, Phillips C, Fay M, Nishikawa R, Cairncross JG, Roa W, Osoba D, Rossiter JP, Sahgal A, Hirte H, Laigle-Donadey F, Franceschi E, Chinot O, Golfinopoulos V, Fariselli L, Wick A, Feuvret L, Back M, Tills M, Winch C, Baumert BG, Wick W, Ding K, Mason WP Trial Investigators. Short-course radiation plus temozolomide in elderly patients with glioblastoma. N Engl J Med. 2017;376:1027–1037. doi: 10.1056/NEJMoa1611977. [DOI] [PubMed] [Google Scholar]
  • 88.Ohno M, Miyakita Y, Takahashi M, Igaki H, Matsushita Y, Ichimura K, Narita Y. Survival benefits of hypofractionated radiotherapy combined with temozolomide or temozolomide plus bevacizumab in elderly patients with glioblastoma aged ≥ 75 years. Radiat Oncol. 2019;14:200. doi: 10.1186/s13014-019-1389-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ney DE, Carlson JA, Damek DM, Gaspar LE, Kavanagh BD, Kleinschmidt-DeMasters BK, Waziri AE, Lillehei KO, Reddy K, Chen C. Phase II trial of hypofractionated intensity-modulated radiation therapy combined with temozolomide and bevacizumab for patients with newly diagnosed glioblastoma. J Neurooncol. 2015;122:135–143. doi: 10.1007/s11060-014-1691-z. [DOI] [PubMed] [Google Scholar]
  • 90.Omuro A, Beal K, Gutin P, Karimi S, Correa DD, Kaley TJ, DeAngelis LM, Chan TA, Gavrilovic IT, Nolan C, Hormigo A, Lassman AB, Mellinghoff I, Grommes C, Reiner AS, Panageas KS, Baser RE, Tabar V, Pentsova E, Sanchez J, Barradas-Panchal R, Zhang J, Faivre G, Brennan CW, Abrey LE, Huse JT. Phase II study of bevacizumab, temozolomide, and hypofractionated stereotactic radiotherapy for newly diagnosed glioblastoma. Clin Cancer Res. 2014;20:5023–5031. doi: 10.1158/1078-0432.CCR-14-0822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Iuchi T, Hatano K, Kodama T, Sakaida T, Yokoi S, Kawasaki K, Hasegawa Y, Hara R. Phase 2 trial of hypofractionated high-dose intensity modulated radiation therapy with concurrent and adjuvant temozolomide for newly diagnosed glioblastoma. Int J Radiat Oncol Biol Phys. 2014;88:793–800. doi: 10.1016/j.ijrobp.2013.12.011. [DOI] [PubMed] [Google Scholar]
  • 92.Reddy K, Gaspar LE, Kavanagh BD, Chen C. Hypofractionated intensity-modulated radiotherapy with temozolomide chemotherapy may alter the patterns of failure in patients with glioblastoma multiforme. J Med Imaging Radiat Oncol. 2014;58:714–21. doi: 10.1111/1754-9485.12185. [DOI] [PubMed] [Google Scholar]
  • 93.Carlson JA, Reddy K, Gaspar LE, Ney D, Kavanagh BD, Damek D, Lillehei K, Chen C. Hypofractionated-intensity modulated radiotherapy (hypo-IMRT) and temozolomide (TMZ) with or without bevacizumab (BEV) for newly diagnosed glioblastoma multiforme (GBM): a comparison of two prospective phase II trials. J Neurooncol. 2015;123:251–257. doi: 10.1007/s11060-015-1791-4. [DOI] [PubMed] [Google Scholar]
  • 94.Gleeson JP, Keane F, Keegan NM, Mammadov E, Harrold E, Alhusaini A, Harte J, Eakin-Love A, O’Halloran PJ, MacNally S, Hennessy BT, Breathnach OS, Grogan L, Morris PG. Similar overall survival with reduced vs. standard dose bevacizumab monotherapy in progressive glioblastoma. Cancer Med. 2020;9:469–475. doi: 10.1002/cam4.2616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Levin VA, Mendelssohn ND, Chan J, Stovall MC, Peak SJ, Yee JL, Hui RL, Chen DM. Impact of bevacizumab administered dose on overall survival of patients with progressive glioblastoma. J Neurooncol. 2015;122:145–150. doi: 10.1007/s11060-014-1693-x. [DOI] [PubMed] [Google Scholar]
  • 96.Raizer JJ, Grimm S, Chamberlain MC, Nicholas MK, Chandler JP, Muro K, Dubner S, Rademaker AW, Renfrow J, 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. 2010;116:5297–5305. doi: 10.1002/cncr.25462. [DOI] [PubMed] [Google Scholar]
  • 97.Bokstein F, Shpigel S, Blumenthal DT. Treatment with bevacizumab and irinotecan for recurrent high-grade glial tumors. Cancer. 2008;112:2267–2273. doi: 10.1002/cncr.23401. [DOI] [PubMed] [Google Scholar]
  • 98.Wong ET, Gautam S, Malchow C, Lun M, Pan E, Brem S. Bevacizumab for recurrent glioblastoma multiforme: a meta-analysis. J Natl Compr Canc Netw. 2011;9:403–407. doi: 10.6004/jnccn.2011.0037. [DOI] [PubMed] [Google Scholar]
  • 99.Menon NS, Shah MJ, Gupta T, Sridhar E, Chatterjee A, Dasgupta A, Sahu A, Sawant R, Kapu V, Trikha M, Shah A, Peelay ZR, Puranik A, Dev I, Jadhav M, Moiyadi A, Shetty P, Sahay A, Singh VK, Patil VM. VAMANA: a phase 2 study of low-dose bevacizumab plus CCNU in relapsed/recurrent glioblastoma. JCO. 2024;42:LBA2064. [Google Scholar]
  • 100.Rubenstein JL, Kim J, Ozawa T, Zhang M, Westphal M, Deen DF, Shuman MA. Anti-VEGF antibody treatment of glioblastoma prolongs survival but results in increased vascular cooption. Neoplasia. 2000;2:306–314. doi: 10.1038/sj.neo.7900102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Fortin D, Morin PA, Belzile F, Mathieu D, Paré FM. Intra-arterial carboplatin as a salvage strategy in the treatment of recurrent glioblastoma multiforme. J Neurooncol. 2014;119:397–403. doi: 10.1007/s11060-014-1504-4. [DOI] [PubMed] [Google Scholar]
  • 102.Lim J, Santo BA, Baig AA, Ciecierska SK, Donnelly B, Balghonaim S, Levy BR, Jaikumar V, Levy EI, Tutino VM, Siddiqui AH. Efficacy of intra-arterial carboplatin and bevacizumab in the C6 rat glioma model of glioblastoma multiforme. J Neurointerv Surg. 2025 doi: 10.1136/jnis-2024-021789. jnis-2024-021789. [DOI] [PubMed] [Google Scholar]
  • 103.Patel NV, Wong T, Fralin SR, Li M, McKeown A, Gruber D, D’Amico RS, Patsalides A, Tsiouris A, Stefanov DG, Flores O, Zlochower A, Filippi CG, Ortiz R, Langer DJ, Boockvar JA. Repeated superselective intraarterial bevacizumab after blood brain barrier disruption for newly diagnosed glioblastoma: a phase I/II clinical trial. J Neurooncol. 2021;155:117–124. doi: 10.1007/s11060-021-03851-2. [DOI] [PubMed] [Google Scholar]
  • 104.Kappel AD, Jha R, Guggilapu S, Smith WJ, Feroze AH, Dmytriw AA, Vicenty-Padilla J, Alcedo Guardia RE, Gessler FA, Patel NJ, Du R, See AP, Peruzzi PP, Aziz-Sultan MA, Bernstock JD. Endovascular applications for the management of high-grade gliomas in the modern era. Cancers (Basel) 2024;16:1594. doi: 10.3390/cancers16081594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Di Filippo LD, Lobato Duarte J, Hofstätter Azambuja J, Isler Mancuso R, Tavares Luiz M, Hugo Sousa Araújo V, Delbone Figueiredo I, Barretto-de-Souza L, Miguel Sábio R, Sasso-Cerri E, Martins Baviera A, Crestani CC, Teresinha Ollala Saad S, Chorilli M. Glioblastoma multiforme targeted delivery of docetaxel using bevacizumab-modified nanostructured lipid carriers impair in vitro cell growth and in vivo tumor progression. Int J Pharm. 2022;618:121682. doi: 10.1016/j.ijpharm.2022.121682. [DOI] [PubMed] [Google Scholar]
  • 106.Sousa F, Costa-Pereira AI, Cruz A, Ferreira FJ, Gouveia M, Bessa J, Sarmento B, Travasso RDM, Mendes Pinto I. Intratumoral VEGF nanotrapper reduces gliobastoma vascularization and tumor cell mass. J Control Release. 2021;339:381–390. doi: 10.1016/j.jconrel.2021.09.031. [DOI] [PubMed] [Google Scholar]
  • 107.de Cristo Soares Alves A, Lavayen V, de Fraga Dias A, Bruinsmann FA, Scholl JN, Cé R, Visioli F, Oliveira Battastini AM, Stanisçuaski Guterres S, Figueiró F, Raffin Pohlmann A. EGFRvIII peptide nanocapsules and bevacizumab nanocapsules: a nose-to-brain multitarget approach against glioblastoma. Nanomedicine (Lond) 2021;16:1775–1790. doi: 10.2217/nnm-2021-0169. [DOI] [PubMed] [Google Scholar]
  • 108.Chu L, Sun Y, Tang X, Duan X, Zhao Y, Xia H, Xu L, Zhang P, Sun K, Yang G, Wang A. The tumor-derived exosomes enhanced bevacizumab across the blood-brain barrier for antiangiogenesis therapy against glioblastoma. Mol Pharm. 2025;22:972–983. doi: 10.1021/acs.molpharmaceut.4c01227. [DOI] [PubMed] [Google Scholar]
  • 109.Norden AD, Young GS, Setayesh K, Muzikansky A, Klufas R, Ross GL, Ciampa AS, Ebbeling LG, Levy B, Drappatz J, Kesari S, Wen PY. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70:779–787. doi: 10.1212/01.wnl.0000304121.57857.38. [DOI] [PubMed] [Google Scholar]
  • 110.Grill J, Massimino M, Bouffet E, Azizi AA, McCowage G, Cañete A, Saran F, Le Deley MC, Varlet P, Morgan PS, Jaspan T, Jones C, Giangaspero F, Smith H, Garcia J, Elze MC, Rousseau RF, Abrey L, Hargrave D, Vassal G. Phase II, open-label, randomized, multicenter trial (HERBY) of bevacizumab in pediatric patients with newly diagnosed high-grade glioma. J. Clin. Oncol. 2018;36:951–958. doi: 10.1200/JCO.2017.76.0611. [DOI] [PubMed] [Google Scholar]
  • 111.Chandra A, Jahangiri A, Chen W, Nguyen AT, Yagnik G, Pereira MP, Jain S, Garcia JH, Shah SS, Wadhwa H, Joshi RS, Weiss J, Wolf KJ, Lin JG, Müller S, Rick JW, Diaz AA, Gilbert LA, Kumar S, Aghi MK. Clonal ZEB1-driven mesenchymal transition promotes targetable oncologic antiangiogenic therapy resistance. Cancer Res. 2020;80:1498–1511. doi: 10.1158/0008-5472.CAN-19-1305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Fu M, Hussain A, Dong Y, Fei Y. A retrospective analysis of GSE84010: cell adhesion molecules might contribute to bevacizumab resistance in glioblastoma. J Clin Neurosci. 2021;86:110–115. doi: 10.1016/j.jocn.2021.01.019. [DOI] [PubMed] [Google Scholar]
  • 113.Lim YC, Jensen KE, Aguilar-Morante D, Vardouli L, Vitting-Seerup K, Gimple RC, Wu Q, Pedersen H, Elbaek KJ, Gromova I, Ihnatko R, Kristensen BW, Petersen JK, Skjoth-Rasmussen J, Flavahan W, Rich JN, Hamerlik P. Non-metabolic functions of phosphofructokinase-1 orchestrate tumor cellular invasion and genome maintenance under bevacizumab therapy. Neuro Oncol. 2023;25:248–260. doi: 10.1093/neuonc/noac135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.DeLay M, Jahangiri A, Carbonell WS, Hu YL, Tsao S, Tom MW, Paquette J, Tokuyasu TA, Aghi MK. Microarray analysis verifies two distinct phenotypes of glioblastomas resistant to antiangiogenic therapy. Clin Cancer Res. 2012;18:2930–2942. doi: 10.1158/1078-0432.CCR-11-2390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Cho SJ, Kim HS, Suh CH, Park JE. Radiological recurrence patterns after bevacizumab treatment of recurrent high-grade glioma: a systematic review and meta-analysis. Korean J Radiol. 2020;21:908–918. doi: 10.3348/kjr.2019.0898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Hovey EJ, Field KM, Rosenthal MA, Barnes EH, Cher L, Nowak AK, Wheeler H, Sawkins K, Livingstone A, Phal P, Goh C, Simes J CABARET/COGNO investigators. Continuing or ceasing bevacizumab beyond progression in recurrent glioblastoma: an exploratory randomized phase II trial. Neurooncol Pract. 2017;4:171–181. doi: 10.1093/nop/npw025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Lucio-Eterovic AK, Piao Y, de Groot JF. Mediators of glioblastoma resistance and invasion during antivascular endothelial growth factor therapy. Clin Cancer Res. 2009;15:4589–4599. doi: 10.1158/1078-0432.CCR-09-0575. [DOI] [PubMed] [Google Scholar]
  • 118.Pham K, Luo D, Siemann DW, Law BK, Reynolds BA, Hothi P, Foltz G, Harrison JK. VEGFR inhibitors upregulate CXCR4 in VEGF receptor-expressing glioblastoma in a TGFβR signaling-dependent manner. Cancer Lett. 2015;360:60–67. doi: 10.1016/j.canlet.2015.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Lu KV, Chang JP, Parachoniak CA, Pandika MM, Aghi MK, Meyronet D, Isachenko N, Fouse SD, Phillips JJ, Cheresh DA, Park M, Bergers G. VEGF inhibits tumor cell invasion and mesenchymal transition through a MET/VEGFR2 complex. Cancer Cell. 2012;22:21–35. doi: 10.1016/j.ccr.2012.05.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Schaub C, Kebir S, Junold N, Hattingen E, Schäfer N, Steinbach JP, Weyerbrock A, Hau P, Goldbrunner R, Niessen M, Mack F, Stuplich M, Tzaridis T, Bähr O, Kortmann RD, Schlegel U, Schmidt-Graf F, Rohde V, Braun C, Hänel M, Sabel M, Gerlach R, Krex D, Belka C, Vatter H, Proescholdt M, Herrlinger U, Glas M. Tumor growth patterns of MGMT-non-methylated glioblastoma in the randomized GLARIUS trial. J Cancer Res Clin Oncol. 2018;144:1581–1589. doi: 10.1007/s00432-018-2671-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Wick W, Chinot OL, Bendszus M, Mason W, Henriksson R, Saran F, Nishikawa R, Revil C, Kerloeguen Y, Cloughesy T. Evaluation of pseudoprogression rates and tumor progression patterns in a phase III trial of bevacizumab plus radiotherapy/temozolomide for newly diagnosed glioblastoma. Neuro Oncol. 2016;18:1434–1441. doi: 10.1093/neuonc/now091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Pope WB, Xia Q, Paton VE, Das A, Hambleton J, Kim HJ, Huo J, Brown MS, Goldin J, Cloughesy T. Patterns of progression in patients with recurrent glioblastoma treated with bevacizumab. Neurology. 2011;76:432–437. doi: 10.1212/WNL.0b013e31820a0a8a. [DOI] [PubMed] [Google Scholar]
  • 123.Nowosielski M, Ellingson BM, Chinot OL, Garcia J, Revil C, Radbruch A, Nishikawa R, Mason WP, Henriksson R, Saran F, Kickingereder P, Platten M, Sandmann T, Abrey LE, Cloughesy TF, Bendszus M, Wick W. Radiologic progression of glioblastoma under therapy-an exploratory analysis of AVAglio. Neuro Oncol. 2018;20:557–566. doi: 10.1093/neuonc/nox162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Franceschi E, Lamberti G, Paccapelo A, Di Battista M, Genestreti G, Minichillo S, Mura A, Bartolini S, Agati R, Brandes AA. Third-line therapy in recurrent glioblastoma: is it another chance for bevacizumab? J Neurooncol. 2018;139:383–388. doi: 10.1007/s11060-018-2873-x. [DOI] [PubMed] [Google Scholar]
  • 125.Brandes AA, Gil-Gil M, Saran F, Carpentier AF, Nowak AK, Mason W, Zagonel V, Dubois F, Finocchiaro G, Fountzilas G, Cernea DM, Chinot O, Anghel R, Ghiringhelli F, Beauchesne P, Lombardi G, Franceschi E, Makrutzki M, Mpofu C, Urban HJ, Pichler J. A randomized phase II trial (TAMIGA) evaluating the efficacy and safety of continuous bevacizumab through multiple lines of treatment for recurrent glioblastoma. Oncologist. 2019;24:521–528. doi: 10.1634/theoncologist.2018-0290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Schaub C, Schäfer N, Mack F, Stuplich M, Kebir S, Niessen M, Tzaridis T, Banat M, Vatter H, Waha A, Herrlinger U, Glas M. The earlier the better? Bevacizumab in the treatment of recurrent MGMT-non-methylated glioblastoma. J Cancer Res Clin Oncol. 2016;142:1825–1829. doi: 10.1007/s00432-016-2187-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Rahman R, Hempfling K, Norden AD, Reardon DA, Nayak L, Rinne ML, Beroukhim R, Doherty L, Ruland S, Rai A, Rifenburg J, LaFrankie D, Alexander BM, Huang RY, Wen PY, Lee EQ. Retrospective study of carmustine or lomustine with bevacizumab in recurrent glioblastoma patients who have failed prior bevacizumab. Neuro Oncol. 2014;16:1523–1529. doi: 10.1093/neuonc/nou118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Shi W, Blomain ES, Siglin J, Palmer JD, Dan T, Wang Y, Werner-Wasik M, Glass J, Kim L, Bar Ad V, Bhamidipati D, Evans JJ, Judy K, Farrell CJ, Andrews DW. Salvage fractionated stereotactic re-irradiation (FSRT) for patients with recurrent high grade gliomas progressed after bevacizumab treatment. J Neurooncol. 2018;137:171–177. doi: 10.1007/s11060-017-2709-0. [DOI] [PubMed] [Google Scholar]
  • 129.Dixit KS, Sachdev S, Amidei C, Kumthekar P, Kruser TJ, Gondi V, Grimm S, Lukas RV, Nicholas MK, Chmura SJ, Fought AJ, Mehta M, Raizer JJ. A multi-center prospective study of re-irradiation with bevacizumab and temozolomide in patients with bevacizumab refractory recurrent high-grade gliomas. J Neurooncol. 2021;155:297–306. doi: 10.1007/s11060-021-03875-8. [DOI] [PubMed] [Google Scholar]
  • 130.You WC, Lee HD, Pan HC, Chen HC. Re-irradiation combined with bevacizumab for recurrent glioblastoma beyond bevacizumab failure: Survival outcomes and prognostic factors. Sci Rep. 2023;13:9442. doi: 10.1038/s41598-023-36290-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Yonezawa H, Ohno M, Igaki H, Miyakita Y, Takahashi M, Tamura Y, Shima S, Matsushita Y, Ichimura K, Narita Y. Outcomes of salvage fractionated re-irradiation combined with bevacizumab for recurrent high-grade gliomas that progressed after bevacizumab treatment*. Jpn J Clin Oncol. 2021;51:1028–1035. doi: 10.1093/jjco/hyab063. [DOI] [PubMed] [Google Scholar]
  • 132.Bergman D, Modh A, Schultz L, Snyder J, Mikkelsen T, Shah M, Ryu S, Siddiqui MS, Walbert T. Randomized prospective trial of fractionated stereotactic radiosurgery with chemotherapy versus chemotherapy alone for bevacizumab-resistant high-grade glioma. J Neurooncol. 2020;148:353–361. doi: 10.1007/s11060-020-03526-4. [DOI] [PubMed] [Google Scholar]
  • 133.Iwamoto FM, Tanguturi SK, Nayak L, Wang TJ, Desai A, Lustig RA, Bagley S, Wong ET, Hertan LM, McCluskey C, Hayden J, Muzikansky A, Nakhawa S, Japo J, Bossi CC, Meylan M, Tian Y, Barlow GL, Speliakos P, Ayoub G, Meredith DM, Ligon KL, Haas-Kogan D, Huang K, Wucherpfennig KW, Wen PY, Reardon DA. Re-irradiation plus pembrolizumab: a phase ii study for patients with recurrent glioblastoma. Clin Cancer Res. 2025;31:316–327. doi: 10.1158/1078-0432.CCR-24-1629. [DOI] [PubMed] [Google Scholar]
  • 134.Pasqualetti F, Pace A, Gonnelli A, Villani V, Cantarella M, Delishaj D, Vivaldi C, Molinari A, Montrone S, Pellerino A, Franchino F, Baldaccini D, Lombardi G, Lolli I, Catania F, Bazzoli E, Morganti R, Fabi A, Zagonel V, Bocci G, Fabrini MG, Rudà R, Soffietti R, Paiar F. Single-agent bevacizumab in recurrent glioblastoma after second-line chemotherapy with fotemustine: the experience of the italian association of neuro-oncology. Am J Clin Oncol. 2018;41:1272–1275. doi: 10.1097/COC.0000000000000464. [DOI] [PubMed] [Google Scholar]
  • 135.Wenger KJ, Wagner M, You SJ, Franz K, Harter PN, Burger MC, Voss M, Ronellenfitsch MW, Fokas E, Steinbach JP, Bähr O. Bevacizumab as a last-line treatment for glioblastoma following failure of radiotherapy, temozolomide and lomustine. Oncol Lett. 2017;14:1141–1146. doi: 10.3892/ol.2017.6251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Brenner AJ, Floyd J, Fichtel L, Michalek J, Kanakia KP, Huang S, Reardon D, Wen PY, Lee EQ. Phase 2 trial of hypoxia activated evofosfamide (TH302) for treatment of recurrent bevacizumab-refractory glioblastoma. Sci Rep. 2021;11:2306. doi: 10.1038/s41598-021-81841-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Ahluwalia MS, Rogers LR, Chaudhary R, Newton H, Ozair A, Khosla AA, Nixon AB, Adams BJ, Seon BK, Peereboom DM, Theuer CP. Endoglin inhibitor TRC105 with or without bevacizumab for bevacizumab-refractory glioblastoma (ENDOT): a multicenter phase II trial. Commun Med (Lond) 2023;3:120. doi: 10.1038/s43856-023-00347-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Ahluwalia MS, Ozair A, Drappatz J, Ye X, Peng S, Lee M, Rath S, Dhruv H, Hao Y, Berens ME, Walbert T, Holdhoff M, Lesser GJ, Cloughesy TF, Sloan AE, Takebe N, Couce M, Peereboom DM, Nabors B, Wen PY, Grossman SA, Rogers LR. Evaluating the base excision repair inhibitor TRC102 and temozolomide for patients with recurrent glioblastoma in the phase 2 adult brain tumor consortium trial BERT. Clin Cancer Res. 2024;30:3167–3178. doi: 10.1158/1078-0432.CCR-23-4098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Takei J, Fukasawa N, Tanaka T, Yamamoto Y, Tamura R, Sasaki H, Akasaki Y, Kamata Y, Murahashi M, Shimoda M, Murayama Y. Impact of neoadjuvant bevacizumab on neuroradiographic response and histological findings related to tumor stemness and the hypoxic tumor microenvironment in glioblastoma: paired comparison between newly diagnosed and recurrent glioblastomas. Front Oncol. 2022;12:898614. doi: 10.3389/fonc.2022.898614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Scheer KG, Ebert LM, Samuel MS, Bonder CS, Gomez GA. Bevacizumab-induced hypertension in glioblastoma patients and its potential as a modulator of treatment response. Hypertension. 2023;80:1590–1597. doi: 10.1161/HYPERTENSIONAHA.123.21119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Hiller-Vallina S, Mondejar-Ruescas L, Caamaño-Moreno M, Cómitre-Mariano B, Alcivar-López D, Sepulveda JM, Hernández-Laín A, Pérez-Núñez Á, Segura-Collar B, Gargini R. Sexual-biased necroinflammation is revealed as a predictor of bevacizumab benefit in glioblastoma. Neuro Oncol. 2024;26:1213–1227. doi: 10.1093/neuonc/noae033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Joshkon A, Tabouret E, Traboulsi W, Bachelier R, Simoncini S, Roffino S, Jiguet-Jiglaire C, Badran B, Guillet B, Foucault-Bertaud A, Leroyer AS, Dignat-George F, Chinot O, Fayyad-Kazan H, Bardin N, Blot-Chabaud M. Soluble CD146, a biomarker and a target for preventing resistance to anti-angiogenic therapy in glioblastoma. Acta Neuropathol Commun. 2022;10:151. doi: 10.1186/s40478-022-01451-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Lallemand C, Ferrando-Miguel R, Auer M, Iglseder S, Czech T, Gaber-Wagener A, Di Pauli F, Deisenhammer F, Tovey MG. Quantification of bevacizumab activity following treatment of patients with ovarian cancer or glioblastoma. Front Immunol. 2020;11:515556. doi: 10.3389/fimmu.2020.515556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Carvalho B, Lopes JM, Silva R, Peixoto J, Leitão D, Soares P, Fernandes AC, Linhares P, Vaz R, Lima J. The role of c-Met and VEGFR2 in glioblastoma resistance to bevacizumab. Sci Rep. 2021;11:6067. doi: 10.1038/s41598-021-85385-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Schell M, Pflüger I, Brugnara G, Isensee F, Neuberger U, Foltyn M, Kessler T, Sahm F, Wick A, Nowosielski M, Heiland S, Weller M, Platten M, Maier-Hein KH, Von Deimling A, Van Den Bent MJ, Gorlia T, Wick W, Bendszus M, Kickingereder P. Validation of diffusion MRI phenotypes for predicting response to bevacizumab in recurrent glioblastoma: post-hoc analysis of the EORTC-26101 trial. Neuro Oncol. 2020;22:1667–1676. doi: 10.1093/neuonc/noaa120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Gilbert MR, Pugh SL, Aldape K, Sorensen AG, Mikkelsen T, Penas-Prado M, Bokstein F, Kwok Y, Lee RJ, Mehta M. NRG oncology RTOG 0625: a randomized phase II trial of bevacizumab with either irinotecan or dose-dense temozolomide in recurrent glioblastoma. J Neurooncol. 2017;131:193–199. doi: 10.1007/s11060-016-2288-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Desjardins A, Herndon JE, McSherry F, Ravelo A, Lipp ES, Healy P, Peters KB, Sampson JH, Randazzo D, Sommer N, Friedman AH, Friedman HS. Single-institution retrospective review of patients with recurrent glioblastoma treated with bevacizumab in clinical practice. Health Sci Rep. 2019;2:e114. doi: 10.1002/hsr2.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Cloughesy TF, Petrecca K, Walbert T, Butowski N, Salacz M, Perry J, Damek D, Bota D, Bettegowda C, Zhu JJ, Iwamoto F, Placantonakis D, Kim L, Elder B, Kaptain G, Cachia D, Moshel Y, Brem S, Piccioni D, Landolfi J, Chen CC, Gruber H, Rao AR, Hogan D, Accomando W, Ostertag D, Montellano TT, Kheoh T, Kabbinavar F, Vogelbaum MA. Effect of vocimagene amiretrorepvec in combination with flucytosine vs standard of care on survival following tumor resection in patients with recurrent high-grade glioma: a randomized clinical trial. JAMA Oncol. 2020;6:1939–1946. doi: 10.1001/jamaoncol.2020.3161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Detti B, Scoccianti S, Teriaca MA, Maragna V, Lorenzetti V, Lucidi S, Bellini C, Greto D, Desideri I, Livi L. Bevacizumab in recurrent high-grade glioma: a single institution retrospective analysis on 92 patients. Radiol Med. 2021;126:1249–1254. doi: 10.1007/s11547-021-01381-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Smolenschi C, Rassy E, Pallud J, Dezamis E, Copaciu R, Parker F, Garcia G, Lezghed N, Colomba E, Khettab M, Ammari S, Fekhi M, Martanovschi L, Benadhou L, Knafo S, Guyon D, Cheaib B, Dhermain F, Dumont SN. Bevacizumab in real-life patients with recurrent glioblastoma: benefit or futility? J Neurol. 2023;270:2702–2714. doi: 10.1007/s00415-023-11600-w. [DOI] [PubMed] [Google Scholar]
  • 151.Witte HM, Riecke A, Steinestel K, Schulz C, Küchler J, Gebauer N, Tronnier V, Leppert J. The addition of chloroquine and bevacizumab to standard radiochemotherapy for recurrent glioblastoma multiforme. Br J Neurosurg. 2024;38:404–410. doi: 10.1080/02688697.2021.1884648. [DOI] [PubMed] [Google Scholar]
  • 152.Lee Y, Lee E, Roh TH, Kim SH. Bevacizumab alone versus bevacizumab plus irinotecan in patients with recurrent glioblastoma: a nationwide population-based study. J Korean Med Sci. 2024;39:e244. doi: 10.3346/jkms.2024.39.e244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Puduvalli VK, Wu J, Yuan Y, Armstrong TS, Vera E, Wu J, Xu J, Giglio P, Colman H, Walbert T, Raizer J, Groves MD, Tran D, Iwamoto F, Avgeropoulos N, Paleologos N, Fink K, Peereboom D, Chamberlain M, Merrell R, Penas Prado M, Yung WKA, Gilbert MR. A Bayesian adaptive randomized phase II multicenter trial of bevacizumab with or without vorinostat in adults with recurrent glioblastoma. Neuro Oncol. 2020;22:1505–1515. doi: 10.1093/neuonc/noaa062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Lee EQ, Zhang P, Wen PY, Gerstner ER, Reardon DA, Aldape KD, deGroot JF, Pan E, Raizer JJ, Kim LJ, Chmura SJ, Robins HI, Connelly JM, Battiste JD, Villano JL, Wagle N, Merrell RT, Wendland MM, Mehta MP. NRG/RTOG 1122: a phase 2, double-blinded, placebo-controlled study of bevacizumab with and without trebananib in patients with recurrent glioblastoma or gliosarcoma. Cancer. 2020;126:2821–2828. doi: 10.1002/cncr.32811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.McCrea HJ, Ivanidze J, O’Connor A, Hersh EH, Boockvar JA, Gobin YP, Knopman J, Greenfield JP. Intraarterial delivery of bevacizumab and cetuximab utilizing blood-brain barrier disruption in children with high-grade glioma and diffuse intrinsic pontine glioma: results of a phase I trial. J Neurosurg Pediatr. 2021;28:371–379. doi: 10.3171/2021.3.PEDS20738. [DOI] [PubMed] [Google Scholar]
  • 156.Galanis E, Anderson SK, Twohy E, Butowski NA, Hormigo A, Schiff D, Omuro A, Jaeckle KA, Kumar S, Kaufmann TJ, Geyer S, Kumthekar PU, Campian J, Giannini C, Buckner JC, Wen PY. Phase I/randomized phase II trial of TRC105 plus bevacizumab versus bevacizumab in recurrent glioblastoma: North Central Cancer Treatment Group N1174 (Alliance) Neurooncol Adv. 2022;4:vdac041. doi: 10.1093/noajnl/vdac041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Cardona AF, Jaramillo-Velásquez D, Ruiz-Patiño A, Polo C, Jiménez E, Hakim F, Gómez D, Ramón JF, Cifuentes H, Mejía JA, Salguero F, Ordoñez C, Muñoz Á, Bermúdez S, Useche N, Pineda D, Ricaurte L, Zatarain-Barrón ZL, Rodríguez J, Avila J, Rojas L, Jaller E, Sotelo C, Garcia-Robledo JE, Santoyo N, Rolfo C, Rosell R, Arrieta O. Efficacy of osimertinib plus bevacizumab in glioblastoma patients with simultaneous EGFR amplification and EGFRvIII mutation. J Neurooncol. 2021;154:353–364. doi: 10.1007/s11060-021-03834-3. [DOI] [PubMed] [Google Scholar]
  • 158.Bota DA, Taylor TH, Lomeli N, Kong XT, Fu BD, Schönthal AH, Singer S, Blumenthal DT, Senecal FM, Linardou H, Rokas E, Antoniou DG, Schijns VEJC, Chen TC, Elliot J, Stathopoulos A. A prospective, cohort study of SITOIGANAP to treat glioblastoma when given in combination with granulocyte-macrophage colony-stimulating factor/cyclophosphamide/bevacizumab/nivolumab or granulocyte-macrophage colony-stimulating factor/cyclophosphamide/bevacizumab/pembrolizumab in patients who failed prior treatment with surgical resection, radiation, and temozolomide. Front Oncol. 2022;12:934638. doi: 10.3389/fonc.2022.934638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Ahluwalia MS, Rauf Y, Li H, Wen PY, Peereboom DM, Reardon DA. Randomized phase 2 study of nivolumab (nivo) plus either standard or reduced dose bevacizumab (bev) in recurrent glioblastoma (rGBM) J. Clin. Oncol. 2021;39:2015–2015. [Google Scholar]
  • 160.She L, Su L, Liu C. Bevacizumab combined with re-irradiation in recurrent glioblastoma. Front Oncol. 2022;12:961014. doi: 10.3389/fonc.2022.961014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Zhang JF, Okai B, Iovoli A, Goulenko V, Attwood K, Lim J, Hess RM, Abad AP, Prasad D, Fenstermaker RA. Bevacizumab and gamma knife radiosurgery for first-recurrence glioblastoma. J Neurooncol. 2024;166:89–98. doi: 10.1007/s11060-023-04524-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Mantica M, Drappatz J, Lieberman F, Hadjipanayis CG, Lunsford LD, Niranjan A. Phase II study of border zone stereotactic radiosurgery with bevacizumab in patients with recurrent or progressive glioblastoma multiforme. J Neurooncol. 2023;164:179–190. doi: 10.1007/s11060-023-04398-0. [DOI] [PubMed] [Google Scholar]
  • 163.Morris SL, Zhu P, Rao M, Martir M, Zhu JJ, Hsu S, Ballester LY, Day AL, Tandon N, Kim DH, Shepard S, Blanco A, Esquenazi Y. Gamma knife stereotactic radiosurgery in combination with bevacizumab for recurrent glioblastoma. World Neurosurg. 2019;127:e523–e533. doi: 10.1016/j.wneu.2019.03.193. [DOI] [PubMed] [Google Scholar]
  • 164.Abbassy M, Missios S, Barnett GH, Brewer C, Peereboom DM, Ahluwalia M, Neyman G, Chao ST, Suh JH, Vogelbaum MA. Phase I trial of radiosurgery dose escalation plus bevacizumab in patients with recurrent/progressive glioblastoma. Neurosurgery. 2018;83:385–392. doi: 10.1093/neuros/nyx369. [DOI] [PubMed] [Google Scholar]
  • 165.Herrlinger U, Schäfer N, Steinbach JP, Weyerbrock A, Hau P, Goldbrunner R, Friedrich F, Rohde V, Ringel F, Schlegel U, Sabel M, Ronellenfitsch MW, Uhl M, Maciaczyk J, Grau S, Schnell O, Hänel M, Krex D, Vajkoczy P, Gerlach R, Kortmann RD, Mehdorn M, Tüttenberg J, Mayer-Steinacker R, Fietkau R, Brehmer S, Mack F, Stuplich M, Kebir S, Kohnen R, Dunkl E, Leutgeb B, Proescholdt M, Pietsch T, Urbach H, Belka C, Stummer W, Glas M. Bevacizumab plus irinotecan versus temozolomide in newly diagnosed O6-methylguanine-DNA methyltransferase nonmethylated glioblastoma: the randomized GLARIUS trial. JCO. 2016;34:1611–1619. doi: 10.1200/JCO.2015.63.4691. [DOI] [PubMed] [Google Scholar]
  • 166.Nagane M, Ichimura K, Onuki R, Narushima D, Honda-Kitahara M, Satomi K, Tomiyama A, Arai Y, Shibata T, Narita Y, Uzuka T, Nakamura H, Nakada M, Arakawa Y, Ohnishi T, Mukasa A, Tanaka S, Wakabayashi T, Aoki T, Aoki S, Shibui S, Matsutani M, Ishizawa K, Yokoo H, Suzuki H, Morita S, Kato M, Nishikawa R. Bevacizumab beyond progression for newly diagnosed glioblastoma (BIOMARK): phase II safety, efficacy and biomarker study. Cancers (Basel) 2022;14:5522. doi: 10.3390/cancers14225522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Matsuda KI, Sakurada K, Nemoto K, Kayama T, Sonoda Y. Treatment outcomes of hypofractionated radiotherapy combined with temozolomide followed by bevacizumab salvage therapy in glioblastoma patients aged > 75 years. Int J Clin Oncol. 2018;23:820–825. doi: 10.1007/s10147-018-1298-z. [DOI] [PubMed] [Google Scholar]
  • 168.Kanamori M, Shibahara I, Shimoda Y, Akiyama Y, Beppu T, Ohba S, Enomoto T, Ono T, Mitobe Y, Hanihara M, Mineharu Y, Ishida J, Asano K, Yoshida Y, Natsumeda M, Nomura S, Abe T, Yonezawa H, Katakura R, Shibui S, Kuroiwa T, Suzuki H, Takei H, Matsushita H, Saito R, Arakawa Y, Sonoda Y, Hirose Y, Kumabe T, Yamaguchi T, Endo H, Tominaga T. Efficacy and safety of carmustine wafers, followed by radiation, temozolomide, and bevacizumab therapy, for newly diagnosed glioblastoma with maximal resection. Int J Clin Oncol. 2025;30:51–61. doi: 10.1007/s10147-024-02650-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Tanaka T, Tamura R, Takei J, Morimoto Y, Teshigawara A, Yamamoto Y, Imai R, Kuranari Y, Tohmoto K, Hasegawa Y, Akasaki Y, Murayama Y, Miyake K, Sasaki H. An exploratory prospective phase II study of preoperative neoadjuvant bevacizumab and temozolomide for newly diagnosed glioblastoma. J Neurooncol. 2024;166:557–567. doi: 10.1007/s11060-023-04544-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Strauss JD, Gilbert MR, Mehta M, Li A, Zhou R, Bondy ML, Sulman EP, Yuan Y, Liu Y, Vera E, Wendland MM, Stieber VW, Puduvalli VK, Choi S, Martinez NL, Robins HI, Hunter GK, Lin CF, Guedes VA, Richard MA, Pugh SL, Armstrong TS, Scheurer ME. Clinical and genetic markers of vascular toxicity in glioblastoma patients: insights from NRG Oncology RTOG-0825. Neuro Oncol. 2025;27:767–778. doi: 10.1093/neuonc/noae234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Kessler T, Schrimpf D, Doerner L, Hai L, Kaulen LD, Ito J, van den Bent M, Taphoorn M, Brandes AA, Idbaih A, Dômont J, Clement PM, Campone M, Bendszus M, von Deimling A, Sahm F, Platten M, Wick W, Wick A. Prognostic markers of DNA methylation and next-generation sequencing in progressive glioblastoma from the EORTC-26101 trial. Clin Cancer Res. 2023;29:3892–3900. doi: 10.1158/1078-0432.CCR-23-0926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Kim M, Park JE, Yoon SK, Kim N, Kim YH, Kim JH, Kim HS. Vessel size and perfusion-derived vascular habitat refines prediction of treatment failure to bevacizumab in recurrent glioblastomas: validation in a prospective cohort. Eur Radiol. 2023;33:4475–4485. doi: 10.1007/s00330-022-09164-w. [DOI] [PubMed] [Google Scholar]
  • 173.Breda-Yepes M, Rodríguez-Hernández LA, Gómez-Figueroa E, Mondragón-Soto MG, Arellano-Flores G, Hernández-Hernández A, Rodríguez-Rubio HA, Martínez P, Reyes-Moreno I, Álvaro-Heredia JA, Gutiérrez Aceves GA, Villanueva-Castro E, Sangrador-Deitos MV, Alonso-Vanegas M, Guerrero-Juárez V, González-Aguilar A. Relative cerebral blood volume as response predictor in the treatment of recurrent glioblastoma with anti-angiogenic therapy. Clin Neurol Neurosurg. 2023;233:107904. doi: 10.1016/j.clineuro.2023.107904. [DOI] [PubMed] [Google Scholar]
  • 174.Ellingson BM, Hagiwara A, Morris CJ, Cho NS, Oshima S, Sanvito F, Oughourlian TC, Telesca D, Raymond C, Abrey LE, Garcia J, Aftab DT, Hessel C, Rachmilewitz Minei T, Harats D, Nathanson DA, Wen PY, Cloughesy TF. Depth of radiographic response and time to tumor regrowth predicts overall survival following anti-VEGF therapy in recurrent glioblastoma. Clin Cancer Res. 2023;29:4186–4195. doi: 10.1158/1078-0432.CCR-23-1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Jiguet-Jiglaire C, Boissonneau S, Denicolai E, Hein V, Lasseur R, Garcia J, Romain S, Appay R, Graillon T, Mason W, Carpentier AF, Brandes AA, Ouafik L’, Wick W, Baaziz A, Gigan JP, Argüello RJ, Figarella-Branger D, Chinot O, Tabouret E. Plasmatic MMP9 released from tumor-infiltrating neutrophils is predictive for bevacizumab efficacy in glioblastoma patients: an AVAglio ancillary study. Acta Neuropathol Commun. 2022;10:1. doi: 10.1186/s40478-021-01305-4. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES