CNS Oncology

Review For reprint orders, please contact: [email protected]

Antiangiogenic therapies for glioblastoma Isabel Arrillaga-Romany1 & Andrew D Norden*,2 Practice points ●● Glioblastoma (GBM) is the most common malignant primary brain tumor in adults and remains incurable.

●● Median overall survival is roughly 15 months with standard of care treatment, which includes 6 weeks of concurrent radiation and temozolomide followed by 6–12 months of adjuvant temozolomide.

●● Early Phase II trials of bevacizumab in recurrent GBM provided evidence of clinical benefit and served as the impetus for the accelerated US FDA approval of bevacizumab in 2009.

●● Two recent Phase III randomized trials of antiangiogenic therapy at initial diagnosis suggested improvement in progression-free survival, but failed to show an overall survival benefit.

●● Antiedema effects of antiangiogenic therapy with concomitant decrease in corticosteroid dependency were reported.

●● Resistance mechanisms are under active investigation and may help to improve overall efficacy of antiangiogenic agents.

Summary Glioblastoma is the most prevalent malignant primary brain tumor in adults and to date effective durable treatments are lacking. Preclinical studies underscore the importance of neovascularization for tumor survival, making angiogenesis an important treatment target. Early clinical experience in recurrent glioblastoma suggested that antiangiogenic agents may provide clinical benefit by prolonging progression-free survival, improving quality of life and decreasing peritumoral edema. Two recent Phase III randomized trials of antiangiogenic therapy at initial diagnosis suggested improvement in progressionfree survival, but failed to show an overall survival benefit. Ongoing preclinical research focuses on mechanisms of resistance and potential predictive biomarkers. Identification of targets to resistance pathways and of predictive biomarkers will hopefully improve efficacy of antiangiogenic therapies.

Keywords 

• antiangiogenesis • bevacizumab • glioblastoma • proangiogenic factors • resistance mechanisms • small-molecule inhibitor • vascular endothelial growth factor • VEGF

Background Glioblastoma (GBM) is the most common primary malignant brain tumor in adults and remains incurable. Median overall survival is roughly 15 months with standard of care treatment, which includes 6 weeks of concurrent radiation and temozolomide followed by 6–12 months of adjuvant temozolomide [1] . GBM is a highly vascularized tumor in which microvascular proliferation is typically observed. The precise mechanism by which angiogenesis impacts tumor growth and survival remains in question. VEGF has been identified as a prominent mediator of tumor angiogenesis [2–4] making interference with VEGF signaling a focus of significant scientific interest. Other antiangiogenic treatment strategies remain under active investigation. In this chapter Stephen E & Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, MA, USA Center for Neuro-Oncology, Dana-Farber/Brigham & Women’s Cancer Center, 450 Brookline Avenue, Boston, MA 02215, USA *Author for correspondence: Tel.: +1 617 632 2166; Fax: +1 617 632 4773; [email protected] 1 2

10.2217/CNS.14.31 © 2014 Future Medicine Ltd

CNS Oncol. (2014) 3(5), 349–358

part of

ISSN 2045-0907

349

Review  Arrillaga-Romany & Norden we will review basic mechanisms, evidence of efficacy and potential benefits of treatment with antiangiogenic therapy. Angiogenesis mechanisms The precise mechanism by which antiangiogenic therapy impacts GBM is uncertain. Classically, angiogenesis was thought to enhance tumorigenesis by increasing delivery of nutrients, oxygen and other growth factors necessary for tumor survival [5] . Blockade of new vessel formation would thereby limit access to these same elements. Early preclinical work indeed supported the role of vascular proliferation in tumor survival and propagation [6,7] . Alternative theories propose that antiangiogenesis works by promoting the ‘normalization’ of tumor vasculature. Tumor blood vessels are plentiful but aberrant in organization, architecture and physiology [8] , thus limiting delivery of chemotherapy agents and oxygen required for radiation to achieve antitumor effects. Tumor vessels are often hyperpermeable with enlarged diameters, thickened basement membranes and a reduced number of associated perivascular cells. These abnormalities elevate interstitial pressures and result in increased peritumoral edema. They diminish vascular perfusion leading to tumor hypoxia. By normalizing the vasculature, antiangiogenic therapy may improve perfusion, oxygenation, delivery of chemotherapy agents [8] and peritumoral edema [9] . This ability of antiangiogenic therapy to normalize tumor vasculature as measured using various imaging parameters has been linked to its efficacy [10–12] . Antiangiogenic targets The complex nature of the angiogenic process in cancer provides numerous targets for therapy. VEGF is the most thoroughly investigated proangiogenic factor in GBM and is likely the central mediator of neovascularization in many tumors [13,14] . Levels of VEGF receptor (VEGFR) ligand correlate with grade of glioma [15] , with mean concentrations of VEGF being 11-fold higher in extracts from grade III and IV gliomas than in extracts from lower grade counterparts. Preclinical work confirms that blockade of VEGF inhibits new tumor vessel formation and halts malignant glioma progression. Members of the VEGF family include VEGF-B, VEGF-C, VEGF-D and PIGF, but VEGF-A has the best established role in pathologic angiogenesis [16] .

350

CNS Oncol. (2014) 3(5)

Three VEGFR subtypes have been identified. VEGFR-1 and VEGFR-2 are essential for the normal development of the vasculature, and their inhibition during embryogenesis is lethal [17] . VEGF-A binds to VEGFR-2 on endothelial cells to promote endothelial cell growth and survival through paracrine signaling pathways [18] . The role of VEGFR-1 is less well defined, but does not appear to directly involve tumor angiogenesis. VEGFR-1 may, however, indirectly influence tumor survival by recruitment of bone marrow-derived cells that can differentiate into cells that promote the structure and integrity of new vasculature [19,20] . VEGFR-3 regulates lymphangiogenesis [21] . The identification of VEGF as an essential regulator of angiogenesis led to the development of VEGF and VEGFR inhibitors. Several antibodies and receptor tyrosine kinase inhibitors (TKI) have been developed and tested against GBM in both clinical and preclinical settings. Antibodies have the benefit of high affinity and selectivity, although their size precludes their ability to cross the blood–brain barrier (BBB). This has been proposed as a potential limitation of antibodies in the treatment of primary brain tumors, although tumor-associated BBB disruption may allow for adequate infiltration of these agents into the tumor. Small-molecule TKIs have the advantage of easy penetration across the BBB, but less selectivity. TKIs also have the advantage of oral bioavailability while antibodies are administered intravenously. Bevacizumab, the recombinant monoclonal anti-VEGF antibody, was first humanized in 1997 from a murine prototype [20] . It binds to all subtypes of VEGF with high affinity and selectivity. In animal models, bevacizumab inhibits angiogenesis and halts tumor growth [22] , including GBM proliferation [3,23] . Early clinical experience with bevacizumab in advanced colon, breast and lung cancers paved the way for it to become the most commonly used and thoroughly investigated antiangiogenic agent for the treatment of high-grade gliomas [24] . Several other proangiogenic molecules have been implicated in tumor angiogenesis and remain under active investigation. Among them are FGF, SDF-1α, angiopoietins, PDGF, IL-8, HIF-1α, neuropilins, endoglins and hepatocyte growth factor, among others [2,15,25–32] . Some, such as angiopoetins [33] and neuropilins [34] , interact directly with receptors on endothelial cells to promote new vessel growth and survival,

future science group

Antiangiogenic therapies for glioblastoma  while others, such as SDF-1α, work indirectly by attracting proangiogenic bone marrow-derived cells [35,36] . Integrins are cell adhesion receptors that help regulate the vascular extracellular matrix and can influence angiogenesis [37] , representing a separate target with therapeutic potential for GBM. Expressivity of the αvβ3 and αvβ5 integrin heterodimer subtypes found on the tumor’s growing edge directly correlates with glioma grade [38] , suggesting a central role in tumorigenesis. Integrins are found on glioma endothelial cells and during angiogenesis are required for endothelial cell migration and proliferation [17,39] . They may also play a role in preventing apoptosis [40] , thereby supporting endothelial cell survival [39] . Efficacy of antiangiogenic therapy ●●Bevacizumab

Early Phase II trials of bevacizumab in recurrent GBM provided evidence of clinical benefit and served as the impetus for the accelerated US FDA approval of bevacizumab in 2009. The BRAIN trial was a randomized Phase II noncomparative trial evaluating the efficacy of bevacizumab monotherapy and bevacizumab plus irinotecan in 167 patients with recurrent GBM [41] . The progression-free survival at 6 months (PFS6) was 42.6 and 50.3% for monotherapy and combination therapy, respectively, which represented a marked improvement over historical controls. For patients with a radiographic response or PFS >6 months, neurocognitive function at 24 weeks relative to baseline was improved or remained stable [42] . Importantly, bevacizumab proved to be well tolerated in these early clinical trials. In the BR AIN trial, 46% of patients treated with bevacizumab alone experienced grade 3 adverse events, the most common of which were hypertension and seizure. More serious adverse events included intracranial hemorrhage, seen in 2.4% of patients in the bevacizumab-only group and in 3.4% of patients in the combination therapy group [41] . Two additional trials provided further support for the utility of bevacizumab in the recurrent GBM setting. The first of these was performed at the National Cancer Institute and evaluated bevacizumab monotherapy in 47 patients with recurrent GBM. PFS6 and radiographic response rates were 29 and 35%, respectively [43] . The second trial – BELOB, a Phase II randomized

future science group

Review

study with assumed little crossover – evaluated the efficacy of bevacizumab, lomustine, and combination of the two agents. Combination treatment yielded an overall survival (OS) benefit at 9 months compared to treatment with either of the agents alone (59% for combination treatment, 42% for lomustine only, and 38% for bevacizumab only) [44] . Two larger, randomized, multicenter, Phase III trials, AVAglio and RTOG 0825, were conducted in newly diagnosed GBM and found a benefit in PFS but not OS. In both studies, patients were treated with standard therapy combined with bevacizumab or placebo. In the RTOG 0825 trial, PFS results favored bevacizumab (bevacizumab: 10.7 months vs placebo: 7.3 months; HR: 0.79; p = 0.007), but did not reach the prespecified thresholds of HR: 0.7 and p = 0.004 [45] . Importantly, criteria for enrollment into the RTOG 0825 trial specified a minimal amount of resected tissue rendering patients who received biopsies ineligible. Some would argue that this requirement excluded the subgroup of patients who might benefit the most from treatment with bevacizumab. In the AVAglio trial, the addition of bevacizumab to standard therapy during the initial 6-week period of chemoradiation produced a clinically meaningful and statistically significant improvement in PFS (6.2 months for the control arm vs 10.6 months for the bevacizumab arm; HR: 0.64; p 50% of those patients were able to decrease corticosteroids by an average of 59% [66] . More recent Phase III trials confirm antiedema effects of bevacizumab with 66% of patients initially on corticosteroids able to discontinue their use after treatment with bevacizumab, versus 44% in the control arm [63] . Several other trials reported corticosteroid reductions in 33–59% of patients with recurrent GBM following bevacizumab treatment [43,67–69] . Preclinical studies further highlight the potent effects of antiangiogenic therapy on peritumoral edema [9] . In three different orthotopic murine glioma models, cediranib significantly alleviated edema through normalization of tumor vasculature. Additionally, cediranib prolonged survival without concomitant effects on tumor [9] , suggesting that the survival benefits of cediranib may be attributable, at least in part, to its antiedema effect. Resistance mechanisms In the hopes of improving efficacy, significant efforts have been devoted to the study of mechanisms underlying resistance to treatment with bevacizumab and other antiangiogeneic agents. Resistance in this setting can be thought of as either intrinsic or acquired, although mechanisms underlying both may overlap [70] . Intrinsic resistance refers to the inherent ability of glioma tumor cells to elude antiangiogenic therapy. Clinical and preclinical evidence suggests that a significant minority of high-grade gliomas might be refractory to VEGF-targeted therapy from treatment onset. For example, treatment with cediranib in a xenograft model of GBM did not affect tumor growth [9] and some patients respond minimally or not at all to treatment with VEGF inhibitors [11,71] . Inherent resistance may stem from redundant proangiogenic signaling pathways or from the existence of a pool of glioma stem cells that reside in a quiescent state unresponsive to antiangiogenic therapy [72,73] . After initial failed treatment with bevacizumab, use of a second-line bevacizumab containing regimen is typically ineffective. In this setting, PFS and response rates are extremely poor, supporting the notion of acquired resistance to antiangiogenic therapy [74] . Modes of acquired resistance under investigation include [72] :

future science group

Antiangiogenic therapies for glioblastoma 

Review

Table 1. Recent selected trials of antiangiogenic therapy for glioblastoma. Study, year (trial)

n  

Design  

Regimens  

Friedman et al., 2009 (BRAIN)

167

Phase II randomized in rGBM

Kreisl et al., 2009 (NCI) Field et al., (CABARET)

67

Phase II prospective in rGBM

122 

Phase II randomized in rGBM

Taal et al., 2013 (BELOB)

148

Phase II randomized in rGBM

Wick et al., 2013 (AvaGlio)

921

Phase III randomized in nGBM

Gilbert et al., 2013 (RTOG 0825)

637

Phase III randomized in nGBM

Batchelor et al., 2013 (REGAL)

325

Phase III randomized in rGBM

Stupp et al., 2013 (CENTRIC)

545

Phase III randomized in rGBM with methylated MGMT

PFS

 

OS

Median 6 months (months)  (%)

  Median (months) 

9 months (%)

Bev alone Bev + IRI Bev alone

 NA  NA 4

42.6 50.3 29

10   9.5   7.8

NA NA NA

Bev alone Bev + carboplatin Bev alone Lomustine alone Bev + lomustine RT/TMZ + TMZ/Plc XRT/TMZ + TMZ/Bev RT/TMZ + TMZ/Plc XRT/TMZ + TMZ/Bev Cediranib alone Lomustine alone Cediranib + lomustine RT/TMX + TMZ/CIL TMZ/RT + TMZ/Plc

NA NA 3 2 4 6.2 10 7.3 10.7 3.1 4 2.5 13.5 10

24 26 18 11 41 NA NA NA NA NA NA NA NA NA

                           

NA NA 38 43 59 NA NA NA NA NA NA NA NA NA

6.4 6  NA  NA  NA 16.8 16.9 16.1 15.7 8 9.4 9.8 26.3 26.3

Ref.

[41]  [43] [62]    [44]

[63] [64] [65]

[60]   

Bev: Bevacizumab; CIL: Cilengitide; IRI: Irinotecan; MGMT: O6-methylguanine-DNA methyltransferase; NA: Not applicable; nGBM: Newly diagnosed glioblastoma; OS: Overall survival; PFS: Progression-free survival; Plc: Placebo; rGBM: Recurrent glioblastoma; RT: Radiation therapy; TMZ: Temozolamide.

●● Adaptive upregulation of proangiogenic

signaling pathways; ●● Recruitment of bone marrow derived

proangiogenic cells to the tumor site; ●● Transformation to a more invasive glioma

phenotype; ●● Promotion of autophagy.

Growth factors with increased expression during periods of resistance to anti-VEGF therapy include FGF-1, FGF-2, Ephrin-A1 (Eph-A1), Eph-A2, angiopoietin-1, VEGF, PlGF, granulocyte colony-stimulating factor, SDF-1α, stem-cell factor, signal transducer and activator of transcription 3 (STAT3) and osteopontin [11,75–78] . Release of these angiogenic factors can be induced by hypoxia, which itself may be provoked by excessive vessel pruning in the setting of angiogenesis inhibition. The upregulation of proangiogenic factors in response to anti-VEGF therapy may have additional clinical implications in that discontinuation of the treatment could result in accelerated tumor growth [70] . Bone marrow-derived proangiogenic cells have also been implicated in acquired resistance to antiangiogenic therapy [28,72,79] . These include tumor-activated macrophages, granulocytes and

future science group

precursor myeloid cells, all of which are recruited to the tumor site in large quantities during resistance and tumor progression [9,80,81] . These cells stimulate the expression of cytokines, growth factors and proteases which may help to promote tumor growth. Precursor myeloid cells, including endothelial and pericyte progenitors, support angiogenesis and neovascularization by differentiating into cells that are essential to the structure and integrity of new vasculature [82,83] . Recruitment of myeloid precursor cells may be associated with the activation of p-STAT3, an important signaling molecule. AZD1480 is an inhibitor of the JAK/STAT pathway that has been used in combination with anti-VEGF TKI treatment in xenograft models of GBM. The combination reduced the influx of proangiogenic macrophages to the tumor site and improved survival [78,84] . Antiangiogenic therapy may also promote resistance by encouraging transformation to a more invasive and mesenchymal glioma phenotype [85,86] . This transformation is likely stimulated by an increase in tumor hypoxia through the upregulation of c-Met expression [87] . In preclinical models, cabozantinib (XL184), a co-inhibitor of c-MET and VEGF-R, amplifies the antiangiogenic phenomena and halts tumor growth

www.futuremedicine.com

353

Review  Arrillaga-Romany & Norden

A

B

C

D

354

CNS Oncol. (2014) 3(5)

future science group

Antiangiogenic therapies for glioblastoma 

Review

Figure 1. Effects of bevacizumab on tumor-associated vasogenic edema and contrast-enhancing tumor progression in glioblastoma multiforme (facing page). T2/FLAIR imaging obtained (A) immediately prior to and (B) 4 months after initiation of bevacizumab. Post-contrast T1 imaging obtained (C) immediately prior to and (D) 4 months after initiation of bevacizumab. 

over blockade of VEGF-R alone [88] . Preliminary Phase II trial results of this agent revealed some efficacy in recurrent GBM [89] , but final results are pending. Onartuzamab, an antibody that inhibits MET, is currently in a Phase II clinical trial as combination therapy with bevacizumab for GBM. Predictive biomarkers Investigations into antiangiogenic therapy resistance mechanisms have gone hand-in-hand with efforts to identify biomarkers of response. Notable candidates have included imaging and serum markers of vascular normalization, which together can be viewed as the ‘vascular normalization index’. In a prospective Phase II trial of recurrent GBM, one dose of cediranib decreased tumor permeability on MRI in the subset of patients who achieved response to therapy and increased overall survival. No survival benefit was observed in patients whose initial permeability imaging was unaffected by treatment [11] . Likewise, in a subset of newly diagnosed GBM patients, the addition of cediranib to standard of care treatment with radiation and temozolomide increased vascular perfusion on MRI. This change in perfusion was evident early in treatment and was associated with an improvement in overall survival, suggesting that early imaging-based response may serve as a predictive biomarker for antiangiogenic therapy [10] . Circulating biomarkers of response may also exist. For example, the level of serum collagen IV (thought to reflect vessel basement membrane thinning) following one dose of cediranib has been associated with a survival benefit [12] . Although important and suggestive, biomarker data remain to be validated in randomized clinical trials. Conclusion & future perspective GBM remains a lethal neoplasm with no effective long-term treatment. The discovery of References 1

2

Stupp R, Mason WP, Van Den Bent MJ et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 352(10), 987–996 (2005). Plate KH, Breier G, Weich HA, Risau W. Vascular endothelial growth factor is a

future science group

angiogenesis and VEGF as a potential driver of tumor growth and survival has led to extensive research in this field. Bevacizumab is a monoclonal antibody to VEGF that has become the prototype for antiangiogenic treatment of GBM. Mechanisms underlying antiangiogenesis are becoming better understood, leading to the development of a diverse group of antiangiogenic agents. Initial clinical investigations with bevacizumab were promising and led to the accelerated approval of this agent for recurrent GBM, conditional on further post-market assessment of efficacy. Results from two Phase III randomized trials revealed efficacy in PFS, but not OS, in the upfront setting. Antiedema effects of antiangiogenic therapy with concomitant decrease in corticosteroid dependency were reported. Additional trials conducted with a diverse variety of antiangiogenic agents have yet to support a beneficial effect on overall survival. Resistance to treatment, either acquired or inherent, may be at play. Ongoing work on predictive biomarkers and potential resistance mechanisms may help improve the outcomes of antiangiogenic therapy for GBM. Additionally, novel-angiogenic targets and combinatorial approaches, including the combination with other targeted therapies and immunotherapies, should be explored. Financial & competing interests disclosure The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. No writing assistance was utilized in the production of this manuscript.

potential tumour angiogenesis factor in human gliomas in vivo. Nature 359(6398), 845–848 (1992). 3

Kim KJ, Li B, Winer J et al. Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 362(6423), 841–844 (1993).

4

Jensen RL, Ragel BT, Whang K, Gillespie D. Inhibition of hypoxia inducible factor-1alpha (HIF-1alpha) decreases vascular endothelial growth factor (VEGF) secretion and tumor growth in malignant gliomas. J. Neurooncol. 78(3), 233–247 (2006).

www.futuremedicine.com

355

Review  Arrillaga-Romany & Norden 5

Folkman J. Angiogenesis: an organizing principle for drug discovery? Nat. Rev. Drug Discov. 6(4), 273–286 (2007).

6

Gullino PM. Angiogenesis and oncogenesis. J. Natl Cancer Inst. 61(3), 639–643 (1978).

7

Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat. Rev. Drug Discov. 3(5), 391–400 (2004).

8

Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307(5706), 58–62 (2005).

9

Kamoun WS, Ley CD, Farrar CT et al. Edema control by cediranib, a vascular endothelial growth factor receptor-targeted kinase inhibitor, prolongs survival despite persistent brain tumor growth in mice. J. Clin. Oncol. 27(15), 2542–2552 (2009).

10 Batchelor TT, Gerstner ER, Emblem KE et al.

Improved tumor oxygenation and survival in glioblastoma patients who show increased blood perfusion after cediranib and chemoradiation. Proc. Natl Acad. Sci. USA 110(47), 19059–19064 (2013). 11 Batchelor TT, Sorensen AG, Di Tomaso E

et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell 11(1), 83–95 (2007). 12 Sorensen AG, Batchelor TT, Zhang WT et al.

A ‘vascular normalization index’ as potential mechanistic biomarker to predict survival after a single dose of cediranib in recurrent glioblastoma patients. Cancer Res. 69(13), 5296–5300 (2009). 13 Kuo CJ, Farnebo F, Yu EY et al. Comparative

evaluation of the antitumor activity of antiangiogenic proteins delivered by gene transfer. Proc. Natl Acad. Sci. USA 98(8), 4605–4610 (2001). 14 Ferrara N, Gerber HP, Lecouter J. The

biology of VEGF and its receptors. Nat. Med. 9(6), 669–676 (2003). 15 Schmidt NO, Westphal M, Hagel C et al.

Levels of vascular endothelial growth factor, hepatocyte growth factor/scatter factor and basic fibroblast growth factor in human gliomas and their relation to angiogenesis. Int. J. Cancer 84(1), 10–18 (1999). 16 Norden AD, Drappatz J, Wen PY. Novel

anti-angiogenic therapies for malignant gliomas. Lancet Neurol. 7(12), 1152–1160 (2008). 17 Shalaby F, Rossant J, Yamaguchi TP et al.

Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 376(6535), 62–66 (1995).

356

18 Millauer B, Shawver LK, Plate KH, Risau W,

Ullrich A. Glioblastoma growth inhibited in vivo by a dominant-negative Flk-1 mutant. Nature 367(6463), 576–579 (1994). 19 Barleon B, Sozzani S, Zhou D, Weich HA,

Mantovani A, Marme D. Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood 87(8), 3336–3343 (1996). 20 Autiero M, Luttun A, Tjwa M, Carmeliet P.

Placental growth factor and its receptor, vascular endothelial growth factor receptor-1: novel targets for stimulation of ischemic tissue revascularization and inhibition of angiogenic and inflammatory disorders. J. Thromb. Haemost. 1(7), 1356–1370 (2003). 21 Coso S, Zeng Y, Opeskin K, Williams ED.

Vascular endothelial growth factor receptor-3 directly interacts with phosphatidylinositol 3-kinase to regulate lymphangiogenesis. PLoS ONE 7(6), e39558 (2012). 22 Presta LG, Chen H, O’Connor SJ et al.

Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Res. 57(20), 4593–4599 (1997). 23 Rubenstein JL, Kim J, Ozawa T et al.

Anti-VEGF antibody treatment of glioblastoma prolongs survival but results in increased vascular cooption. Neoplasia 2(4), 306–314 (2000). 24 Norden AD, Drappatz J, Wen PY.

Antiangiogenic therapies for high-grade glioma. Nat. Rev. Neurol. 5(11), 610–620 (2009). 25 Stefanik DF, Rizkalla LR, Soi A, Goldblatt

SA, Rizkalla WM. Acidic and basic fibroblast growth factors are present in glioblastoma multiforme. Cancer Res. 51(20), 5760–5765 (1991). 26 Reiss Y, Machein MR, Plate KH. The role of

angiopoietins during angiogenesis in gliomas. Brain Pathol. 15(4), 311–317 (2005). 27 Shih AH, Holland EC. Platelet-derived

growth factor (PDGF) and glial tumorigenesis. Cancer Lett. 232(2), 139–147 (2006). 28 Du R, Lu KV, Petritsch C et al. HIF1alpha

induces the recruitment of bone marrowderived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell 13(3), 206–220 (2008). 29 Folkins C, Shaked Y, Man S et al. Glioma

tumor stem-like cells promote tumor angiogenesis and vasculogenesis via vascular endothelial growth factor and stromal-derived factor 1. Cancer Res. 69(18), 7243–7251 (2009).

CNS Oncol. (2014) 3(5)

30 Carmeliet P, Jain RK. Molecular

mechanisms and clinical applications of angiogenesis. Nature 473(7347), 298–307 (2011). 31 Wang LF, Fokas E, Juricko J et al. Increased

expression of EphA7 correlates with adverse outcome in primary and recurrent glioblastoma multiforme patients. BMC Cancer 8, 79 (2008). 32 Brat DJ, Bellail AC, Van Meir EG. The role

of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro. Oncol. 7(2), 122–133 (2005). 33 Holash J, Maisonpierre PC, Compton D

et al. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science 284(5422), 1994–1998 (1999). 34 Soker S, Fidder H, Neufeld G, Klagsbrun M.

Characterization of novel vascular endothelial growth factor (VEGF) receptors on tumor cells that bind VEGF165 via its exon 7-encoded domain. J. Biol. Chem. 271(10), 5761–5767 (1996). 35 Kioi M, Vogel H, Schultz G, Hoffman RM,

Harsh GR, Brown JM. Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J. Clin. Invest. 120(3), 694–705 (2010). 36 Cheng L, Huang Z, Zhou W et al.

Glioblastoma stem cells generate vascular pericytes to support vessel function and tumor growth. Cell 153(1), 139–152 (2013). 37 Stupp R, Ruegg C. Integrin inhibitors

reaching the clinic. J. Clin. Oncol. 25(13), 1637–1638 (2007). 38 Bello L, Francolini M, Marthyn P et al.

Alpha(v)beta3 and alpha(v)beta5 integrin expression in glioma periphery. Neurosurgery 49(2), 380–389; discussion 390 (2001). 39 Alghisi GC, Ruegg C. Vascular integrins in

tumor angiogenesis: mediators and therapeutic targets. Endothelium 13(2), 113–135 (2006). 40 Byzova TV, Rabbani R, D’Souza SE, Plow

EF. Role of integrin alpha(v)beta3 in vascular biology. Thromb. Haemost. 80(5), 726–734 (1998). 41 Friedman HS, Prados MD, Wen PY et al.

Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J. Clin. Oncol. 27(28), 4733–4740 (2009). 42 Wefel JS, Cloughesy T, Zazzali JL et al.

Neurocognitive function in patients with recurrent glioblastoma treated with bevacizumab. Neuro. Oncol. 13(6), 660–668 (2011).

future science group

Antiangiogenic therapies for glioblastoma  43 Kreisl TN, Kim L, Moore K et al. Phase II trial

of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J. Clin. Oncol. 27(5), 740–745 (2009). 44 Taal W OH, Walenkamp AM, Beerepoot LV

et al. A randomized Phase II study of bevacizumab versus bevacizumab plus lomustine versus lomustine single agent in recurrent glioblastoma: The Dutch BELOB study. J. Clin. Oncol. 31(Suppl.), Abstract 2001 (2013). 45 Gilbert MR, Dignam JJ, Armstrong TS et al. A

randomized trial of bevacizumab for newly diagnosed glioblastoma. N. Engl. J. Med. 370(8), 699–708 (2014). 46 Chinot OL, Wick W, Mason W et al.

Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N. Engl. J. Med. 370(8), 709–722 (2014). 47 Hainsworth JD, Ervin T, Friedman E et al.

Concurrent radiotherapy and temozolomide followed by temozolomide and sorafenib in the first-line treatment of patients with glioblastoma multiforme. Cancer 116(15), 3663–3669 (2010). 48 Iwamoto FM, Lamborn KR, Robins HI et al.

Phase II trial of pazopanib (GW786034), an oral multi-targeted angiogenesis inhibitor, for adults with recurrent glioblastoma (North American Brain Tumor Consortium Study 06–02). Neuro. Oncol. 12(8), 855–861 (2010). 49 Reardon DA, Vredenburgh JJ, Coan A et al.

Phase I study of sunitinib and irinotecan for patients with recurrent malignant glioma. J. Neurooncol. 105(3), 621–627 (2011). 50 Wick W, Puduvalli VK, Chamberlain MC

et al. Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J. Clin. Oncol. 28(7), 1168–1174 (2010). 51 Holash J, Davis S, Papadopoulos N et al.

VEGF-Trap: a VEGF blocker with potent antitumor effects. Proc. Natl Acad. Sci. USA 99(17), 11393–11398 (2002). 52 De Groot JF, Lamborn KR, Chang SM et al.

Phase II study of aflibercept in recurrent malignant glioma: a North American Brain Tumor Consortium study. J. Clin. Oncol. 29(19), 2689–2695 (2011). 53 Mamluk R, Carvajal IM, Morse BA et al.

Anti-tumor effect of CT-322 as an adnectin inhibitor of vascular endothelial growth factor receptor-2. mAbs 2(2), 199–208 (2010). 54 Adnexus AB-MSRDC: CT-322 in treating

patients with recurrentglioblastoma multiforme and combination therapy with irinotecan. www.clinicaltrials.gov/show/NCT00562419

future science group

55 Chamberlain MC, Cloughsey T, Reardon DA,

Wen PY. A novel treatment for glioblastoma: integrin inhibition. Expert. Rev. Neurother. 12(4), 421–435 (2012). 56 Xiong JP, Stehle T, Zhang R et al. Crystal

structure of the extracellular segment of integrin alpha Vbeta3 in complex with an Arg-Gly-Asp ligand. Science 296(5565), 151–155 (2002). 57 Reardon DA, Fink KL, Mikkelsen T et al.

Randomized Phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme. J. Clin. Oncol. 26(34), 5610–5617 (2008). 58 Gerstner Er LM, Ye X, Mikkelsen T et al.

A Phase I study of cediranib in combination with cilengitide in patients with recurrent glioblastoma. J. Clin. Oncol. 31(Suppl.), Abstract 2054 (2013). 59 Stupp R, Hegi ME, Neyns B et al. Phase I/IIa

study of cilengitide and temozolomide with concomitant radiotherapy followed by cilengitide and temozolomide maintenance therapy in patients with newly diagnosed glioblastoma. J. Clin. Oncol. 28(16), 2712–2718 (2010). 60 Stupp R HM, Gorlia T, Erridge S et al.

Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma and methylated O6methylguanine-DNA methyltransferase (MGMT) gene promoter: key results of the multicenter, randomized, open-label, controlled, Phase III CENTRIC study. J. Clin. Oncol. 31(Suppl.), Abstract LBA2009 (2013). 61 Serono E. Cilengitide, temozolomide, and

radiationtherapy in treating patients with newly diagnosed glioblastoma and unmethylated gene promoter status CORE. www.clinicaltrials.gov/ct2/show/ NCT00813943 62 Field KM, Rosenthal M, Wheeler H. A

randomized Phase II study of carboplatin and bevacizumab in recurrent glioblastoma multiforme (CABARET). J. Clin. Oncol. 31(Suppl.), Abstract 2017 (2013). 63 Wick W, Cloughesy TF, Nishikawa R et al.

Tumor response based on adapted Macdonald criteria and assessment of pseudoprogression (PsPD) in the Phase III AVAglio trial of bevacizumab (Bv) plus temozolomide (T) plus radiotherapy (RT) in newly diagnosed glioblastoma (GBM). J. Clin. Oncol. 31(Suppl.), Abstract 2002 (2013). 64 Gilbert MR, Dignam J, Won M et al.

RTOG 0825: Phase III double-blind placebo-controlled trial evaluating

Review

bevacizumab (Bev) in patients (Pts) with newly diagnosed glioblastoma (GBM). J. Clin. Oncol. 31(Suppl.), Abstract 1 (2013). 65 Batchelor TT, Mulholland P, Neyns B et al.

Phase III randomized trial comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, versus lomustine alone in patients with recurrent glioblastoma. J. Clin. Oncol. 31(26), 3212–3218 (2013). 66 Vredenburgh JJ, Cloughesy T, Samant M

et al. Corticosteroid use in patients with glioblastoma at first or second relapse treated with bevacizumab in the BRAIN study. Oncologist 15(12), 1329–1334 (2010). 67 Vredenburgh JJ, Desjardins A, Herndon JE,

2nd et al. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin. Cancer Res. 13(4), 1253–1259 (2007). 68 Norden AD, Young GS, Setayesh K et al.

Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology 70(10), 779–787 (2008). 69 Chamberlain MC, Johnston SK. Salvage

therapy with single agent bevacizumab for recurrent glioblastoma. J. Neurooncol. 96(2), 259–269 (2010). 70 Loges S, Schmidt T, Carmeliet P.

Mechanisms of resistance to anti-angiogenic therapy and development of third-generation anti-angiogenic drug candidates. Genes Cancer 1(1), 12–25 (2010). 71 Kerbel RS. Tumor angiogenesis. N. Engl.

J. Med. 358(19), 2039–2049 (2008). 72 Bergers G, Hanahan D. Modes of resistance

to anti-angiogenic therapy. Nat. Rev. Cancer 8(8), 592–603 (2008). 73 Rahman R, Smith S, Rahman C, Grundy R.

Antiangiogenic therapy and mechanisms of tumor resistance in malignant glioma. J. Oncol. 2010, 251231 (2010). 74 Quant EC, Norden AD, Drappatz J et al.

Role of a second chemotherapy in recurrent malignant glioma patients who progress on bevacizumab. Neuro. Oncol. 11(5), 550–555 (2009). 75 Ebos JM, Lee CR, Christensen JG, Mutsaers

AJ, Kerbel RS. Multiple circulating proangiogenic factors induced by sunitinib malate are tumor-independent and correlate with antitumor efficacy. Proc. Natl Acad. Sci. USA 104(43), 17069–17074 (2007). 76 Casanovas O, Hicklin DJ, Bergers G,

Hanahan D. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 8(4), 299–309 (2005).

www.futuremedicine.com

357

Review  Arrillaga-Romany & Norden 77 Fernando NT, Koch M, Rothrock C et al.

Tumor escape from endogenous, extracellular matrix-associated angiogenesis inhibitors by up-regulation of multiple proangiogenic factors. Clin. Cancer Res. 14(5), 1529–1539 (2008). 78 de Groot J, Liang J, Kong LY et al.

Modulating antiangiogenic resistance by inhibiting the signal transducer and activator of transcription 3 pathway in glioblastoma. Oncotarget 3(9), 1036–1048 (2012). 79 Lu-Emerson C, Snuderl M, Kirkpatrick ND

et al. Increase in tumor-associated macrophages after antiangiogenic therapy is associated with poor survival among patients with recurrent glioblastoma. Neuro. Oncol. 15(8), 1079–1087 (2013). 80 Piao Y, Liang J, Holmes L et al.

Glioblastoma resistance to anti-VEGF therapy is associated with myeloid cell infiltration, stem cell accumulation, and a mesenchymal phenotype. Neuro. Oncol. 14(11), 1379–1392 (2012).

358

81 Shojaei F, Wu X, Malik AK et al. Tumor

refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat. Biotechnol. 25(8), 911–920 (2007). 82 Asahara T, Murohara T, Sullivan A et al.

Isolation of putative progenitor endothelial cells for angiogenesis. Science 275(5302), 964–967 (1997). 83 Song S, Ewald AJ, Stallcup W, Werb Z,

Bergers G. PDGFRbeta+ perivascular progenitor cells in tumours regulate pericyte differentiation and vascular survival. Nat. Cell Biol. 7(9), 870–879 (2005). 84 Mcfarland BC, Ma JY, Langford CP et al.

Therapeutic potential of AZD1480 for the treatment of human glioblastoma. Mol. Cancer Therap. 10(12), 2384–2393 (2011). 85 Lu KV, Chang JP, Parachoniak CA et al.

VEGF inhibits tumor cell invasion and mesenchymal transition through a MET/VEGFR2 complex. Cancer Cell 22(1), 21–35 (2012).

CNS Oncol. (2014) 3(5)

86 Piao Y, Liang J, Holmes L, Henry V,

Sulman E, de Groot JF. Acquired resistance to anti-VEGF therapy in glioblastoma is associated with a mesenchymal transition. Clin. Cancer Res. 19(16), 4392–4403 (2013). 87 Jahangiri A, De Lay M, Miller LM et al.

Gene expression profile identifies tyrosine kinase c-Met as a targetable mediator of antiangiogenic therapy resistance. Clin. Cancer Res. 19(7), 1773–1783 (2013). 88 You WK, Sennino B, Williamson CW et al.

VEGF and c-Met blockade amplify angiogenesis inhibition in pancreatic islet cancer. Cancer Res. 71(14), 4758–4768 (2011). 89 Wen Py PM, Schiff D et al. Phase II study of

XL184 (BMS 907351), an inhibitor of MET, VEGFR2, and RET, in patients with progressive glioblastoma. J. Clin. Oncol. 28(15), 181S (2010).

future science group

Antiangiogenic therapies for glioblastoma.

Glioblastoma is the most prevalent malignant primary brain tumor in adults and to date effective durable treatments are lacking. Preclinical studies u...
3MB Sizes 2 Downloads 6 Views