Abstract
Angiogenesis has long been considered an important target for cancer therapy. Initial efforts have primarily focused on targeting of endothelial and tumor-derived vascular endothelial growth factor signaling. As evidence emerges that angiogenesis has significant mechanistic complexity, therapeutic resistance and escape have become practical limitations to drug development. Here, we review the mechanisms by which dynamic changes occur in the tumor microenvironment in response to antiangiogenic therapy, leading to drug resistance. These mechanisms include direct selection of clonal cell populations with the capacity to rapidly upregulate alternative proangiogenic pathways, increased invasive capacity, and intrinsic resistance to hypoxia. The implications of normalization of vasculature with subsequently improved vascular function as a result of antiangiogenic therapy are explored, as are the implications of the ability to incorporate and co-opt otherwise normal vasculature. Finally, we consider the extent to which a better understanding of the biology of hypoxia and reoxygenation, as well as the depth and breadth of systems invested in angiogenesis, may offer putative biomarkers and novel therapeutic targets. Insights gained through this work may offer solutions for personalizing antiangiogenesis approaches and improving the outcome of patients with cancer.
INTRODUCTION
Angiogenesis plays a critical role in tumor growth and progression.1 Tumors acquire their supply by a variety of means including vasculogenesis, co-option of previously established vasculature, and vascular mimicry.2,3 Efforts to target tumor angiogenesis have focused on the vascular endothelial growth factor (VEGF) pathway.4 VEGF targeting has shown promise for some cancers but has not proven as efficacious as hoped.5,6 Evidence suggests mechanisms of escape mediated by tumor cells and by members of the microenvironment, leading to the hypothesis that simultaneous targeting of complementary and redundant pathways may hold promise in the treatment of solid tumors.7,8
TARGETING VEGF
More than 30 years ago, on the basis of the recognition that tumor-associated endothelial cells play a fundamental role in tumor neovascularization, and given their presumed genetic stability, these cells were proposed as a therapeutic target. VEGF-A was identified as a central endothelial cell survival factor and angiogenesis promoter.9 Bevacizumab (monoclonal antibody against VEGF-A165) was among the initial antiangiogenic agents developed.10 This therapeutic strategy has provided clinical benefit in several solid tumor types, and bevacizumab remains approved for colorectal, renal, nonsquamous/non–small-cell lung cancer, and glioblastoma.11 Although bevacizumab was approved for metastatic breast cancer in 2008, approval was withdrawn secondary to concerns about efficacy relative to toxicity.11 In ovarian cancer, GOG 218 (Gynecologic Oncology Group 218; three-arm trial: paclitaxel/carboplatin chemotherapy (CT) v CT plus concurrent bevacizumab v CT plus concurrent and maintenance bevacizumab) and GCIG ICON7 (Gynaecologic Cancer InterGroup International Collaboration on Ovarian Neoplasms 7; two-arm trial: CT ± concurrent and maintenance bevacizumab) were both conducted in the first-line adjuvant setting after tumor cytoreduction.12,13 In both trials, modest improvements in progression-free survival (PFS) were noted in the groups receiving maintenance bevacizumab. Overall survival (OS) data are not mature but are not expected to be positive. Although interval to progression has improved, there seems to have been no improvement in the total number of patients who progressed. Small-molecule inhibitor data are less mature, but similar observations have been made regarding sorafenib and sunitinib, prompting investigation into potential mechanisms of escape from anti-VEGF therapy.14,15
ESCAPE/RESISTANCE MECHANISMS
Evolutionary biology teaches that disruptions in an ecosystem by an imposed selection pressure produce reactionary dynamics and interactions, and natural selection will result in a resilient system. Cancer cells are characteristically heterogeneous and genetically unstable.16 Furthermore, normal cells in the tumor microenvironment such as endothelial cells, pericytes, platelets, fibroblasts, and leukocytes are known to have normal functions that are co-opted to support tumor growth and progression. Potential mechanisms of resistance to antiangiogenic therapy, therefore, may result from the selection of directly and indirectly advantaged subpopulations of tumor and tumor-associated cells. If anti-VEGF therapy is considered as a selection pressure, and surviving cell populations are considered to be advantaged in the new environment, multiple plausible mechanisms of resistance and escape emerge for consideration (Fig 1).
Fig 1.
Resistance and escape from antiangiogenesis therapy is multifactorial; it is driven by the intrinsic properties of cancer cell subpopulations and members of the tumor microenvironment, resulting in the evolution of an advantaged tumor ecosystem in response to the stimulus of antiangiogenic therapy. AldoA, Aldolase-A; APO, apolipoprotein; DLL, delta-like ligand; ENA, epithelial neutrophil-activating peptide; FGF, fibroblast growth factor; GCSF, granulocyte colony-stimulating factor; Glut-1, glucose transporter 1; GRO, growth-regulated oncogene; HGF, hepatocyte growth factor; HIF1-α, hypoxia-inducible factor-1 alpha; HRG, histidine-rich glycoprotein; IGF, insulin-like growth factor; IGFBP, IGF binding protein; IL, interleukin; MCP, monocyte chemotactic protein; MIP, macrophage inflammatory protein; MMP, matrix metalloproteinase; mTOR, mammalian target of rapamycin; PAI, plasminogen activator inhibitor; PDGF, platelet-derived growth factor; PI3K, phosphoinositide 3–kinase; PlGF, placental growth factor; RANTES, regulated and normal T cell expressed and secreted; SDF, stromal cell–derived factor; U-PA, urokinase-type plasminogen activator; VEGF, vascular endothelial growth factor.
Direct Selection Benefit to Tumor Cells
Selection of clonal populations with upregulated alternative and compensatory pathways.
Various data raise the question of whether anti-VEGF therapy selects for clonal populations that have upregulated alternative and compensatory proangiogenic signaling cascades. In pancreatic cancer models, treatment with a VEGF receptor 2 (VEGFR-2) –inhibiting antibody resulted in 10 to 14 days of tumor stasis followed by tumor regrowth, with apparent acceleration of tumor vascularity and increased expression of fibroblast growth factor 1 (FGF-1), FGF-2, Ephrin-A1 (Eph-A1), Eph-A2, and angiopoietin-1.7 In tumors whose angiogenesis should otherwise be suppressed by ectopic expression of angiogenesis inhibitors, compensatory upregulation of VEGF, platelet-derived growth factor (PDGF), and FGF-2 is observed.17 In human studies, predictable upregulation of FGF-2, VEGF, and placental growth factor (PlGF) has been detected in response to antiangiogenic therapy.18 In sunitinib-treated mice, VEGF, granulocyte colony-stimulating factor, stromal cell–derived factor 1-alpha (SDF-1α), stem-cell factor, and osteopontin have all been found to be increased.18
FGFs are important promoters of angiogenic and mitogenic activity.19 FGF-1 and FGF-2 induce angiogenesis, and preclinical models suggest that FGF binding protein releases FGF-2 from the extracellular matrix, allowing it access to receptors.20,21 Cross talk between VEGF and FGF may stimulate angiogenesis synergistically, with variable effects on vessel size and function.19 In a phase II trial of FOLFIRI+B (folinic acid, fluorouracil, irinotecan, and bevacizumab) in colorectal cancer, significant increases in FGF-2 as well as hepatocyte growth factor, PlGF, SDF-1, and macrophage chemoattractant protein-3 were observed in plasma samples.22 In patients with glioblastoma, treatment with cediranib, a pan-VEGFR inhibitor, resulted in decreased tumor edema by dynamic magnetic resonance imaging; subsequent increases in FGF-2 and SDF-1 levels correlated with progression.23 Murine models of pancreatic and renal-cell carcinoma further support that upregulation of FGF-2 is associated with tumor progression in the face of VEGFR-2 blockade, and upregulation of FGF-2, angiopoietin-2, and PDGF-A has been implicated in bypass of antiangiogenic signaling.17
Recognizing the complexity of angiogenesis, other signaling systems are potentially implicated. In tumor models without hypoxia inducible factor 1-alpha (HIF-1α), interleukin-8 (IL-8) expression has been shown to support angiogenesis.24 In a phase II trial of FOLFIRI+B in colorectal cancer, increased baseline IL-8 correlated with decreased PFS.22 Increased insulin-like growth factor 1 has been associated with increased prostaglandin E2 expression, which correlates with increased VEGF expression and enhanced angiogenesis.25 VEGF precursor mRNA undergoes alternative splicing that generates both pro- and antiangiogenic forms, and variant splicing has been hypothesized to explain both initial and acquired resistance to antiangiogenic therapy.26 Studies of the murine double minute (MDM2) oncogene have demonstrated a p53-independent, hypoxia-driven translocation to the cytoplasm, with binding and stabilization of VEGF mRNA, increasing translation, VEGF protein production, cell survival, and angiogenesis.27 Epidermal growth factor receptor (EGFR) –mediated activation of the phosphoinositide 3–kinase/Akt/mammalian target of rapamycin pathway can also upregulate HIF-1α and VEGF, and there have been separate pathways described by which VEGF production via phosphoinositide 3–kinase is independent of HIF.28 One report suggested that blockade with anti-EGFR antibody resulted in selection of tumor cell subpopulations with increased angiogenic potential.29 These studies form the basis of dual targeting of EGFR and VEGFR with drugs such as vandetanib.30
Selection of clones with greater invasive capacity in nonangiogenic environment.
Evidence suggests that when VEGFR/PDGF receptor (PDGFR) signaling is blocked with sunitinib (preclinical pancreatic and glioblastoma models) in addition to anticipated antitumor effects, a tumor phenotype is observed with heightened invasive and metastatic potential. In an orthotopic mouse model of glioblastoma multiforme, it was observed that blockade of VEGF with a neutralizing antibody resulted in a tumor phenotype that continued to grow, albeit more slowly, with persistent invasive capacity.31 These models remain unsubstantiated and controversial, but they raise potential concerns regarding unintended consequences of blockade of these pathways, and mechanisms remain under exploration.32,33
Aside from the upregulated mitogenic pathways discussed previously, the hypoxic, acidic, high–interstitial fluid pressure features of the tumor microenvironment seem to favor tumor clones with more aggressive behavior.34 It has been argued that this microenvironment fuels nonproductive angiogenesis that fails to relieve the hypoxic stress, perpetuating a self-reinforcing loop of pathologic angiogenesis.35 Hypoxic conditions created by VEGF pathway inhibitors correlate with upregulation of the MET oncogene, promoting invasive behavior.36,37 MET overexpression has been observed in variety of solid tumors including advanced ovarian cancer.36 Cabozantinib (XL-184) is an oral, potent inhibitor of MET and VEGFR-2, and a phase II trial (100 mg once daily orally over 12 weeks) in advanced, progressive epithelial ovarian cancer showed a promising clinical response rate (overall, 24%).38
Selection of clones resistant to hypoxia.
Pancreatic ductal adenocarcinoma is characteristically hypovascular and in clinical trials has shown no treatment response to bevacizumab.37,39 This tumor type seems to have underlying resistance to hypoxia, indicating a high probability that other cell populations could evolve this trait. Approximately 75% of these tumors carry inactivating mutations of the p53 gene, which may contribute to survival in hypoxic conditions.40,41 Additionally, 75% of pancreatic cancer cell lines in one study constitutively expressed HIF-1α, correlating with resistance to apoptosis induced by hypoxia and/or glucose deprivation. Constitutive HIF-1α expression correlates with increased expression of glucose transporter 1 and Aldolase-A, both of which are associated with anaerobic metabolism.42 These data suggest that preferential utilization of particular metabolic pathways may favor certain cell populations when VEGF-targeted therapy imposes a hypoxic environment. The combination of p53 loss supporting cell survival and the utilization of metabolic pathways that additionally favor survival in a hypoxic environment would offer a simple mechanism by which clonal populations may thrive in the hypoxic environment.
Compensation for VEGF blockade and resistance to hypoxia can be mediated by the microenvironment. Hypoxic conditions promote recruitment of bone marrow–derived cells that include vascular progenitors (eg, endothelial and pericyte progenitors) and vascular modulators.43,44 Vascular modulators are a class of cells including tumor-associated macrophages, immature monocytes, VEGFR-1 hemangiocytes, and CD11b-myeloid cells.45,46 HIF-1α has been implicated in the recruitment of CD45 myeloid cells with various subpopulations expressing Tie-2, VEGFR-1, CD11b, mature F4/80 tumor-associated macrophages, and endothelial and pericyte progenitors.46,47 This environment favors selection of tumor clones protected by CD11b+Gr1+ myeloid cells that express proangiogenic factors such as Bv8, which has been shown to be partially responsible for angiogenesis promotion during VEGF blockade.48,49 These cells facilitate progression to frank carcinoma and may represent an important alternative therapeutic target.50
Indirect Selection Benefit to Tumor Cells
Selection and support of normalized vasculature, supporting tumor cell growth and function.
Anti-VEGF therapy results in vascular normalization that remains of unclear therapeutic significance.35 Abnormal tumor vasculature functions poorly, reflected by chaotic, stagnant, even reversed flow.34,51 The hypoxic, acidic microenvironment promotes protease-mediated matrix remodeling, anchorage-independent growth, and resistance to apoptosis despite increasing genetic instability.52,53 Inhibition of VEGF reduces vessel size and tortuosity; the remaining vessels have greater pericyte coverage, and the basement membrane is normalized.54 Some studies have posited a therapeutic drug-delivery advantage (within a limited therapeutic window) gained by changes in vascular permeability.34 Other studies have suggested that normalization and maturation may represent mechanisms of therapeutic escape. In one study, myeloid cell–driven angiogenesis was selectively ablated in a mouse model of breast cancer, resulting in reduced VEGF, reduced vascular density, increased maturation, and concomitant maturation/normalization; this normalization correlated with increased tumor growth and progression.55
The oxygen-sensing prolyl hydroxylase domain (PHD) proteins may also play a role in vascular normalization and maturation and contribute to more aggressive tumor behavior after antiangiogenic therapy. PHD-2 is an oxygen-sensing enzyme that hydroxylates HIF when sufficient oxygen is available, targeting the protein for degradation.56 Under hypoxic conditions resulting from anti-VEGF treatment, a compensatory release of proangiogenic cytokines generates vessels characterized by irregular borders, hypermobile cells, loosely attached layers, and denuded areas.34,57 In PHD-2 heterozygous mice, endothelial cells upregulate VEGFR-1 and VE-cadherin to stabilize vessels, reduce leakage, and improve vessel perfusion, allowing normalization of neovasculature; homozygous deficiency does not allow this function.57 In some non–small-cell lung cancers, PHDs are present and upregulated, suggesting a possible vascular normalization function associated with overall tumor growth.58
The hyperactivated angiogenic state characteristic of some tumor microenvironments results in a dense, nonproductive vascular network that fails to support tumor growth. Recent data suggest that the Notch ligand Delta-like ligand-4 (DLL4), which is normally induced by VEGF, is actually a negative-feedback regulator of vascular sprouting and branching. In mice lacking DLL4, or in circumstances where it is inhibited, there is excessive and nonproductive angiogenesis (blind-end budding).59 Despite the excess of angiogenesis, tumor growth is decreased, even in tumors resistant to anti-VEGF therapy. The current hypothesis is that in the absence of DLL4, there is inadequate maturation, and therefore inadequate function, of the vessels.59 It has been shown that tumor resistance to bevacizumab can be induced by transfection of DLL4; blockade of Notch signaling reverses this resistance. Mechanisms implicated include increased stromal VEGFR-1, decreased vascular VEGFR-2, diffusely reduced VEGFR-3, and increased signaling through FGF-2/FGF receptor and Ephrin-B4/Ephrin-B2. Cell lines transfected with DLL4 were also resistant to a VEGFR-targeted multikinase inhibitor.60
Among vascular support structures, pericytes provide survival factors and temper the proliferation rate of endothelial cells.61–63 Pericytes are positioned around endothelial cell junctions, provide physical support, and release low levels of endothelial survival factors such as VEGF; for new vascular branches to form, pericytes must detach.64 Within tumors, pericyte coverage is variable, but it is less extensive than seen in normal tissue, and cells take on an abnormal shape and express markers characteristic of immature, less contractile mural cells.64–66 Evidence suggests that tumor cell shedding into the circulation is inversely proportional to pericyte coverage.67 Chronic VEGF expression in the tumor microenvironment seems to interfere with PDGF-B signaling, interfering with vascular smooth muscle cell function and smooth muscle recruitment.68 VEGF blockade increases the signaling of angiopoietin-1, resulting in improved endothelial cell function and pericyte recruitment.69
PDGF/PDGFR signaling itself is implicated in rescue and escape from VEGF blockade.70 The PDGF family provides mitogenic signaling necessary for pericyte recruitment and maturation. Immature vessels with poor pericyte investment seem vulnerable to anti-VEGF treatment, and post-treatment (surviving) vessels seem to have relatively high pericyte coverage.71,72 In one study, PDGF-BB was expressed by endothelial and tumor cells, and PDGFRβ was expressed in pericyte-like cells; PDGF-BB increased the migration and VEGF production of these pericyte-like cells, and these noted functions could be blocked by PDGFRβ inhibitors.73 AX102, a highly specific inhibitor of PDGF-B signaling, was highly effective in combination with bevacizumab in ovarian cancer models.74 Dual targeting of endothelial cells and pericytes has been considered to hold potential as an antivascular therapeutic approach in ovarian carcinoma, and agents such as pazopanib, sunitinib, sorafenib, and BIBF-1120 are being tested in clinical trials.
The role of PlGF in tumor angiogenesis remains controversial and poorly understood. PlGF overexpression in tumors in vivo has been show to correlate with decreased tumor growth, and it has also been shown to correlate with normalization of tumor vasculature, possibly through heterodimerization with VEGF, reducing VEGF potency.75,76 PlGF-null mice demonstrate reduced response to VEGF. However, PlGF is chemotactic for endothelial cells and monocytes in vitro; it may be involved with mobilization of bone marrow–derived cells, and it increases the response of cultured endothelial cells to VEGF-induced survival, proliferation, and migration.77,78 Aflibercept (VEGF Trap) is a protein that contains the VEGF-binding regions of VEGFR-1 and VEGFR-2 fused to the Fc portion of human IgG1. It acts as a high-affinity soluble VEGFR decoy receptor and therefore inhibits the activity of both VEGF and PlGF.79 Two randomized phase II studies showed that even in heavily pretreated patients, single-agent aflibercept could induce tumor response and delay progression.80,81 A recent combined phase I/II trial of docetaxel plus aflibercept in patients with recurrent ovarian, primary peritoneal, or fallopian tube cancer resulted in a 54% response rate, and 24% of patients had stable disease.82
Ang1 and Ang2 exist in balance to promote organization and maturation of neovasculature. Overexpression of Ang1 compared with Ang2 results in dense hypervascularization with large vessels, and excess Ang2 binding to Tie-2 results in destabilized and leaky blood vessels.83 Perivascular Tie-2–expressing monocytes are also proangiogenic.35 Coordinated Ang1 and Ang2 signaling through Tie-2 receptors is thought to be an alternative pathway to VEGF through which neovascularization can occur, and coordinated upregulation may represent an additional mode of anti-VEGF–therapy bypass.
Selection of subpopulations capable of co-opting normal/existing vasculature.
Grade 2 and 3 astrocytomas have been observed to develop neovasculature without an identified proliferative endothelial component.84,85 Vascular acquisition in this case seems to be via perivascular tumor invasion with incorporation of normal vasculature into the tumor structure. As an apparent corollary, three human studies reported patients with multifocal recurrence while receiving bevacizumab therapy.37,86,87 These findings, in conjunction with data suggesting that fibroblasts can draw existing vessels toward and into a fresh wound, point toward cell populations that have the phenotypic capacity to home toward existing vasculature and to incorporate it rather than relying on a cytokine-driven neovascular response. These clonal populations would be favored in an environment exposed to antiangiogenic therapy.
Endothelial cell genetic instability and resistance to therapy.
Contrary to previous dogma stating that endothelial cells within tumors are genetically stable and therefore less likely to evolve resistance to therapy, recent work has identified cytogenetic abnormalities in human tumor-associated endothelial cells.88,89 Although they have not been validated, these studies raise the possibility that high rates of endothelial cell mutation within tumors could contribute to therapeutic resistance to a greater degree than previously surmised.88,89 What remains to be elucidated is why tumor-based endothelial cells have significant cytogenetic instability and whether this high rate of mutation would confer the capacity to select for unique subpopulations of endothelial cells that would develop therapeutic resistance over (relatively) short periods of time.90
ATTRACTIVENESS OF MULTIPLE-PATHWAY INHIBITORS
Given our increasing knowledge regarding the myriad alternative pathways for the development and maintenance of tumor vasculature, several new inhibitors have been engineered to block multiple proangiogenic signaling pathways. Sorafenib, axitinib, sunitinib, cediranib, and pazopanib variously target proangiogenic signaling cascades and are in clinical trials. The most promising of these is a phase III trial of pazopanib in advanced renal cell carcinoma. Compared with placebo, pazopanib improved PFS from 4.2 to 9.2 months. The difference in PFS was even more striking in patients who were treatment naive (11.1 v 2.8 months; P < .001). Response rate was also improved (30% v 3%), and median duration of response was > 1 year.91
Other drugs have been developed to target VEGF and FGF signaling. Brivanib (dual FGF/VEGF inhibitor) has shown activity in preclinical pancreatic cancer models that have developed resistance to VEGF inhibition. BIBF-1120 is a unique, triple-angiokinase inhibitor of VEGFR, PDGFR, and FGF receptor. In a randomized, phase II placebo-controlled trial, patients who had completed chemotherapy for relapsed ovarian cancer with evidence of response were treated with BIBF-1120. Three-year PFS rates were 16.3% and 5.0% in the BIBF 1120 and placebo groups, respectively (hazard ratio, 0.65; 95% CI, 0.42 to 1.02; P = .06).92 A current phase III (NCT01015118) trial is evaluating the addition of BIBF-1120 to carboplatin/paclitaxel in first-line chemotherapy in ovarian cancer.93
AMG386 is an investigational, angiopoietin antagonist peptide-Fc fusion protein that selectively binds Ang1 and Ang2. This binding prevents the interaction of Ang1 and Ang2 with Tie-2 and inhibits tumor endothelial cell proliferation and tumor growth.94 In a randomized, double-blind, placebo-controlled phase II study to evaluate the safety and tolerability of AMG386 in combination with paclitaxel, the addition of AMG386 to paclitaxel demonstrated dose-responsive improvements in PFS with a manageable safety profile distinct from that of VEGF inhibition.95 AMG386 has entered phase III investigation in the setting of recurrent ovarian cancer.
Attention has also shifted to nonreceptor kinases such as Src and Fak, which are implicated in multiple tumorigenic behaviors including angiogenesis.96 The Src/Fak complex, in particular, has been the target of drug development.97–99 In phase II investigation, the Src inhibitor dasatinib has been used as first-line therapy in advanced non–small-cell lung cancer. Of 34 patients, the overall disease control rate was 43%; however, only one patient had a partial response. Interestingly, 11 patients had a metabolic response, suggesting poor patient selection and/or dose selection.100 An additional target of interest is EZH2. Increased EZH2 in endothelial cells results from paracrine VEGF stimulation, resulting in silencing of VASH1. Silencing of EZH2 in the endothelium results in decreased tumor angiogenesis and reduced ovarian cancer growth in an orthotopic model. EZH2 silencing in tumor cells has a similar effect.101 EZH2 may also be subject to regulation by FGF-2 and miR101 signaling.102,103
STRATEGIC CLINICAL INVESTIGATION
Given the exponential number of new drug combinations, as well as the cost of drug trials, careful consideration must be given to trial design and rational combination therapy. Conventional clinical trials rely on randomly assigning a heterogeneous array of eligible patients to a small number of therapeutic arms. These studies consistently fail to acknowledge the fundamental biologic variability of cancer and thus necessarily expose patients to potentially toxic medication in the absence of an expectation of benefit. Because of limited resources, validated, biomarker-driven designs are of great interest. These smart trials have the potential of enrolling fewer patients, anticipating larger treatment effects, and reducing unnecessary harms.
An alternative approach may be trial designs in which a pharmacodynamic or pharmacokinetic marker could be assessed early in therapy, and if the marker is undermodulated, the experimental agent could be withdrawn. In GOG 262 (randomized, phase III trial in primary ovarian cancer that includes a bevacizumab maintenance arm), a computed tomography–based perfusion scan is being evaluated to identify those who might benefit from long-term bevacizumab exposure. With regard to biologic tumor variation, circulating tumor cells and cell-free nucleic acids may offer a real-time assay for making more informed choices regarding dose and duration of therapy.
NEW DIRECTIONS
Surgery and cytotoxic chemotherapy remain the mainstays of primary therapy. The use of targeted biologics combined with one another or with traditional therapy is intended to increase tumor response without significant increases in toxicity. Some have interpreted results of phase III studies of biologics as reflecting poor drug responsiveness. The true shortcoming of these trials lies in the failure of the trial design to account for tumor cell heterogeneity, both among individuals and within a given individual's tumor cell population; different underlying aberrant biology should be anticipated to be vulnerable to different drug targeting. As targeted biologic therapies push forward, it becomes increasingly important to identify susceptible tumor cell populations within a given patient to identify which cytotoxics and biologic inhibitors may provide that individual with clinical benefit.
Fresh targets are needed, and the microenvironment provides a rich array of necessary, targetable, angiogenic participants such as pericytes, endothelial cells, and bone marrow–derived precursors. Although multiple-kinase inhibitors have risk of increased toxicity, carefully considered combinatorial choices in well-designed trials hold real promise of clinical benefit. Additionally, vascular disrupting agents are a unique class of drugs that aim to collapse existing vascular structures.104 Among these, combretastatin A-4 (modified to AC7700; now known as ombrabulin) was encouraging in preclinical sarcoma and lung cancer models.105 CA4P, pinabulin, and ombrabulin have shown some promise in phase II clinical trials.106 Combination with traditional antiangiogenic agents might further improve the overall antitumor effect.107 Table 1 summarizes selected classes of agents in current development that represent novel and promising approaches to targeting the biology of angiogenesis through pathways other than VEGF signaling.
Table 1.
Selected Classes of Agents That Represent Novel Approaches to Targeting Biology of Angiogenesis Through Pathways Other Than VEGF Signaling
Mechanism of Action/Class | Representative Drugs | Current Clinical Phase |
---|---|---|
Vascular disrupting agents | ASA-404 | II-III |
AVE-8062 | II-III | |
Ombrabulin | II-III | |
CA4P | II-III | |
Crolibulin | I-II | |
DMXAA | I-II | |
NPI-2358 | I-II | |
Plinabulin | I-II | |
Soblidotin | I-II | |
Denibulin | I | |
Oxi-4503 | I | |
ZD-6126 | I | |
FGFR targeting | Dovitinib | III |
BIBF-1120 | II-III | |
Brivanib | II-III | |
Pazopanib | II-III | |
BGJ-398 | I | |
Angiopoietin targeting | AMG-386 | II-III |
MEDI-3617 | I-II | |
EphrinA2 targeting | Dasatinib | II-III |
DLL4/Notch targeting | MEDI-0639 | I |
REGN-421 | I | |
Demcizumab | I | |
PI3K/mTOR targeting | BKM-120 | II-III |
Everolimus | II-III | |
Temsirolimus | II-III | |
BEZ-235 | I-II | |
BGT-226 | I-II | |
DS7-423 | I-II | |
GDC-0941 | I-II | |
GSK-2110183 | I-II | |
PF-04691502 | I-II | |
PX-866 | I-II | |
XL-147 | I-II | |
XL-765 | I-II | |
INK-1117 | I | |
GSK-1059615 | I | |
GSK-2126458 | I | |
PKI-179 | I | |
SF-1126 | I | |
ZSTK-474 | I | |
EGFR targeting | Cetuximab | II-III |
Erlotinib | II-III | |
Gefitinib | II-III | |
Panitumumab | II-III | |
Vandetanib | II-III | |
Lapatanib | I-II | |
MM-121 | I-II | |
MET targeting | Cabozantinib | II-III |
PDGFR targeting | Axitinib | II-III |
BIBF1120 | II-III | |
Cediranib | II-III | |
Dovitinib | II-III | |
Pazopanib | II-III | |
Sunitinib | II-III | |
Sorafenib | II-III | |
PlGF targeting | Aflibercept | II-III |
Src/Fak complex targeting | Dasatinib | II-III |
Sunitinib | II-III | |
KX2-391 | I-II | |
IL-6 targeting | Siltuximab | II-III |
AKT targeting | MK-2206 | I-II |
IGF1-R targeting | MK-0646 | I-II |
Abbreviations: DLL4, delta-like ligand-4; EGFR, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; IGF1-R, insulin-like growth factor receptor 1; IL-6, interleukin-6; mTOR, mammalian target of rapamycin; PDGFR, platelet-derived growth factor receptor; PI3K, phosphoinositide 3– kinase; PlGF, placental growth factor.
A recent, compelling strategy involves metronomic dosing of chemotherapy, in which lower doses of cytotoxic agents at more frequent intervals is thought to target the tumor vasculature.108,109 Clinically, semimetronomic, or dose-dense, regimens have shown intriguing superiority to traditional dosing regimens.110–113
MicroRNAs are a recent addition to the angiogenesis literature. More than 20 microRNAs have been identified that either target genes involved in angiogenesis or respond to angiogenic stimuli such as VEGF.114 These microRNAs may represent transmissible genetic elements that that can be dysregulated by tumors. The new finding of endothelial genetic instability may imply the presence of transposable genetic elements by which a genetically unstable cell can transmit unstable elements to surrounding cells.
Finally, the hematologic contribution to angiogenesis and tumorigenesis must be more fully explored and targeted. Transgenic mouse models of de novo skin carcinogenesis provide evidence that early hyperplasia and early increases in angiogenesis are correlated with mast cell recruitment and degranulation.115 B lymphocytes initiate reactions resulting in mast cell recruitment, which correlates with increased angiogenesis and progression from hyperplasia to dysplasia to carcinoma.116 Causality is supported by the fact that blocking B-lymphocyte responses, mast cell infiltration, and recruitment of immature myeloid cells decreases angiogenesis and tumor progression.115 In a transgenic mouse model of breast cancer, the transition from adenoma to mammary intraepithelial neoplasia was accompanied by activation of angiogenesis that coincided with macrophage infiltration of the tumor. Depletion of these macrophages reduces angiogenesis and tumor progression; the provision of VEGF after macrophage depletion restores the progression of malignancy.117 Platelets are targetable mediators of angiogenesis. Purinergic signaling from platelets influences cell migration and proliferation.118–120 Alpha granule contents are known mediators of pro- and antiangiogenic effectors, supporting the notion that platelets may be sophisticated and integrated angiogenesis regulators.121 IL-6 is implicated in paraneoplastic thrombocytosis, and targeting with siltuximab (a monoclonal antibody targeting IL-6) was shown in preclinical models to abrogate thrombocytosis and to have antitumor effects additive to conventional cytotoxic agents.
A better understanding of the biology of hypoxia and reoxygenation, including the biochemical, microenvironmental, and ecologic effects, could provide a model of solid tumor biology, facilitating wise choices of biomarkers and therapeutic interventions. Similarly, it may provide insights into natural history and prognosis that would aid in more holistic decision making for and by the patient as we aim to enter an age of individually focused care and improved outcomes.
Acknowledgment
We thank Chad Pecot, MD, and Behrouz Zand, MD, for their helpful discussions. Neither of these individuals received compensation for their contributions.
Footnotes
Supported in part by the Gynecologic Cancer Foundation, National Institutes of Health (NIH; Grants No. CA 110793, 109298, P50 CA083639, P50 CA098258, CA128797, RC2GM092599, and U54 CA151668), Ovarian Cancer Research Fund (Program Project Development grant), Department of Defense (Grants No. OC073399, W81XWH-10-1-0158, and BC085265), Zarrow Foundation, Marcus Foundation, Estate of C.G. Johnson Jr, Gilder Foundation, Betty Anne Asche Murray Distinguished Professorship, and Ann Rife Cox Chair in Gynecology and Cancer Prevention Research Institute of Texas (Grants No. RP120214 and RP110595), and by T32 Training Grant No. CA101642 from the National Cancer Institute, Department of Health and Human Services, NIH, and a career development grant from the Ovarian Cancer National Alliance (J.N.B.-M.).
Authors' disclosures of potential conflicts of interest and author contributions are found at the end of this article.
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
Although all authors completed the disclosure declaration, the following author(s) indicated a financial or other interest that is relevant to the subject matter under consideration in this article. Certain relationships marked with a “U” are those for which no compensation was received; those relationships marked with a “C” were compensated. For a detailed description of the disclosure categories, or for more information about ASCO's conflict of interest policy, please refer to the Author Disclosure Declaration and the Disclosures of Potential Conflicts of Interest section in Information for Contributors.
Employment or Leadership Position: None Consultant or Advisory Role: Robert L. Coleman, Genentech-Roche (U), GlaxoSmithKline (U), AstraZeneca (U), Boehringer Ingelheim (U) Stock Ownership: None Honoraria: Robert L. Coleman, Genentech-Roche, GlaxoSmithKline, AstraZeneca, Boehringer Ingelheim Research Funding: Robert L. Coleman, sanofi-aventis, Amgen, Novartis, Merck, Merrimack Pharmaceuticals Expert Testimony: None Other Remuneration: None
AUTHOR CONTRIBUTIONS
Financial support: Robert L. Coleman, Anil K. Sood
Administrative support: All authors
Provision of study materials or patients: All authors
Manuscript writing: All authors
Final approval of manuscript: All authors
REFERENCES
- 1.Folkman J, Merler E, Abernathy C, et al. Isolation of a tumor factor responsible for angiogenesis. J Exp Med. 1971;133:275–288. doi: 10.1084/jem.133.2.275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Potente M, Gerhardt H, Carmeliet P. Basic and therapeutic aspects of angiogenesis. Cell. 2011;146:873–887. doi: 10.1016/j.cell.2011.08.039. [DOI] [PubMed] [Google Scholar]
- 3.Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 4.Spannuth WA, Sood AK, Coleman RL. Angiogenesis as a strategic target for ovarian cancer therapy. Nat Clin Pract Oncol. 2008;5:194–204. doi: 10.1038/ncponc1051. [DOI] [PubMed] [Google Scholar]
- 5.Eskander RN, Randall LM. Bevacizumab in the treatment of ovarian cancer. Biologics. 2011;5:1–5. doi: 10.2147/BTT.S13071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Beal K, Abrey LE, Gutin PH. Antiangiogenic agents in the treatment of recurrent or newly diagnosed glioblastoma: Analysis of single-agent and combined modality approaches. Radiat Oncol. 2011;6:2. doi: 10.1186/1748-717X-6-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Casanovas O, Hicklin DJ, Bergers G, et al. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell. 2005;8:299–309. doi: 10.1016/j.ccr.2005.09.005. [DOI] [PubMed] [Google Scholar]
- 8.Xu L, Duda DG, di Tomaso E, et al. Direct evidence that bevacizumab, an anti-VEGF antibody, up-regulates SDF1alpha, CXCR4, CXCL6, and neuropilin 1 in tumors from patients with rectal cancer. Cancer Res. 2009;69:7905–7910. doi: 10.1158/0008-5472.CAN-09-2099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hu DE, Fan TP. Suppression of VEGF-induced angiogenesis by the protein tyrosine kinase inhibitor, lavendustin A. Br J Pharmacol. 1995;114:262–268. doi: 10.1111/j.1476-5381.1995.tb13221.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Rosen LS. Clinical experience with angiogenesis signaling inhibitors: Focus on vascular endothelial growth factor (VEGF) blockers. Cancer Control. 2002;9(suppl):36–44. doi: 10.1177/107327480200902S05. [DOI] [PubMed] [Google Scholar]
- 11.Jackson DB, Sood AK. Personalized cancer medicine-advances and socio-economic challenges. Nat Rev Clin Oncol. 2011;8:735–741. doi: 10.1038/nrclinonc.2011.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Burger RA, Brady MF, Bookman MA, et al. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med. 2011;365:2473–2483. doi: 10.1056/NEJMoa1104390. [DOI] [PubMed] [Google Scholar]
- 13.Perren TJ, Swart AM, Pfisterer J, et al. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med. 2011;365:2484–2496. doi: 10.1056/NEJMoa1103799. [DOI] [PubMed] [Google Scholar]
- 14.Scagliotti G, Novello S, von Pawel J, et al. Phase III study of carboplatin and paclitaxel alone or with sorafenib in advanced non–small-cell lung cancer. J Clin Oncol. 2010;28:1835–1842. doi: 10.1200/JCO.2009.26.1321. [DOI] [PubMed] [Google Scholar]
- 15.Barrios CH, Liu MC, Lee SC, et al. Phase III randomized trial of sunitinib versus capecitabine in patients with previously treated HER2-negative advanced breast cancer. Breast Cancer Res Treat. 2010;121:121–131. doi: 10.1007/s10549-010-0788-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–615. doi: 10.1038/nature10166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.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. 2008;14:1529–1539. doi: 10.1158/1078-0432.CCR-07-4126. [DOI] [PubMed] [Google Scholar]
- 18.Ebos JM, Lee CR, Christensen JG, et al. Multiple circulating proangiogenic factors induced by sunitinib malate are tumor-independent and correlate with antitumor efficacy. Proc Natl Acad Sci U S A. 2007;104:17069–17074. doi: 10.1073/pnas.0708148104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lieu C, Heymach J, Overman M, et al. Beyond VEGF: Inhibition of the fibroblast growth factor pathway and antiangiogenesis. Clin Cancer Res. 2011;17:6130–6139. doi: 10.1158/1078-0432.CCR-11-0659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Greenhalgh DG, Sprugel KH, Murray MJ, et al. PDGF and FGF stimulate wound healing in the genetically diabetic mouse. Am J Pathol. 1990;136:1235–1246. [PMC free article] [PubMed] [Google Scholar]
- 21.Tassi E, Al-Attar A, Aigner A, et al. Enhancement of fibroblast growth factor (FGF) activity by an FGF-binding protein. J Biol Chem. 2001;276:40247–40253. doi: 10.1074/jbc.M104933200. [DOI] [PubMed] [Google Scholar]
- 22.Kopetz S, Hoff PM, Morris JS, et al. Phase II trial of infusional fluorouracil, irinotecan, and bevacizumab for metastatic colorectal cancer: Efficacy and circulating angiogenic biomarkers associated with therapeutic resistance. J Clin Oncol. 2010;28:453–459. doi: 10.1200/JCO.2009.24.8252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Batchelor TT, Sorensen AG, di Tomaso E, et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007;11:83–95. doi: 10.1016/j.ccr.2006.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Mizukami Y, Jo WS, Duerr EM, et al. Induction of interleukin-8 preserves the angiogenic response in HIF-1alpha-deficient colon cancer cells. Nat Med. 2005;11:992–997. doi: 10.1038/nm1294. [DOI] [PubMed] [Google Scholar]
- 25.Tian J, Lambertz I, Berton TR, et al. Transgenic insulin-like growth factor-1 stimulates activation of COX-2 signaling in mammary glands. Mol Carcinog. doi: 10.1002/mc.20868. [epub ahead of print on October 17, 2011] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hilmi C, Guyot M, Pagès G. VEGF spliced variants: Possible role of anti-angiogenesis therapy. J Nucleic Acids. 2012;2012:162692. doi: 10.1155/2012/162692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhou S, Gu L, He J, et al. MDM2 regulates vascular endothelial growth factor mRNA stabilization in hypoxia. Mol Cell Biol. 2011;31:4928–4937. doi: 10.1128/MCB.06085-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Karar J, Maity A. PI3K/AKT/mTOR pathway in angiogenesis. Front Mol Neurosci. 2011;4:51. doi: 10.3389/fnmol.2011.00051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Viloria-Petit A, Crombet T, Jothy S, et al. Acquired resistance to the antitumor effect of epidermal growth factor receptor-blocking antibodies in vivo: A role for altered tumor angiogenesis. Cancer Res. 2001;61:5090–5101. [PubMed] [Google Scholar]
- 30.Natale RB, Thongprasert S, Greco FA, et al. Phase III trial of vandetanib compared with erlotinib in patients with previously treated advanced non–small-cell lung cancer. J Clin Oncol. 2011;29:1059–1066. doi: 10.1200/JCO.2010.28.5981. [DOI] [PubMed] [Google Scholar]
- 31.Rubenstein JL, Kim J, Ozawa T, et al. 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]
- 32.Pàez-Ribes M, Allen E, Hudock J, et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 2009;15:220–231. doi: 10.1016/j.ccr.2009.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ebos JM, Lee CR, Cruz-Munoz W, et al. Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell. 2009;15:232–239. doi: 10.1016/j.ccr.2009.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Jain RK. Normalization of tumor vasculature: An emerging concept in antiangiogenic therapy. Science. 2005;307:58–62. doi: 10.1126/science.1104819. [DOI] [PubMed] [Google Scholar]
- 35.Carmeliet P, Jain RK. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat Rev Drug Discov. 2011;10:417–427. doi: 10.1038/nrd3455. [DOI] [PubMed] [Google Scholar]
- 36.Eder JP, Vande Woude GF, Boerner SA, et al. Novel therapeutic inhibitors of the c-Met signaling pathway in cancer. Clin Cancer Res. 2009;15:2207–2214. doi: 10.1158/1078-0432.CCR-08-1306. [DOI] [PubMed] [Google Scholar]
- 37.Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008;8:592–603. doi: 10.1038/nrc2442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Buckanovich R, Berger R, Sella A, et al. Activity of cabozantinib (XL184) in advanced ovarian cancer patients (pts): Results from a phase II randomized discontinuation trial (RDT) J Clin Oncol. 2011;29(suppl 15):334s. abstr 5008. [Google Scholar]
- 39.Kindler HL, Friberg G, Singh DA, et al. Phase II trial of bevacizumab plus gemcitabine in patients with advanced pancreatic cancer. J Clin Oncol. 2005;23:8033–8040. doi: 10.1200/JCO.2005.01.9661. [DOI] [PubMed] [Google Scholar]
- 40.Schneider G, Schmid RM. Genetic alterations in pancreatic carcinoma. Mol Cancer. 2003;2:15. doi: 10.1186/1476-4598-2-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Yu JL, Rak JW, Coomber BL, et al. Effect of p53 status on tumor response to antiangiogenic therapy. Science. 2002;295:1526–1528. doi: 10.1126/science.1068327. [DOI] [PubMed] [Google Scholar]
- 42.Akakura N, Kobayashi M, Horiuchi I, et al. Constitutive expression of hypoxia-inducible factor-1alpha renders pancreatic cancer cells resistant to apoptosis induced by hypoxia and nutrient deprivation. Cancer Res. 2001;61:6548–6554. [PubMed] [Google Scholar]
- 43.De Falco E, Porcelli D, Torella AR, et al. SDF-1 involvement in endothelial phenotype and ischemia-induced recruitment of bone marrow progenitor cells. Blood. 2004;104:3472–3482. doi: 10.1182/blood-2003-12-4423. [DOI] [PubMed] [Google Scholar]
- 44.Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004;10:858–864. doi: 10.1038/nm1075. [DOI] [PubMed] [Google Scholar]
- 45.Yang L, DeBusk LM, Fukuda K, et al. Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell. 2004;6:409–421. doi: 10.1016/j.ccr.2004.08.031. [DOI] [PubMed] [Google Scholar]
- 46.Du R, Lu KV, Petritsch C, et al. HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell. 2008;13:206–220. doi: 10.1016/j.ccr.2008.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Aghi M, Cohen KS, Klein RJ, et al. Tumor stromal-derived factor-1 recruits vascular progenitors to mitotic neovasculature, where microenvironment influences their differentiated phenotypes. Cancer Res. 2006;66:9054–9064. doi: 10.1158/0008-5472.CAN-05-3759. [DOI] [PubMed] [Google Scholar]
- 48.Shojaei F, Wu X, Malik AK, et al. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat Biotechnol. 2007;25:911–920. doi: 10.1038/nbt1323. [DOI] [PubMed] [Google Scholar]
- 49.Shojaei F, Wu X, Qu X, et al. G-CSF-initiated myeloid cell mobilization and angiogenesis mediate tumor refractoriness to anti-VEGF therapy in mouse models. Proc Natl Acad Sci U S A. 2009;106:6742–6747. doi: 10.1073/pnas.0902280106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Murdoch C, Muthana M, Coffelt SB, et al. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8:618–631. doi: 10.1038/nrc2444. [DOI] [PubMed] [Google Scholar]
- 51.Fukumura D, Duda DG, Munn LL, et al. Tumor microvasculature and microenvironment: Novel insights through intravital imaging in pre-clinical models. Microcirculation. 2010;17:206–225. doi: 10.1111/j.1549-8719.2010.00029.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lunt SJ, Chaudary N, Hill RP. The tumor microenvironment and metastatic disease. Clin Exp Metastasis. 2009;26:19–34. doi: 10.1007/s10585-008-9182-2. [DOI] [PubMed] [Google Scholar]
- 53.Graeber TG, Osmanian C, Jacks T, et al. Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature. 1996;379:88–91. doi: 10.1038/379088a0. [DOI] [PubMed] [Google Scholar]
- 54.Tong RT, Boucher Y, Kozin SV, et al. Vascular normalization by vascular endothelial growth factor receptor 2 blockade induces a pressure gradient across the vasculature and improves drug penetration in tumors. Cancer Res. 2004;64:3731–3736. doi: 10.1158/0008-5472.CAN-04-0074. [DOI] [PubMed] [Google Scholar]
- 55.Stockmann C, Doedens A, Weidemann A, et al. Deletion of vascular endothelial growth factor in myeloid cells accelerates tumorigenesis. Nature. 2008;456:814–818. doi: 10.1038/nature07445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Aragonés J, Fraisl P, Baes M, et al. Oxygen sensors at the crossroad of metabolism. Cell Metab. 2009;9:11–22. doi: 10.1016/j.cmet.2008.10.001. [DOI] [PubMed] [Google Scholar]
- 57.Mazzone M, Dettori D, Leite de Oliveira R, et al. Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization. Cell. 2009;136:839–851. doi: 10.1016/j.cell.2009.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chen SF, Zhang J, Li XB, et al. The expression of prolyl hydroxylase domain enzymes are up-regulated and negatively correlated with Bcl-2 in non-small cell lung cancer. Mol Cell Biochem. 2011;358:257–263. doi: 10.1007/s11010-011-0976-1. [DOI] [PubMed] [Google Scholar]
- 59.Thurston G, Noguera-Troise I, Yancopoulos GD. The Delta paradox: DLL4 blockade leads to more tumour vessels but less tumour growth. Nat Rev Cancer. 2007;7:327–331. doi: 10.1038/nrc2130. [DOI] [PubMed] [Google Scholar]
- 60.Li JL, Sainson RC, Oon CE, et al. DLL4-Notch signaling mediates tumor resistance to anti-VEGF therapy in vivo. Cancer Res. 2011;71:6073–6083. doi: 10.1158/0008-5472.CAN-11-1704. [DOI] [PubMed] [Google Scholar]
- 61.Darland DC, Massingham LJ, Smith SR, et al. Pericyte production of cell-associated VEGF is differentiation-dependent and is associated with endothelial survival. Dev Biol. 2003;264:275–288. doi: 10.1016/j.ydbio.2003.08.015. [DOI] [PubMed] [Google Scholar]
- 62.Song S, Ewald AJ, Stallcup W, et al. PDGFRbeta+ perivascular progenitor cells in tumours regulate pericyte differentiation and vascular survival. Nat Cell Biol. 2005;7:870–879. doi: 10.1038/ncb1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Orlidge A, D'Amore PA. Inhibition of capillary endothelial cell growth by pericytes and smooth muscle cells. J Cell Biol. 1987;105:1455–1462. doi: 10.1083/jcb.105.3.1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Díaz-Flores L, Gutiérrez R, Madrid JF, et al. Pericytes: Morphofunction, interactions and pathology in a quiescent and activated mesenchymal cell niche. Histol Histopathol. 2009;24:909–969. doi: 10.14670/HH-24.909. [DOI] [PubMed] [Google Scholar]
- 65.Morikawa S, Baluk P, Kaidoh T, et al. Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors. Am J Pathol. 2002;160:985–1000. doi: 10.1016/S0002-9440(10)64920-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ozawa MG, Yao VJ, Chanthery YH, et al. Angiogenesis with pericyte abnormalities in a transgenic model of prostate carcinoma. Cancer. 2005;104:2104–2115. doi: 10.1002/cncr.21436. [DOI] [PubMed] [Google Scholar]
- 67.Gerhardt H, Semb H. Pericytes: Gatekeepers in tumour cell metastasis? J Mol Med (Berl) 2008;86:135–144. doi: 10.1007/s00109-007-0258-2. [DOI] [PubMed] [Google Scholar]
- 68.Greenberg JI, Shields DJ, Barillas SG, et al. A role for VEGF as a negative regulator of pericyte function and vessel maturation. Nature. 2008;456:809–813. doi: 10.1038/nature07424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Winkler F, Kozin SV, Tong RT, et al. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: Role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell. 2004;6:553–563. doi: 10.1016/j.ccr.2004.10.011. [DOI] [PubMed] [Google Scholar]
- 70.Hamdan R, Zhou Z, Kleinerman ES. SDF-1α induces PDGF-B expression and the differentiation of bone marrow cells into pericytes. Mol Cancer Res. 2011;9:1462–1470. doi: 10.1158/1541-7786.MCR-11-0190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Bergers G, Song S, Meyer-Morse N, et al. Benefits of targeting both pericytes and endothelial cells in the tumor vasculature with kinase inhibitors. J Clin Invest. 2003;111:1287–1295. doi: 10.1172/JCI17929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lu C, Kamat AA, Lin YG, et al. Dual targeting of endothelial cells and pericytes in antivascular therapy for ovarian carcinoma. Clin Cancer Res. 2007;13:4209–4217. doi: 10.1158/1078-0432.CCR-07-0197. [DOI] [PubMed] [Google Scholar]
- 73.Lu C, Thaker PH, Lin YG, et al. Impact of vessel maturation on antiangiogenic therapy in ovarian cancer. Am J Obstet Gynecol. 2008;198:477.e1–477.e9. doi: 10.1016/j.ajog.2007.12.028. discussion 477.e9-477.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Lu C, Shahzad MM, Moreno-Smith M, et al. Targeting pericytes with a PDGF-B aptamer in human ovarian carcinoma models. Cancer Biol Ther. 2010;9:176–182. doi: 10.4161/cbt.9.3.10635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hedlund EM, Hosaka K, Zhong Z, et al. Malignant cell-derived PlGF promotes normalization and remodeling of the tumor vasculature. Proc Natl Acad Sci U S A. 2009;106:17505–17510. doi: 10.1073/pnas.0908026106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Fischer C, Mazzone M, Jonckx B, et al. FLT1 and its ligands VEGFB and PlGF: Drug targets for anti-angiogenic therapy? Nat Rev Cancer. 2008;8:942–956. doi: 10.1038/nrc2524. [DOI] [PubMed] [Google Scholar]
- 77.Carmeliet P, Moons L, Luttun A, et al. Synergism between vascular endothelial growth factor and placental growth factor contributes to angiogenesis and plasma extravasation in pathological conditions. Nat Med. 2001;7:575–583. doi: 10.1038/87904. [DOI] [PubMed] [Google Scholar]
- 78.Mac Gabhann F, Popel AS. Model of competitive binding of vascular endothelial growth factor and placental growth factor to VEGF receptors on endothelial cells. Am J Physiol Heart Circ Physiol. 2004;286:153–164. doi: 10.1152/ajpheart.00254.2003. [DOI] [PubMed] [Google Scholar]
- 79.Holash J, Davis S, Papadopoulos N, et al. VEGF-Trap: A VEGF blocker with potent antitumor effects. Proc Natl Acad Sci U S A. 2002;99:11393–11398. doi: 10.1073/pnas.172398299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Tew WP, Colombo N, Ray-Coquard I, et al. VEGF-Trap for patients (pts) with recurrent platinum-resistant epithelial ovarian cancer (EOC): Preliminary results of a randomized, multicenter phase II study. J Clin Oncol. 2007;25(suppl 18):276s. abstr 5508. [Google Scholar]
- 81.Vergote I, Amant F, Advani S, et al. Intravenous aflibercept (VEGF Trap) in advanced ovarian cancer patients with recurrent symptomatic malignant ascites: Main efficacy and safety results of a phase II, randomized, double-blind, placebo-controlled study. Presented at the 16th European Society of Gynaecological Oncology; October 11-15, 2009; Belgrade, Serbia. [Google Scholar]
- 82.Coleman RL, Duska LR, Ramirez PT, et al. Phase 1-2 study of docetaxel plus aflibercept in patients with recurrent ovarian, primary peritoneal, or fallopian tube cancer. Lancet Oncol. 2011;12:1109–1117. doi: 10.1016/S1470-2045(11)70244-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Augustin HG, Koh GY, Thurston G, et al. Control of vascular morphogenesis and homeostasis through the angiopoietin-Tie system. Nat Rev Mol Cell Biol. 2009;10:165–177. doi: 10.1038/nrm2639. [DOI] [PubMed] [Google Scholar]
- 84.Kaur B, Tan C, Brat DJ, et al. Genetic and hypoxic regulation of angiogenesis in gliomas. J Neurooncol. 2004;70:229–243. doi: 10.1007/s11060-004-2752-5. [DOI] [PubMed] [Google Scholar]
- 85.Berger MS, Wilson CB. The Gliomas. Philadelphia, PA: W.B. Saunders; 1999. [Google Scholar]
- 86.Norden AD, Young GS, Setayesh K, et al. Bevacizumab for recurrent malignant gliomas: Efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70:779–787. doi: 10.1212/01.wnl.0000304121.57857.38. [DOI] [PubMed] [Google Scholar]
- 87.Narayana A, Kelly P, Golfinos J, et al. Antiangiogenic therapy using bevacizumab in recurrent high-grade glioma: Impact on local control and patient survival. J Neurosurg. 2009;110:173–180. doi: 10.3171/2008.4.17492. [DOI] [PubMed] [Google Scholar]
- 88.Akino T, Hida K, Hida Y, et al. Cytogenetic abnormalities of tumor-associated endothelial cells in human malignant tumors. Am J Pathol. 2009;175:2657–2667. doi: 10.2353/ajpath.2009.090202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Xiong YQ, Sun HC, Zhang W, et al. Human hepatocellular carcinoma tumor-derived endothelial cells manifest increased angiogenesis capability and drug resistance compared with normal endothelial cells. Clin Cancer Res. 2009;15:4838–4846. doi: 10.1158/1078-0432.CCR-08-2780. [DOI] [PubMed] [Google Scholar]
- 90.Dudley AC, Klagsbrun M. Tumor endothelial cells join the resistance. Clin Cancer Res. 2009;15:4787–4789. doi: 10.1158/1078-0432.CCR-09-0902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: Results of a randomized phase III trial. J Clin Oncol. 2010;28:1061–1068. doi: 10.1200/JCO.2009.23.9764. [DOI] [PubMed] [Google Scholar]
- 92.Ledermann JA, Hackshaw A, Kaye S, et al. Randomized phase II placebo-controlled trial of maintenance therapy using the oral triple angiokinase inhibitor BIBF 1120 after chemotherapy for relapsed ovarian cancer. J Clin Oncol. 2011;29:3798–3804. doi: 10.1200/JCO.2010.33.5208. [DOI] [PubMed] [Google Scholar]
- 93.Allen E, Walters IB, Hanahan D. Brivanib, a dual FGF/VEGF inhibitor, is active both first and second line against mouse pancreatic neuroendocrine tumors developing adaptive/evasive resistance to VEGF inhibition. Clin Cancer Res. 2011;17:5299–5310. doi: 10.1158/1078-0432.CCR-10-2847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Herbst RS, Hong D, Chap L, et al. Safety, pharmacokinetics, and antitumor activity of AMG 386, a selective angiopoietin inhibitor, in adult patients with advanced solid tumors. J Clin Oncol. 2009;27:3557–3565. doi: 10.1200/JCO.2008.19.6683. [DOI] [PubMed] [Google Scholar]
- 95.Karlan BY, Oza AM, Richardson GE, et al. Randomized, double-blind, placebo-controlled phase II study of AMG 386 combined with weekly paclitaxel in patients with recurrent ovarian cancer. J Clin Oncol. 2012;20:362–371. doi: 10.1200/JCO.2010.34.3178. [DOI] [PubMed] [Google Scholar]
- 96.Chang YM, Kung HJ, Evans CP. Nonreceptor tyrosine kinases in prostate cancer. Neoplasia. 2007;9:90–100. doi: 10.1593/neo.06694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Mayer EL, Krop IE. Advances in targeting SRC in the treatment of breast cancer and other solid malignancies. Clin Cancer Res. 2010;16:3526–3532. doi: 10.1158/1078-0432.CCR-09-1834. [DOI] [PubMed] [Google Scholar]
- 98.Bolós V, Gasent JM, López-Tarruella S, et al. The dual kinase complex FAK-Src as a promising therapeutic target in cancer. Onco Targets Ther. 2010;3:83–97. doi: 10.2147/ott.s6909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wheeler DL, Iida M, Dunn EF. The role of Src in solid tumors. Oncologist. 2009;14:667–678. doi: 10.1634/theoncologist.2009-0009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Johnson FM, Bekele BN, Feng L, et al. Phase II study of dasatinib in patients with advanced non–small-cell lung cancer. J Clin Oncol. 2010;28:4609–4615. doi: 10.1200/JCO.2010.30.5474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Lu C, Han HD, Mangala LS, et al. Regulation of tumor angiogenesis by EZH2. Cancer Cell. 2010;18:185–197. doi: 10.1016/j.ccr.2010.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Smits M, Nilsson J, Mir SE, et al. MiR-101 is down-regulated in glioblastoma resulting in EZH2-induced proliferation, migration, and angiogenesis. Oncotarget. 2010;1:710–720. doi: 10.18632/oncotarget.205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Kottakis F, Polytarchou C, Foltopoulou P, et al. FGF-2 regulates cell proliferation, migration, and angiogenesis through an NDY1/KDM2B-miR-101-EZH2 pathway. Mol Cell. 2011;43:285–298. doi: 10.1016/j.molcel.2011.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Lippert JW., 3rd Vascular disrupting agents. Bioorg Med Chem. 2007;15:605–615. doi: 10.1016/j.bmc.2006.10.020. [DOI] [PubMed] [Google Scholar]
- 105.Hori K, Saito S, Kubota K. A novel combretastatin A-4 derivative, AC7700, strongly stanches tumour blood flow and inhibits growth of tumours developing in various tissues and organs. Br J Cancer. 2002;86:1604–1614. doi: 10.1038/sj.bjc.6600296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Petrillo M, Borriello M, Fuoco G, et al. Novel VEGF-independent strategies targeting tumor vasculature: Clinical aspects. Curr Pharm Des. 2012;18:2702–2712. doi: 10.2174/138161212800626184. [DOI] [PubMed] [Google Scholar]
- 107.Taylor M, Billiot F, Marty V, et al. Reversing resistance to vascular-disrupting agents by blocking late mobilization of circulating endothelial progenitor cells. Cancer Discov. 2012;2:434–449. doi: 10.1158/2159-8290.CD-11-0171. [DOI] [PubMed] [Google Scholar]
- 108.Shaked Y, Emmenegger U, Man S, et al. Optimal biologic dose of metronomic chemotherapy regimens is associated with maximum antiangiogenic activity. Blood. 2005;106:3058–3061. doi: 10.1182/blood-2005-04-1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Kerbel RS, Kamen BA. The anti-angiogenic basis of metronomic chemotherapy. Nat Rev Cancer. 2004;4:423–436. doi: 10.1038/nrc1369. [DOI] [PubMed] [Google Scholar]
- 110.Gogas H, Dafni U, Karina M, et al. Postoperative dose-dense sequential versus concomitant administration of epirubicin and paclitaxel in patients with node-positive breast cancer: 5-year results of the Hellenic Cooperative Oncology Group HE 10/00 phase III trial. Breast Cancer Res Treat. 2012;132:609–619. doi: 10.1007/s10549-011-1913-4. [DOI] [PubMed] [Google Scholar]
- 111.Katsumata N, Yasuda M, Takahashi F, et al. Dose-dense paclitaxel once a week in combination with carboplatin every 3 weeks for advanced ovarian cancer: A phase 3, open-label, randomised controlled trial. Lancet. 2009;374:1331–1338. doi: 10.1016/S0140-6736(09)61157-0. [DOI] [PubMed] [Google Scholar]
- 112.Fujiwara K, Aotani E, Hamano T, et al. A randomized phase II/III trial of 3 weekly intraperitoneal versus intravenous carboplatin in combination with intravenous weekly dose-dense paclitaxel for newly diagnosed ovarian, fallopian tube and primary peritoneal cancer. Jpn J Clin Oncol. 2011;41:278–282. doi: 10.1093/jjco/hyq182. [DOI] [PubMed] [Google Scholar]
- 113.Kelly CM, Green MC, Broglio K, et al. Phase III trial evaluating weekly paclitaxel versus docetaxel in combination with capecitabine in operable breast cancer. J Clin Oncol. 2012;30:930–935. doi: 10.1200/JCO.2011.36.2079. [DOI] [PubMed] [Google Scholar]
- 114.Caporali A, Emanueli C. MicroRNA regulation in angiogenesis. Vascul Pharmacol. 2011;55:79–86. doi: 10.1016/j.vph.2011.06.006. [DOI] [PubMed] [Google Scholar]
- 115.Coussens LM, Raymond WW, Bergers G, et al. Inflammatory mast cells up-regulate angiogenesis during squamous epithelial carcinogenesis. Genes Dev. 1999;13:1382–1397. doi: 10.1101/gad.13.11.1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.de Visser KE, Korets LV, Coussens LM. De novo carcinogenesis promoted by chronic inflammation is B lymphocyte dependent. Cancer Cell. 2005;7:411–423. doi: 10.1016/j.ccr.2005.04.014. [DOI] [PubMed] [Google Scholar]
- 117.Lin EY, Li JF, Gnatovskiy L, et al. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res. 2006;66:11238–11246. doi: 10.1158/0008-5472.CAN-06-1278. [DOI] [PubMed] [Google Scholar]
- 118.Burnstock G. Purinergic signaling and vascular cell proliferation and death. Arterioscler Thromb Vasc Biol. 2002;22:364–373. doi: 10.1161/hq0302.105360. [DOI] [PubMed] [Google Scholar]
- 119.Anitua E, Andia I, Ardanza B, et al. Autologous platelets as a source of proteins for healing and tissue regeneration. Thromb Haemost. 2004;91:4–15. doi: 10.1160/TH03-07-0440. [DOI] [PubMed] [Google Scholar]
- 120.Lansdown AB. Calcium: A potential central regulator in wound healing in the skin. Wound Repair Regen. 2002;10:271–285. doi: 10.1046/j.1524-475x.2002.10502.x. [DOI] [PubMed] [Google Scholar]
- 121.Italiano JE, Jr, Richardson JL, Patel-Hett S, et al. Angiogenesis is regulated by a novel mechanism: Pro- and antiangiogenic proteins are organized into separate platelet alpha granules and differentially released. Blood. 2008;111:1227–1233. doi: 10.1182/blood-2007-09-113837. [DOI] [PMC free article] [PubMed] [Google Scholar]