Anti-VEGF pathway therapies primarily target immature blood vessels in tumors. However, emerging approaches to combine with targeted therapies impacting the later stages of remodeling and vessel maturation are expected to improve clinical efficacy by expanding the target vessel population. The angiopoietin/Tie ligand/receptor system is a prototypic regulator of vessel remodeling and maturation. Angiopoietin-2 (Ang2) appears to be a particularly attractive therapeutic target. In fact, the experimental proof-of-concept showing improved efficacy when VEGF and Ang2-targeting therapies are combined has been solidly established in preclinical models, and several Ang2-targeting drugs are in clinical trials. However, rational development of these second-generation combination therapies is hampered by a limited understanding of the biological complexity that is generated from agonistic and antagonistic Ang/Tie signaling. This review discusses recent mechanistic advances in angiopoietin signaling, particularly in light of the recent study published on REGN910 and summarizes the status quo of Ang2-targeting therapies. In light of the clarified partial agonist function of Ang2, we propose that clarity on the expression profile of the angiopoietin ligands and Tie1 and Tie2 receptors in subsets of cancer vessels and cancer cells will provide clearer hypotheses for more focused rational clinical trials to exploit this seminal pathway and improve current antiangiogenic therapies. Cancer Res; 73(6); 1649–57. ©2013 AACR.

In the last 9 years, antiangiogenic therapy has become part of standard antitumor treatment. However, the clinical efficacy of such therapies is limited, and it appears that the full therapeutic potential of antiangiogenic intervention has not been fully exploited. Moreover, the mechanistic goals of antiangiogenic intervention are presently less clear than ever. While concepts developed by Folkman in the 1970s suggested that antiangiogenic therapy was supposed to starve tumors to death by driving the tumor-associated vasculature into regression (1), we find that clinically established anti-VEGF/VEGF receptor therapies primarily promote stable disease by preferentially targeting the most immature blood vessels leaving behind normalized and resistant blood vessels (2). The enigmatic question around the importance of vascular regression, tumor vessel normalization, or a spatiotemporal combination of both mechanisms for the efficacy to antiangiogenic intervention has increased to the forefront of basic and applied vascular biology research to guide future pharmaceutical development. One of the most challenging questions at the moment is: What drug combinations improve the clinical efficacy of targeted VEGF pathway therapies without causing toxicity by damaging the mature, resting organ vasculature outside tumors?

The vascular receptor tyrosine kinase Tie2 is physiologically activated by its agonistic ligand Ang1. Ang1- and Tie2-deficient mice die in midgestation around embryonic day 10.5 as a consequence of perturbed vessel remodeling and maturation (3). The phenotypic similarity between Ang1 ligand and Tie2 receptor–deficient mice has supported the concept that Ang1 is the single, nonredundant agonistic ligand of Tie2 (4, 5). Using similar logic, the antagonistic mode of action of the second angiopoietin ligand, angiopoietin-2 (Ang2), was inferred from genetic manipulation experiments in mice: Ang2-overexpressing transgenic mice essentially phenocopy the midgestational embryonic lethal phenotype of Ang1-deficient mice (6). In other words, too much Ang2 resembles too little Ang1. In addition, correlative expression of Ang2 at times of vessel regression in ovaries, tumor vascular cooption, and hyaloid vessel regression supported the concept that the dominant biologic role of Ang2 is destabilization of established blood vessels through the interruption of Tie2 signaling, as a prerequisite to sprouting angiogenesis in the presence of proangiogenic stimulation or physiologic vascular regression in the absence of such stimuli (7–10). The molecular mechanism of this vessel destabilization phenotype has recently been bolstered to include the Ang2-induced internalization and degradation of endothelial cell surface integrins, which may eventually drive the cells into apoptosis (anoikis; ref. 11). The antagonistic mode of Ang2 function is also supported by the positional presentation of the angiopoietin ligands; whereas Ang1 is produced by periendothelial cells, including pericytes, to act in a paracrine manner, Ang2 is almost exclusively produced by endothelial cells and acts on endothelial cells in an autocrine manner, possibly even involving intracrine signaling mechanisms (12). In fact, Ang2 is not just produced by endothelial cells, but endothelial cells store it in Weibel–Palate bodies where it can be rapidly released in seconds to minutes to enable the responsiveness of endothelial cells to angiogenic, inflammatory, and other cytokines (13, 14). Tools to examine the impact of Ang2 on Tie2 phosphorylation are poor and have hampered extensive interrogation of this hypothesis; however, there is one study in which transgenic overexpression of Ang2 in endothelial cells was shown to be capable of reducing Tie2 phosphorylation in vivo (15). Ang2 production by endothelial cells is strongly regulated at the transcriptional level. In fact, almost any form of endothelial cell activation leads to upregulation of Ang2 mRNA. The mRNA induction of Ang2 in tumor endothelium has made Ang2 a very attractive circulating biomarker of angiogenic activation (e.g., ref. 16; summary in ref. 3). Thus, while the in vivo evidence for a predominately antagonistic mode of action of Ang2 is overwhelming, there is still only very limited examination of the molecular mechanisms of this phenotype.

As a challenge to the antagonist hypothesis, a number of cellular studies have provided compelling evidence that Ang2 may under certain conditions also be able to act agonistically on its receptor Tie2 (e.g., ref. 17). The typically higher concentrations of Ang2 required to observe this agonism have raised questions around the physiologic relevance of the in vitro data. However, there are a series of in vivo data suggesting that there are cellular contexts in which Ang2 may offer a proangiogenic function through agonism of the Tie2 receptor. One example is the situation in lymphatics, in which deletion of Ang2 led to lymphatic dysfunction with chylous ascites and edema. This lymphatic phenotype was corrected by replacing endogenous Ang2 with a cDNA for Ang1 (18). Thus, Ang2 seemed to substitute for Ang1-agonistic functions in lymphatics in vivo, which may be deprived of Ang1 signaling due to their low pericyte coverage. Similarly, in monocytes there is evidence of agonist activity by Ang2. A subpopulation of Tie2-expressing monocytes (TEM) are associated with tumor blood vessels and exhibit proangiogenic activity, which could be inhibited by Tie2 silencing or Ang2 neutralization (19, 20). In vitro, TEMs migrated toward Ang2, which induced Tie2 phosphorylation (21). Thus, Ang2 also functioned as an agonist of Tie2 in these cells.

To further complicate the question of Tie2 antagonism versus agonism, there is a series of reports that incorporate the orphan receptor Tie1 into the context of Ang2 function. Deletion of Tie1 in mice results in defects of vascular integrity, localized hemorrhaging, and cardiac underdevelopment, indicating its essential role in vascular maintenance (22). Tie1 is highly expressed by arteries and their associated capillaries at branching points and is induced by disturbed flow, suggesting dynamic physiologic regulation of Tie1 (23). Multiple studies have addressed potential cross-talk between Tie1 and Tie2, as well as feedback regulatory loops that respond differently to Ang1 and Ang2 (24, 25). These data are still emerging but a take-home hypothesis generated by Seegar and colleagues suggests that in the presence of Tie1, Ang2 is unable to activate Tie2; however, loss of Tie1 reveals the agonist capabilities of Ang2 in vitro (26). These tantalizing data await a more physiologic context and confirmation in vivo.

Ang2 has recently been shown to exert proangiogenic functions in a Tie2-independent manner. Angiogenic endothelial tip cells downregulate the Tie2 receptor and Ang2 signals in these cells through activated integrins to promote focal adhesion kinase (FAK) signaling and, subsequently, angiogenic sprouting (27). While not acting through an agonism of Tie2, this is nevertheless an unexpected proangiogenic function of Ang2 where neutralization of the ligand could be beneficial in a cancer context. It is notable that in terms of Ang2 functions, both its role in destabilizing a mature vasculature to facilitate cooption and neoangiogenesis and its role in promoting integrin stimulation of sprouting tip cells suggest that the consequences of Ang2 blockade will likely be contextual with different net outcomes on sprouting Tie2-negative tip cells and more mature Tie2-expressing stalk and phalanx endothelial cells (see Fig. 1). This is consistent with published reports using neutralizing molecules to Ang2 where tumor growth and retinal sprouting are blocked (e.g., ref. 28). These studies use antibodies that were developed to block Tie2 interactions and often to block the in vitro activation of Tie2 that can be observed in the absence of Ang1, suggesting that the antibodies or peptides block the Ang2–Tie2–interacting domain. It is unclear whether the observed Ang2 interactions with integrins use the same binding domains and whether this function is sensitive to the currently available blocking antibodies and peptides to Ang2 in therapeutic development described below.

Figure 1.

Endothelial cell phenotypes during tumor angiogenesis. A, endothelial cells during angiogenesis maintain distinct functions and acquire specific phenotypes during the individual steps of the angiogenic cascade. Sprouting tip cells (1) have an invasive filopodia-rich promigratory phenotype. They sense the gradient of angiogenic cytokines and migrate towards the angiogenic stimuli. Tip cells are followed by stalk cells (2), which proliferate and remodel to establish lumen-forming microvessels. Tip cell– and stalk cell–derived PDGFB is the best characterized pericyte-recruiting cytokine during angiogenesis. Pericyte recruitment leads to microvessel maturation. Loosely attached pericytes cover remodeling phalanx cells (3). Fully mature phalanx endothelial cells (4) are in intimate contact with pericytes, which control the quiescent endothelial cell phenotype. B, application of the tip cell/stalk cell/phalanx cell model of angiogenesis applied to the partial Ang2 agonist concept for cancer: The mature organ vasculature outside tumors (4) expresses Tie2, and the cells are surrounded by Ang1-producing pericytes. There is low-level Ang2 production, which is stored in quiescent phalanx endothelial cells, that can be induced and secreted upon activation. Release of Ang2 would likely result in a quenching of Ang1/Tie2 signal transduction indicative of an antagonistic mode of action. Conversely, the most immature tip cell vasculature (1) downregulates Tie2 and upregulates Ang2 creating a milieu in which Ang2 can signal agonistically through integrins. The exact positional information on Ang ligands and Tie receptors in stalk cells and in remodeling phalanx cells awaits further analysis, but it is most likely that Tie2- and Ang2-expressing endothelial cells in a low-Ang1 environment could be found in this zone (2, 3) where the partial agonist Ang2 results in a net activation for Tie2. The function and the positional expression of the Ang/Tie2 response, including the role of Tie1 in this context, similarly await further clarification.

Figure 1.

Endothelial cell phenotypes during tumor angiogenesis. A, endothelial cells during angiogenesis maintain distinct functions and acquire specific phenotypes during the individual steps of the angiogenic cascade. Sprouting tip cells (1) have an invasive filopodia-rich promigratory phenotype. They sense the gradient of angiogenic cytokines and migrate towards the angiogenic stimuli. Tip cells are followed by stalk cells (2), which proliferate and remodel to establish lumen-forming microvessels. Tip cell– and stalk cell–derived PDGFB is the best characterized pericyte-recruiting cytokine during angiogenesis. Pericyte recruitment leads to microvessel maturation. Loosely attached pericytes cover remodeling phalanx cells (3). Fully mature phalanx endothelial cells (4) are in intimate contact with pericytes, which control the quiescent endothelial cell phenotype. B, application of the tip cell/stalk cell/phalanx cell model of angiogenesis applied to the partial Ang2 agonist concept for cancer: The mature organ vasculature outside tumors (4) expresses Tie2, and the cells are surrounded by Ang1-producing pericytes. There is low-level Ang2 production, which is stored in quiescent phalanx endothelial cells, that can be induced and secreted upon activation. Release of Ang2 would likely result in a quenching of Ang1/Tie2 signal transduction indicative of an antagonistic mode of action. Conversely, the most immature tip cell vasculature (1) downregulates Tie2 and upregulates Ang2 creating a milieu in which Ang2 can signal agonistically through integrins. The exact positional information on Ang ligands and Tie receptors in stalk cells and in remodeling phalanx cells awaits further analysis, but it is most likely that Tie2- and Ang2-expressing endothelial cells in a low-Ang1 environment could be found in this zone (2, 3) where the partial agonist Ang2 results in a net activation for Tie2. The function and the positional expression of the Ang/Tie2 response, including the role of Tie1 in this context, similarly await further clarification.

Close modal

In a recent issue of Cancer Research, Daly and colleagues reveal insights into the complexity of Ang/Tie signaling that provides a model for the contextuality of Ang2-antagonistic and -agonistic functions of Ang2 on Tie2 (Fig. 1). The authors show that a selective antibody to Ang2 (REGN910) can decrease tumor growth in a manner that is reversed by high-dose administration of an engineered form of Ang1 called Ang1-F1-Fc-F1 (which has an IC50 of 9.7 nmol/L to Tie2 and is a forced tetramer), hinting that Tie2 signaling was attenuated (29, 30). This Ang1 construct has previously been shown to maximally activate Tie2 in vitro and in vivo and to block mustard oil–induced vascular leak. While this report falls short of directly showing the impact of REGN910 on activation of Tie2 by Ang2 in vivo, the tumor growth and resulting gene expression data presented, along with the above described cellular contexts in which Ang2 can have proangiogenic and possibly even Tie2-agonistic activity, provide a compelling case to revisit the simplistic view of the Yin-Yang of Ang1 and Ang2.

The data presented in the article by Daly and colleagues are compatible with a partial agonist model of Ang2 function. In line with classical pharmacologic concepts, the partial agonist competes with the strong agonist. In the presence of the strong agonist (in this case, Ang1), the partial agonist Ang2 quenches the signal induced by the strong agonist, thereby exerting an inhibitory activity. In the absence of the strong agonist, the weak agonist may function as receptor-stimulating ligand. This model of Ang2 as a partial agonist can perhaps provide a unifying hypothesis to explain some of the contextuality of Ang/Tie biology. For example, the embryonic lethal phenotype of Ang1-deficient mice may be explained by the fact that Ang2 is only later expressed in endothelial cells during embryonic development. Conversely, the puzzling observation that Ang1 deletion in adult mice did not result in overt pathology unless the mice were pathologically challenged (31) may well be attributed by a compensatory agonistic function elicited by the partial antagonist Ang2, which would be a testable hypothesis. Clearly, the model proposed by Daly and colleagues provokes a long list of experiments on the contextuality of Ang/Tie function, which emerges at a time when better reagents for the tracing of receptor and ligand expression and for the conditional mutagenesis of the ligands and receptors becomes more readily available. An accelerated pace in the elucidation of the molecular mechanisms of Ang/Tie signaling in the tumor context holds great promise to enable the rational translation of Ang/Tie pathway therapeutics into clinical application.

There are no selective Tie2 kinase inhibitors in clinical development; however, several approved agents show cellular activity against this pathway. Regorafenib is one of several small-molecule VEGFR2 inhibitors that also exhibit some activity against Tie2. Regorafenib has a 3 to 10 nmol/L cellular activity against VEGFR2, Ret, Kit, and Raf and modest (30–150 nmol/L) cellular activity against fibroblast growth factor receptor (FGFR), platelet-derived growth factor receptor (PDGFR), and Tie2. Phase III trials showed a significant increase in progression-free survival (PFS) and overall survival in second-line metastatic colorectal cancer (CRC) leading to the accelerated approval of the drug (32). Cabozopanib, a related VEGFR TKIs that is also active against Ret, Tie2, and other kinases, is now approved in medullary thyroid cancer (33). Similarly, MGCD-265 is a Ron, Tie2, cMET, and a VEGFR1/2/3 inhibitor that is being currently tested in phase II trials in patients with a variety of advanced metastatic malignancies (34). In addition, there are several small-molecule inhibitors (ACTB-1003, CEP-11981, ARRY-614) in phase I trials. As it is unlikely that any of these molecules has dominant impact via Tie2 signaling, the remainder of this discussion will focus on the variety of more selective biologics currently under development.

There are 2 types of biologics targeting the angiopoietins whose references can be found in Table 1. The first and most advanced are peptide based, using peptides with high affinity and differing selectivity to Ang1 and Ang2. These peptides are either linked to an Fc domain or to an irrelevant antibody. Trebananib (AMG-386) and PF04856884 (CVX-060) are the 2 peptide-based traps that are most advanced, in phase III and 2 trials, respectively. Trebananib is a peptide–Fc fusion protein (peptibody) that blocks the interaction of both Ang1 and Ang2 to Tie2 with an affinity of 0.67 nmol/L by BiAcore (35). Several phase II trials were conducted with trebananib in which some hints of clinical efficacy and experience with pharmacokinetics (PK)/pharmacodynamics (PD) has led to further development in phase III, particularly in ovarian cancer. Phase II data showed an improved response rate but no difference in PFS when trebananib was combined with sorafenib in patients with metastatic clear cell renal carcinoma (RCC; ref. 36). However, an encouraging dose–response effect was observed when trebananib was used in combination with paclitaxel in patients with advanced recurrent epithelial ovarian or primary peritoneal cancer (37). PK/PD analysis of the phase II data suggest that maximum benefit in PFS may not have been reached with a trebananib dose of 10 mg/kg once every week and predict that a dose of 15 mg/kg once every week may provide greater exposure and may yield further improvements in PFS (38). The 3 phase III trials now ongoing at 15 mg/kg once-a-week dose of trebananib combine either paclitaxel (NCT01204749/TRINOVA-1), pegylated liposomal doxorubicin (NCT01281254/TRINOVA-2), or a combination with paclitaxel and carboplatin (NCT01493505/TRINOVA-3). TRINOVA-1 and -2 are in recurrent platinum-sensitive or -resistant epithelial ovarian, primary peritoneal, or fallopian tube cancers, whereas TRINOVA-3 is in first-line FIGO Stage III-IV epithelial ovarian, primary peritoneal, or fallopian tube cancers (39).

Table 1.

Therapeutics targeting Ang-Tie axis

CompanyTargetHighest phase of developmentClinical dataPreclinical data
Small-molecule inhibitors 
 Regorafenib Bayer AG FGFR, Kit, Tie2, VEGFR1,2,3, RET, BRAF, CRAF, PDGFR and caspase-3/9 stimulator Phase III Positive phase III trials (significant increase in PFS) in second-line metastatic CRC and third-line gastrointestinal stromal tumors (46) Showed inhibition of tumor growth in several tumor xenograft models including pancreatic, lung, colon carcinoma and regression in breast carcinoma model (47). In addition showed a significant increase in survival with fewer metastasis in syngeneic MC38 CRC liver metastasis model (48). 
 Cabozantinib (XL-184/BMS-907351) Exelixis MET, VEGFR2, RET, Kit, FLT3, Tie2 Phase III Positive phase III trials in medullary thyroid cancer and ongoing phase III trials in metastatic castration-resistant prostate cancer. Reduced metastasis in RIP-TAG2 model of pancreatic neuroendocrine carcinoma compared with VEGF inhibitor alone (49). 
 MGCD-265 MethylGene, Inc. RON, Tie2, VEGFR1, 2, 3, cMET Phase II A parallel-arm, open-label trial in advanced metastatic malignancies and NSCLC began for assessing antitumor activity, PFS, and safety (50). Showed tumor regression in MKN45 gastric cancer xenograft model in combination either with erlotinib or docetaxel. Showed antitumor activity and tumor growth regression in MET-driven human xenograft cells (gastric carcinoma) and non–MET-driven tumor cells (NSCLC and prostate) in combination with paclitaxel and docetaxel (34). 
 Foretinib Exelixis, Inc./GSK MET, VEGFR1, 2, Tie2, RON, AXL, FLT3 and PDGFR-beta Phase 2 A phase1/2 trial in HCC and phase 2 trials in recurrent or metastatic squamous cell head and neck cancer, metastatic gastric cancer and hereditary or sporadic papillary RCC (NCT00725764, NCT00743067, NCT00920192). Dose-dependently inhibited tumor growth in several models including glioblastoma, breast, CRC, and NSCLC (51). 
 ACTB-1003 ACT Biotech/Bayer AG VEGFR2, Tie2, RSK, FGFR1 and S6K Phase I None. Showed tumor growth inhibition in cell lines with FGFR genetic alterations—OPM2 human multiple myeloma and the murine leukemia Ba/F3-TEL-FGFR1 and in H460 (52). 
 CEP-11981 Cephalon, Inc./Teva Tie2, VEGFR1,2,3, FGFR1 Phase I Linear, dose-related increases in plasma pharmacokinetic exposure (mean Cmax and AUC) with oral administration. Exposure-related increases in VEGFR2 inhibition ex vivo were shown in a human plasma-based cellular Dose-dependent tumor growth inhibition in U251 MG human glioblastoma xenograft and A375 human melanoma xenograft model (53). 
    Bioassay (NCT00875264).  
 ARRY-614 Array BioPharma, Inc. p38, Tie2 Phase I Drug was well tolerated and the MTD was not reached with QD dosing. Recently tested a new formulation with enhanced drug exposure and lower variability for future trials (54). Antitumor activity shown in multiple myeloma in combination with lenalidomide. Complete regressions in BCR-Abl–driven model of chronic myeloid leukemia (55). 
Large-molecule inhibitors 
 Trebananib- peptide: Fc fusion-peptibody, AMG-386 Amgen Ang1 and Ang2 Phase III No improvement in PFS in combination with sorafenib in metastatic RCC (phase II; ref. 36). Significantly inhibited tumor growth of several tumor types, including Colo205 (CRC) and A431 (epidermoid carcinoma). 
    Evidence of antitumor activity and a dose-response effect in combination with paclitaxel in advanced recurrent epithelial ovarian or primary peritoneal cancer (37). A dose-dependent antiangiogenic effect in VEGF-mediated rat corneal angiogenesis assay. Had greater tumor growth suppression than inhibiting Ang1 or Ang2 alone (57). 
    No apparent prolongation of PFS in combination with paclitaxel in HER2-negative, metastatic, or locally recurrent breast cancer (56).  
 CVX-060/PF04856884, recombinant humanized CovX body CovX Pharmaceuticals (Pfizer) Ang2 Phase II A randomized, crossover, open-label phase II trial in metastatic RCC in combination with axitinib was initiated (NCT01441414). As efficacious as bevacizumab in a colon cancer xenograft model and found to improve the response to irinotecan or docetaxel when administered together (35). 
 AMG-780, fully human anti- ang1/2 mAb Amgen Inhibits Tie2-Ang1/2 interaction Phase I Dose-escalation and expansion in patients with advanced solid malignancies (NCT01137552). None. 
 MEDI-3617, fully human anti-Ang 2 mAb(3.19.3) AstraZeneca Ang2 (Ang1 weakly) Phase I Dose-escalation in patients with advanced solid malignancies. Modest toxicity was observed. No objective response. Five of 17 evaluated patients had stable disease at 12 wks (58). Inhibited tumor growth in mice with human pancreatic and hepatocellular xenografts 
     Efficacy enhanced in combination with sorafenib and bevacizumab 
     Efficacy also enhanced in human CRC models in combination with vandetanib, cediranib, or DC101 (59). 
 REGN-910, fully human anti-Ang2 mAb Sanofi/Regeneron Ang2 Phase I Dose-escalation and expansion study in patients with advanced solid malignancies (60). Significantly inhibited growth of several tumors types including PC3 (prostate), Colo205 (CRC), and A431 (epidermois carcinoma). 
     Reduced Colo205 tumor vascularity and tumor perfusion more than single agents, as per histologic and microultrasound analyses (61). 
     Potentiates the effects of VEGF-Trap (Aflibercept; ref. 62). 
 TAvi6, a CrossMab tetravalent IgG-like bispecific antibody Roche Ang2 and VEGF-A Discovery None. Effectively reduced angiogenesis, tumor growth, and metastasis in several subcutaneous and orthotopic in vivo models that are resistant to anti-VEGF treatment. Ang2-selective approach was found to have safety-related advantages over simultaneous Ang1 and Ang2 inhibition (63). 
 AT-006, fully human mab Roche/Affitech Ang2 Discovery None. Inhibited tumor growth and neoangiogenesis (64). 
 Anti-Her2/anti-Ang2 mab, multivalent mab Zyngenia Her2 and Ang2 Discovery None. Remained intact and functional in mouse serum and can bind to Her2 and Ang2 simultaneously (65). 
 A-11, a peptide inhibitor of Ang2 and Tie2 interaction Compugen Ang2 Discovery None. Inhibited retinal neovascularization in a rat model of oxygen-induced retinopathy. Showed synergy with anti-VEGF antibody (66). 
CompanyTargetHighest phase of developmentClinical dataPreclinical data
Small-molecule inhibitors 
 Regorafenib Bayer AG FGFR, Kit, Tie2, VEGFR1,2,3, RET, BRAF, CRAF, PDGFR and caspase-3/9 stimulator Phase III Positive phase III trials (significant increase in PFS) in second-line metastatic CRC and third-line gastrointestinal stromal tumors (46) Showed inhibition of tumor growth in several tumor xenograft models including pancreatic, lung, colon carcinoma and regression in breast carcinoma model (47). In addition showed a significant increase in survival with fewer metastasis in syngeneic MC38 CRC liver metastasis model (48). 
 Cabozantinib (XL-184/BMS-907351) Exelixis MET, VEGFR2, RET, Kit, FLT3, Tie2 Phase III Positive phase III trials in medullary thyroid cancer and ongoing phase III trials in metastatic castration-resistant prostate cancer. Reduced metastasis in RIP-TAG2 model of pancreatic neuroendocrine carcinoma compared with VEGF inhibitor alone (49). 
 MGCD-265 MethylGene, Inc. RON, Tie2, VEGFR1, 2, 3, cMET Phase II A parallel-arm, open-label trial in advanced metastatic malignancies and NSCLC began for assessing antitumor activity, PFS, and safety (50). Showed tumor regression in MKN45 gastric cancer xenograft model in combination either with erlotinib or docetaxel. Showed antitumor activity and tumor growth regression in MET-driven human xenograft cells (gastric carcinoma) and non–MET-driven tumor cells (NSCLC and prostate) in combination with paclitaxel and docetaxel (34). 
 Foretinib Exelixis, Inc./GSK MET, VEGFR1, 2, Tie2, RON, AXL, FLT3 and PDGFR-beta Phase 2 A phase1/2 trial in HCC and phase 2 trials in recurrent or metastatic squamous cell head and neck cancer, metastatic gastric cancer and hereditary or sporadic papillary RCC (NCT00725764, NCT00743067, NCT00920192). Dose-dependently inhibited tumor growth in several models including glioblastoma, breast, CRC, and NSCLC (51). 
 ACTB-1003 ACT Biotech/Bayer AG VEGFR2, Tie2, RSK, FGFR1 and S6K Phase I None. Showed tumor growth inhibition in cell lines with FGFR genetic alterations—OPM2 human multiple myeloma and the murine leukemia Ba/F3-TEL-FGFR1 and in H460 (52). 
 CEP-11981 Cephalon, Inc./Teva Tie2, VEGFR1,2,3, FGFR1 Phase I Linear, dose-related increases in plasma pharmacokinetic exposure (mean Cmax and AUC) with oral administration. Exposure-related increases in VEGFR2 inhibition ex vivo were shown in a human plasma-based cellular Dose-dependent tumor growth inhibition in U251 MG human glioblastoma xenograft and A375 human melanoma xenograft model (53). 
    Bioassay (NCT00875264).  
 ARRY-614 Array BioPharma, Inc. p38, Tie2 Phase I Drug was well tolerated and the MTD was not reached with QD dosing. Recently tested a new formulation with enhanced drug exposure and lower variability for future trials (54). Antitumor activity shown in multiple myeloma in combination with lenalidomide. Complete regressions in BCR-Abl–driven model of chronic myeloid leukemia (55). 
Large-molecule inhibitors 
 Trebananib- peptide: Fc fusion-peptibody, AMG-386 Amgen Ang1 and Ang2 Phase III No improvement in PFS in combination with sorafenib in metastatic RCC (phase II; ref. 36). Significantly inhibited tumor growth of several tumor types, including Colo205 (CRC) and A431 (epidermoid carcinoma). 
    Evidence of antitumor activity and a dose-response effect in combination with paclitaxel in advanced recurrent epithelial ovarian or primary peritoneal cancer (37). A dose-dependent antiangiogenic effect in VEGF-mediated rat corneal angiogenesis assay. Had greater tumor growth suppression than inhibiting Ang1 or Ang2 alone (57). 
    No apparent prolongation of PFS in combination with paclitaxel in HER2-negative, metastatic, or locally recurrent breast cancer (56).  
 CVX-060/PF04856884, recombinant humanized CovX body CovX Pharmaceuticals (Pfizer) Ang2 Phase II A randomized, crossover, open-label phase II trial in metastatic RCC in combination with axitinib was initiated (NCT01441414). As efficacious as bevacizumab in a colon cancer xenograft model and found to improve the response to irinotecan or docetaxel when administered together (35). 
 AMG-780, fully human anti- ang1/2 mAb Amgen Inhibits Tie2-Ang1/2 interaction Phase I Dose-escalation and expansion in patients with advanced solid malignancies (NCT01137552). None. 
 MEDI-3617, fully human anti-Ang 2 mAb(3.19.3) AstraZeneca Ang2 (Ang1 weakly) Phase I Dose-escalation in patients with advanced solid malignancies. Modest toxicity was observed. No objective response. Five of 17 evaluated patients had stable disease at 12 wks (58). Inhibited tumor growth in mice with human pancreatic and hepatocellular xenografts 
     Efficacy enhanced in combination with sorafenib and bevacizumab 
     Efficacy also enhanced in human CRC models in combination with vandetanib, cediranib, or DC101 (59). 
 REGN-910, fully human anti-Ang2 mAb Sanofi/Regeneron Ang2 Phase I Dose-escalation and expansion study in patients with advanced solid malignancies (60). Significantly inhibited growth of several tumors types including PC3 (prostate), Colo205 (CRC), and A431 (epidermois carcinoma). 
     Reduced Colo205 tumor vascularity and tumor perfusion more than single agents, as per histologic and microultrasound analyses (61). 
     Potentiates the effects of VEGF-Trap (Aflibercept; ref. 62). 
 TAvi6, a CrossMab tetravalent IgG-like bispecific antibody Roche Ang2 and VEGF-A Discovery None. Effectively reduced angiogenesis, tumor growth, and metastasis in several subcutaneous and orthotopic in vivo models that are resistant to anti-VEGF treatment. Ang2-selective approach was found to have safety-related advantages over simultaneous Ang1 and Ang2 inhibition (63). 
 AT-006, fully human mab Roche/Affitech Ang2 Discovery None. Inhibited tumor growth and neoangiogenesis (64). 
 Anti-Her2/anti-Ang2 mab, multivalent mab Zyngenia Her2 and Ang2 Discovery None. Remained intact and functional in mouse serum and can bind to Her2 and Ang2 simultaneously (65). 
 A-11, a peptide inhibitor of Ang2 and Tie2 interaction Compugen Ang2 Discovery None. Inhibited retinal neovascularization in a rat model of oxygen-induced retinopathy. Showed synergy with anti-VEGF antibody (66). 

Abbreviations: NSCLC, non–small cell lung cancer; qd, once daily.

PF04856884 (CVX-060) is a CovX-body specific to Ang2. CovX-bodies are an antibody platform that are generated by linking an irrelevant (aldolase) IgG to a target-binding peptide. CVX-060 has a binding affinity of 4.9 nmol/L to Ang2, similar to published affinity for Ang2 to Tie2. The peptide is linked using a branched azetidinone (AZD). The antibody scaffold imparts IgG-like half-life, and distribution and the peptide pharmacophores are responsible for functional activities (6, 40). CVX-060 inhibited tumor growth, tumor microvessel density, and intratumor proangiogenic Tie2/CD11b-positive cells (TEMs) in Colo205 xenograft tumors. Even greater efficacy (>80%) was observed when combined with sunitinib, sorafenib, bevacizumab, irinotecan, or docetaxel in this model (35). Significant mean decreases in tumor blood flow assessed by Ktrans (DCE-MRI) and increase in serum Ang2 levels were observed in a phase I study of CVX-060. The maximum tolerated dose (MTD) was not defined and the most common adverse event was fatigue. PK/PD analysis has recommended 15 mg/kg once weekly dosing (41). It is unclear whether increased serum Ang2 levels in this study are due to peptibody binding and accumulation of stabilized complexes in the circulation as opposed to a pharmacodynamics induction of de novo Ang2 expression. A phase II study is ongoing in combination with axitinib in patients with previously treated metastatic RCC (NCT01441414; ref. 39). In general, peptides tend to have lower affinities for Ang2 binding than antibodies, and understanding how competitive they are as neutralizing agents clinically awaits further development of the antibodies described below.

The second type of biologic therapeutics are antibodies. Two selective anti-Ang2 antibodies are in development, MEDI-3617 (AstraZeneca) and REGN-910 (Sanofi). Both MEDI-3617 and REGN-910 have ongoing phase I dose-escalation studies as single agents or in combination with other chemotherapy in patients with advanced solid tumors (NCT01248949 and NCT01271972). MEDI-3617 reported drug-related adverse events (% of patients) including fatigue (24%), diarrhea (19%), nausea (19%), dysgeusia (14%), and headache (14%), all of which were ≤grade II (42). AMG-780, an antibody that binds both Ang1 and Ang2, is also in phase I development (NCT01137552) (39). In addition, Roche has a preclinical stage CrossMab, which is a bispecific antibody that binds and neutralizes both Ang2 and VEGF-A. This bifunctional approach is notable, as several preclinical studies have shown an advantage to dual inhibition of both VEGF/VEGFR and Ang/Tie2 pathways, including the study by Daly and colleagues, in a recent issue of Cancer Research (29, 42).

The prominent upregulation of Ang2 in the vasculature of most solid tumors makes Ang2 an attractive candidate for therapeutic intervention. Preclinical and clinical studies suggest that Ang2 may augment the activity of VEGF-targeted therapies without clear evidence of enhanced toxicity. However, the pleiotropic activities of Ang2 in blood, lymphatic, and myeloid biology, as well as complexities in receptor use and contextual presentation demand further investigation to clarify the critical aspects of Ang2 that are drivers in cancer progression. Regardless of which effects turn out to be dominant in cancer, current evidence supports the therapeutic rationale to antagonize Ang2.

In tumor angiogenesis, Ang2 appears to function as a potent Tie2 agonist based on the ability of Ang1 to counter the efficacy of anti-Ang2 antibodies. However, modulation of Tie2 activation in tumor vessels remains to be explored (29). Ectopic administration of Ang1 can substitute for Ang2 function, raising the question of the difference between autocrine Ang2 (released by endothelial cells subjected to stress) or paracrine Ang2. This may be particularly relevant in some tumor settings where Ang2 is produced by tumor cells. Ang2 has also been associated with tumor metastases. High Ang2 expression in primary human breast cancer specimens correlates with an increased metastatic phenotype (44). Furthermore, overexpression of Ang2 in MCF-7 promoted their metastasis to lymph nodes and lungs, an effect postulated to result from direct interaction between Ang2 and beta1 integrins on tumor cells (much the way Ang2 was reported to facilitate sprouting of Tie2-negative tip cells; refs. 11, 27). The delineation of autocrine versus paracrine presentation of Ang2 still needs to be carefully investigated.

The partial agonist concept developed by Daly and colleagues raises important questions as to when and where angiopoietin ligands and Tie2 receptor are expressed in different tumor settings (Fig. 1; ref. 1). Endothelial cells in mature tumor-associated vessels express Tie2 in an Ang1-rich/Ang2-poor microenvironment. When this balance is disturbed by increased expression of Ang2 (a weak agonist), the net result is a paradoxical decrease in Tie2 phosphorylation. Conversely, endothelial cells in the most immature tumor vasculature downregulate Tie2 expression, while at the same time upregulating Ang2 and favoring proangiogenic Ang2 function through integrin signaling. The contextual presentation of Ang1 in tumors is not well-defined, but an intermediate zone of Tie2- and Ang2-expressing endothelial cells in an Ang1-low microenvironment could likely be present in the actively proliferating stalk cell zone (Fig. 1).

Extrapolating from rapidly growing mouse tumors with a very immature vasculature, it is widely believed that the tumor vasculature is low in Ang1-producing pericytes. However, this is most likely an overstatement as microvessels in human tumors are often well-invested by pericytes. These pericytes may not be as tightly associated with endothelial cells as in resting blood vessels but are widely detected in human tumors (45). In fact, there are distinct differences in the pericyte coverage of different human tumor blood vessels. It is tempting, therefore, to speculate that pericyte-mediated differences in tumor blood vessel maturation could account for differences in the efficacy of immature blood vessels targeted by anti-VEGF/VEGFR therapies. These concepts strongly support a rationale for inducing vascular maturation as part of the treatment of tumors.

The development of Ang/Tie pathway drugs and the deciphering of the intricate details of Ang/Tie signaling occur at a time when the goals of antiangiogenic intervention are under serious consideration. Understanding the cross-talk between vessel maturation and remodeling (i.e., Ang/Tie) and primarily sprouting angiogenesis-inducing pathways (i.e., VEGF) holds great promise to rationally develop safe and improved antiangiogenic cancer therapy.

No potential conflicts of interest were disclosed.

Conception and design: D. Gerald, S. Chintharlapalli, H.G. Augustin, L.E. Benjamin

Writing, review, and/or revision of the manuscript: D. Gerald, S. Chintharlapalli, H.G. Augustin, L.E. Benjamin

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): D. Gerald, L.E. Benjamin

The authors thank Greg Plowman for edits and helpful suggestions.

1.
Folkman
J
. 
Tumor angiogenesis: therapeutic implications
.
N Engl J Med
1971
;
285
:
1182
6
.
2.
Carmeliet
P
,
Jain
RK
. 
Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases
.
Nat Rev Drug Discov
2011
;
10
:
417
27
.
3.
Augustin
HG
,
Koh
GY
,
Thurston
G
,
Alitalo
K
. 
Control of vascular morphogenesis and homeostasis through the angiopoietin-Tie system
.
Nat Rev Mol Cell Biol
2009
;
10
:
165
77
.
4.
Dumont
DJ
,
Gradwohl
G
,
Fong
GH
,
Puri
MC
,
Gertsenstein
M
,
Auerbach
A
, et al
Dominant-negative and targeted null mutations in the endothelial receptor tyrosine kinase, tek, reveal a critical role in vasculogenesis of the embryo
.
Genes Dev
1994
;
8
:
1897
909
.
5.
Suri
C
,
Jones
PF
,
Patan
S
,
Bartunkova
S
,
Maisonpierre
PC
,
Davis
S
, et al
Requisite role of angiopoietin-1, a ligand for the TIE2 receptor, during embryonic angiogenesis
.
Cell
1996
;
87
:
1171
80
.
6.
Maisonpierre
PC
,
Suri
C
,
Jones
PF
,
Bartunkova
S
,
Wiegand
SJ
,
Radziejewski
C
, et al
Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis
.
Science
1997
;
277
:
55
60
.
7.
Goede
V
,
Schmidt
T
,
Kimmina
S
,
Kozian
D
,
Augustin
HG
. 
Analysis of blood vessel maturation processes during cyclic ovarian angiogenesis
.
Lab Invest
1998
;
78
:
1385
94
.
8.
Hanahan
D
. 
Signaling vascular morphogenesis and maintenance
.
Science
1997
;
277
:
48
50
.
9.
Holash
J
,
Maisonpierre
PC
,
Compton
D
,
Boland
P
,
Alexander
CR
,
Zagzag
D
, et al
Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF
.
Science
1999
;
284
:
1994
8
.
10.
Rao
S
,
Lobov
IB
,
Vallance
JE
,
Tsujikawa
K
,
Shiojima
I
,
Akunuru
S
, et al
Obligatory participation of macrophages in an angiopoietin 2-mediated cell death switch
.
Development
2007
;
134
:
4449
58
.
11.
Thomas
M
,
Felcht
M
,
Kruse
K
,
Kretschmer
S
,
Deppermann
C
,
Biesdorf
A
, et al
Angiopoietin-2 stimulation of endothelial cells induces alphavbeta3 integrin internalization and degradation
.
J Biol Chem
2010
;
285
:
23842
9
.
12.
Scharpfenecker
M
,
Fiedler
U
,
Reiss
Y
,
Augustin
HG
. 
The Tie-2 ligand angiopoietin-2 destabilizes quiescent endothelium through an internal autocrine loop mechanism
.
J Cell Sci
2005
;
118
:
771
80
.
13.
Fiedler
U
,
Reiss
Y
,
Scharpfenecker
M
,
Grunow
V
,
Koidl
S
,
Thurston
G
, et al
Angiopoietin-2 sensitizes endothelial cells to TNF-alpha and has a crucial role in the induction of inflammation
.
Nat Med
2006
;
12
:
235
9
.
14.
Fiedler
U
,
Scharpfenecker
M
,
Koidl
S
,
Hegen
A
,
Grunow
V
,
Schmidt
JM
, et al
The Tie-2 ligand angiopoietin-2 is stored in and rapidly released upon stimulation from endothelial cell Weibel-Palade bodies
.
Blood
2004
;
103
:
4150
6
.
15.
Reiss
Y
,
Droste
J
,
Heil
M
,
Tribulova
S
,
Schmidt
MH
,
Schaper
W
, et al
Angiopoietin-2 impairs revascularization after limb ischemia
.
Circ Res
2007
;
101
:
88
96
.
16.
Stratmann
A
,
Risau
W
,
Plate
KH
. 
Cell type-specific expression of angiopoietin-1 and angiopoietin-2 suggests a role in glioblastoma angiogenesis
.
Am J Pathol
1998
;
153
:
1459
66
.
17.
Yuan
HT
,
Khankin
EV
,
Karumanchi
SA
,
Parikh
SM
. 
Angiopoietin 2 is a partial agonist/antagonist of Tie2 signaling in the endothelium
.
Mol Cell Biol
2009
;
29
:
2011
22
.
18.
Gale
NW
,
Thurston
G
,
Hackett
SF
,
Renard
R
,
Wang
Q
,
McClain
J
, et al
Angiopoietin-2 is required for postnatal angiogenesis and lymphatic patterning, and only the latter role is rescued by Angiopoietin-1
.
Dev Cell
2002
;
3
:
411
23
.
19.
Mazzieri
R
,
Pucci
F
,
Moi
D
,
Zonari
E
,
Ranghetti
A
,
Berti
A
, et al
Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells
.
Cancer Cell
2011
;
19
:
512
26
.
20.
Venneri
MA
,
De Palma
M
,
Ponzoni
M
,
Pucci
F
,
Scielzo
C
,
Zonari
E
, et al
Identification of proangiogenic TIE2-expressing monocytes (TEMs) in human peripheral blood and cancer
.
Blood
2007
;
109
:
5276
85
.
21.
Coffelt
SB
,
Tal
AO
,
Scholz
A
,
De Palma
M
,
Patel
S
,
Urbich
C
, et al
Angiopoietin-2 regulates gene expression in TIE2-expressing monocytes and augments their inherent proangiogenic functions
.
Cancer Res
2010
;
70
:
5270
80
.
22.
Puri
MC
,
Rossant
J
,
Alitalo
K
,
Bernstein
A
,
Partanen
J
. 
The receptor tyrosine kinase TIE is required for integrity and survival of vascular endothelial cells
.
EMBO J
1995
;
14
:
5884
91
.
23.
Porat
RM
,
Grunewald
M
,
Globerman
A
,
Itin
A
,
Barshtein
G
,
Alhonen
L
, et al
Specific induction of tie1 promoter by disturbed flow in atherosclerosis-prone vascular niches and flow-obstructing pathologies
.
Circ Res
2004
;
94
:
394
401
.
24.
Hansen
TM
,
Singh
H
,
Tahir
TA
,
Brindle
NP
. 
Effects of angiopoietins-1 and -2 on the receptor tyrosine kinase Tie2 are differentially regulated at the endothelial cell surface
.
Cell Signal
2010
;
22
:
527
32
.
25.
Saharinen
P
,
Kerkela
K
,
Ekman
N
,
Marron
M
,
Brindle
N
,
Lee
GM
, et al
Multiple angiopoietin recombinant proteins activate the Tie1 receptor tyrosine kinase and promote its interaction with Tie2
.
J Cell Biol
2005
;
169
:
239
43
.
26.
Seegar
TC
,
Eller
B
,
Tzvetkova-Robev
D
,
Kolev
MV
,
Henderson
SC
,
Nikolov
DB
, et al
Tie1-Tie2 interactions mediate functional differences between angiopoietin ligands
.
Mol Cell
2010
;
37
:
643
55
.
27.
Felcht
M
,
Luck
R
,
Schering
A
,
Seidel
P
,
Srivastava
K
,
Hu
J
, et al
Angiopoietin-2 differentially regulates angiogenesis through TIE2 and integrin signaling
.
J Clin Invest
2012
;
122
:
1991
2005
.
28.
Coxon
A
,
Bready
J
,
Min
H
,
Kaufman
S
,
Leal
J
,
Yu
D
, et al
Context-dependent role of angiopoietin-1 inhibition in the suppression of angiogenesis and tumor growth: implications for AMG 386, an angiopoietin-1/2-neutralizing peptibody
.
Mol Cancer Ther
2010
;
9
:
2641
51
.
29.
Daly
C
,
Eichten
A
,
Castanaro
C
,
Pasnikowski
E
,
Adler
A
,
Lalani
AS
, et al
Angiopoietin-2 functions as a Tie2 agonist in tumor models, where it limits the effects of VEGF inhibition
.
Cancer Res
2013
;
73
:
108
18
.
30.
Davis
S
,
Papadopoulos
N
,
Aldrich
TH
,
Maisonpierre
PC
,
Huang
T
,
Kovac
L
, et al
Angiopoietins have distinct modular domains essential for receptor binding, dimerization and superclustering
.
Nat Struct Biol
2003
;
10
:
38
44
.
31.
Jeansson
M
,
Gawlik
A
,
Anderson
G
,
Li
C
,
Kerjaschki
D
,
Henkelman
M
, et al
Angiopoietin-1 is essential in mouse vasculature during development and in response to injury
.
J Clin Invest
2011
;
121
:
2278
89
.
32.
Strumberg
D
,
Schultheis
B
. 
Regorafenib for cancer
.
Expert Opin Invest Drugs
2012
;
21
:
879
89
.
33.
McTigue
M
,
Murray
BW
,
Chen
JH
,
Deng
YL
,
Solowiej
J
,
Kania
RS
. 
Feature Article: Molecular conformations, interactions, and properties associated with drug efficiency and clinical performance among VEGFR TK inhibitors
.
Proc Natl Acad Sci U S A
2012
;
109
:
18281
9
.
34.
Besterman
JM
,
Fournel
M
,
Dupont
I
,
Bonfils
C
,
Dubay
M
,
Ste-Croix
CR
, et al
Potent preclinical antitumor activity of MGCD265, an oral Met/VEGFR kinase inhibitor in phase II clinical development, in combination with taxanes or erlotinib
.
J Clin Oncol
28
, 
2010
(
suppl; abstr e13595
).
35.
Huang
H
,
Lai
JY
,
Do
J
,
Liu
D
,
Li
L
,
Del Rosario
J
, et al
Specifically targeting angiopoietin-2 inhibits angiogenesis, Tie2-expressing monocyte infiltration, and tumor growth
.
Clin Cancer Res
2011
;
17
:
1001
11
.
36.
Rini
BI
,
Szczylik
C
,
Tannir
NM
,
Koralewski
P
,
Tomczak
P
,
Deptala
A
, et al
AMG 386 in combination with sorafenib in patients (pts) with metastatic renal cell cancer (mRCC): A randomized, double-blind, placebo-controlled, phase II study
.
J Clin Oncol
29
: 
2011
(
suppl 7; abstr 309
).
37.
Karlan
BY
,
Oza
AM
,
Richardson
GE
,
Provencher
DM
,
Hansen
VL
,
Buck
M
, 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
;
30
:
362
71
.
38.
Gaast
AV
,
van Hagen
P
,
Hulshof
M
,
Richel
D
,
van Berge Henegouwen
MI
,
Nieuwenhuijzen
GA
, et al
Effect of preoperative concurrent chemoradiotherapy on survival of patients with resectable esophageal or esophagogastric junction cancer: Results from a multicenter randomized phase III study
.
J Clin Oncol
28
:
400s
, 
2010
(
suppl; abstr 5042
).
39.
Available from
: www.clinicaltrial.gov.
[Cited 2012 Dec.]
40.
Doppalapudi
VR
,
Huang
J
,
Liu
D
,
Jin
P
,
Liu
B
,
Li
L
, et al
Chemical generation of bispecific antibodies
.
Proc Natl Acad Sci U S A
2010
;
107
:
22611
6
.
41.
Rosen
LS
,
Mendelson
DS
,
Cohen
RB
,
Gordon
MS
,
Goldman
JW
,
Bear
IK
, et al
First-in-human dose-escalation safety and PK trial of a novel intravenous humanized monoclonal CovX body inhibiting angiopoietin 2
.
J Clin Oncol
28
:
210s
, 
2010
(
suppl 15; abstr 2524
).
42.
Natale
RB
,
Rizvi
NA
,
Infante
JR
,
Joseph
LJ
,
Viner
J
,
McDevitt
JT
, et al
Safety, pharmacokinetics, and antitumor activity of MEDI3617, a selective angiopoietin inhibitor, in adult patients with advanced solid tumors
.
J Clin Oncol
30
: 
2012
(
suppl; abstr TPS2621
).
43.
Hashizume
H
,
Falcon
BL
,
Kuroda
T
,
Baluk
P
,
Coxon
A
,
Yu
D
, et al
Complementary actions of inhibitors of angiopoietin-2 and VEGF on tumor angiogenesis and growth
.
Cancer Res
2010
;
70
:
2213
23
.
44.
Imanishi
Y
,
Hu
B
,
Jarzynka
MJ
,
Guo
P
,
Elishaev
E
,
Bar-Joseph
I
, et al
Angiopoietin-2 stimulates breast cancer metastasis through the alpha(5)beta(1) integrin-mediated pathway
.
Cancer Res
2007
;
67
:
4254
63
.
45.
Baluk
P
,
Hashizume
H
,
McDonald
DM
. 
Cellular abnormalities of blood vessels as targets in cancer
.
Curr Opin Genet Dev
2005
;
15
:
102
11
.
46.
Grothey
A
,
Cutsem
EV
,
Sobrero
A
,
Siena
S
,
Falcone
A
,
Ychou
M
, et al
Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial
.
Lancet
2013
;
381
:
303
12
.
47.
Wilhelm
SM
,
Dumas
J
,
Adnane
L
,
Lynch
M
,
Carter
CA
,
Schutz
G
, et al
Regorafenib (BAY 73-4506): a new oral multikinase inhibitor of angiogenic, stromal and oncogenic receptor tyrosine kinases with potent preclinical antitumor activity
.
Int J Cancer
2011
;
129
:
245
55
.
48.
Schmieder
R
,
Ellinghaus
P
,
Scholz
A
,
Bhargava
A
,
Wilhelm
SM
,
Zopf
D
, et al
Regorafenib (BAY 73–4506): anti-metastatic activity in a mouse model of colorectal cancer [abstract]
. In:
Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31–Apr 4; Chicago, IL
.
Philadelphia (PA)
:
AACR
; 
2012
.
Abstract nr 2337
.
49.
Sennino
B
,
Ishiguro-Oonuma
T
,
Wei
Y
,
Naylor
RM
,
Williamson
CW
,
Bhagwandin
V
, et al
Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors
.
Cancer Discov
2012
;
2
:
270
87
.
50.
MethylGene initiates phase II program for MGCD265 in solid tumors and non-small cell lung cancer patients
.
MethylGene Inc Press Release
, 
2009
:
September 02
.
51.
Kataoka
Y
,
Mukohara
T
,
Tomioka
H
,
Funakoshi
Y
,
Kiyota
N
,
Fujiwara
Y
, et al
Foretinib (GSK1363089), a multi-kinase inhibitor of MET and VEGFRs, inhibits growth of gastric cancer cell lines by blocking inter-receptor tyrosine kinase networks
.
Invest New Drugs
2012
;
30
:
1352
60
.
52.
Patel
K
,
Fattaey
A
,
Burd
A
. 
ACTB-1003: An oral kinase inhibitor targeting cancer mutations (FGFR), angiogenesis (VEGFR2, TEK), and induction of apoptosis (RSK and p70S6K)
J Clin Oncol
28
, 
2010
(
suppl; abstr e13665
).
53.
Hudkins
RL
,
Becknell
NC
,
Zulli
AL
,
Underiner
TL
,
Angeles
TS
,
Aimone
LD
, et al
Synthesis and biological profile of the pan-vascular endothelial growth factor receptor/tyrosine kinase with immunoglobulin and epidermal growth factor-like homology domains 2 (VEGF-R/TIE-2) inhibitor 11-(2-methylpropyl)-12,13-dihydro-2-methyl-8-(pyrimidin-2-ylamino)-4H-indazolo[5, 4-a]pyrrolo[3,4-c]carbazol-4-one (CEP-11981): a novel oncology therapeutic agent
.
J Med Chem
2012
;
55
:
903
13
.
54.
Komrokji
RS
,
List
AF
,
Khoury
HJ
,
Lancet
JE
,
Jabbour
E
,
Foudray
MC
, et al
Phase 1 dose-escalation/expansion study of the p38/Tie2 inhibitor ARRY-614 in patients with IPSS Low/Int-1 risk myelodysplastic syndromes [abstract]
. In:
Proceedings of the 53rd ASH Annual Meeting and Exposition; 2011 Dec 10–13
San Diego, CA
.
Abstract nr
118
.
55.
Gross
S
,
Winski
S
,
Pheneger
J
,
Anderson
D
,
Munson
M
,
Rodriguez
M
, et al
ARRY-614, a small molecule inhibitor of p38, Abl, Tie2 and VEGFR2 with multiple potential therapeutic applications [abstract]
. In:
Kinases Proceedings of the Cancer Meeting: Lessons from the Clinic 2006
; 
2006
Feb 14–19
;
Santa Fe, New Mexico
,
Abstract nr 14
.
56.
Dieras
V
,
Jassem
J
,
Dirix
LY
,
Guastalla
JP
,
Bono
P
,
Hurvitz
SA
, et al
A randomized, placebo-controlled phase II study of AMG 386 plus bevacizumab (Bev) and paclitaxel (P) or AMG 386 plus P as first-line therapy in patients (pts) with HER2-negative, locally recurrent or metastatic breast cancer (LR/MBC)
.
J Clin Oncol
29
:
2011
(
suppl; abstr 544
).
57.
Oliner
J
,
Min
H
,
Leal
J
,
Yu
D
,
Rao
S
,
You
E
, et al
Suppression of angiogenesis and tumor growth by selective inhibition of angiopoietin-2
.
Cancer Cell
2004
;
6
:
507
16
.
58.
MEDI-3617 ANGIO 2012: antiangiogenic therapy
. In:
Proceedings of the 14th International Symposium
; 
2012
Feb 2–4
;
La Jolla, CA
.
59.
Leow
CC
,
Coffman
K
,
Inigo
I
,
Breen
S
,
Czapiga
M
,
Soukharev
S
, et al
MEDI3617, a human anti-angiopoietin 2 monoclonal antibody, inhibits angiogenesis and tumor growth in human tumor xenograft models
.
Int J Oncol
2012
;
40
:
1321
30
.
60.
Papadopoulos
KP
,
Chau
NG
,
Patnaik
A
,
Adriaens
L
,
Lalani
AS
,
Daly
C
, et al
A phase I first-in-human study of REGN910, a fully human and selective angiopoietin-2 monoclonal antibody, in patients with advanced solid tumor malignancies
.
J Clin Oncol
29
:
2011
(
suppl; abstr TPS159
).
61.
Lalani
AS
,
Castanaro
C
,
Kyle
AH
,
Minchinton
AI
,
Thurston
G
,
Daly
C
. 
Combined blockade of VEGF and Ang2 promotes dramatic tumor necrosis and growth inhibition of colorectal cancer xenografts [abstract]
. In:
Proceedings of the Molecular Targets and Cancer Therapeutics Meeting;
2011
Nov 12–16
;
San Francisco, CA
.
Abstract nr A10.
62.
Daly
C
,
Castanaro
C
,
Adler
AP
,
Eichten
A
,
Lalani
AS
,
Papadopoulos
NJ
, et al
REGN910, a fully-human, Ang2-specific monoclonal antibody, inhibits tumor growth as a monotherapy and dramatically potentiates the effects of VEGF Trap (Aflibercept) [abstract]
. In:
Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2–6; Chicago, IL
.
Philadelphia (PA)
:
AACR
; 
2011
.
Abstract nr 3290
.
63.
Thomas
M
,
Kienast
Y
,
Scheuer
W
,
Lorenzon
E
,
Herting
F
,
Odin
M
, et al
Dual inhibition of Ang-2 and VEGF via a novel human bispecific bivalent IgG1 CrossMAb shows potent antiangiogenic, antitumoral, and antimetastatic efficacy and leads to a reduced side effect profile compared to single therapies [abstract]
. In:
Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31–Apr 4; Chicago, IL.
.
Philadelphia (PA)
:
AACR
; 
2012
.
Abstract nr 2319
.
64.
Rennel
ES
,
Regula
JT
,
Harper
SJ
,
Thomas
M
,
Klein
C
,
Bates
DO
, et al
A human neutralizing antibody specific to Ang-2 inhibits ocular angiogenesis
.
Microcirculation
2011
;
18
:
598
607
.
65.
Roschke
V
,
Lafleur
D
,
Hilbert
D
,
Kiener
P
,
inventors; Zyngenia, Inc., assignee
. 
ANG-2 binding complexes and uses thereof
.
United States patent WO-2012009705. 2012 Jan 19
.
66.
Palmer
GM
,
Tiran
Z
,
Zhou
Z
,
Capozzi
ME
,
Park
W
,
Coletta
C
, et al
A novel angiopoietin-derived peptide displays antiangiogenic activity and inhibits tumour-induced and retinal neovascularization
.
Br J Pharmacol
2012
;
165
:
1891
903
.