Tumor growth requires induction of an angiogenic program, and targeting of this program with antiangiogenic drugs shows an impact on tumor progression. However, although they are effective at reducing angiogenesis, these therapies have not produced widespread or enduring clinical benefit, which openly exposes their limitations. Here, we describe the current limitations of these therapies, including the known mechanisms and current controversies. Further, we present some of the recent approaches to predict these limitations and strategies to overcome them. With the development of meaningful predictive biomarkers and effective treatments that impede these limitations, longer and more robust efficacies will be achieved for a wider population of patients.

Significance: The clinical benefit of antiangiogenic drugs is restricted because of intrinsic and acquired limitations. Acknowledging and understanding these limitations will not only allow the development of effective predictive biomarkers but also help in devising new therapeutic strategies that achieve longer efficacies for a wider population of patients. Cancer Discov; 4(1); 31–41. ©2013 AACR.

Neoplastic growth requires increasing amounts of nutrients and oxygen to sustain expansive proliferation of tumor cells. Therefore, switching on an angiogenic program is a limiting step during neoplastic progression that provides the growing tumor with a supply of oxygen and nutrients. Although many cytokines and growth factors have been described to have proangiogenic properties, it is VEGF and its receptors (VEGFR) that are essential for tumor angiogenesis (1). Inhibitors of this signaling axis exhibit antiangiogenic properties with consequent antitumor effects; hence, these drugs have been robustly introduced into clinical protocols for the treatment of several types of tumors, either alone or in combination with traditional cytotoxic agents (2).

Although effective at reducing angiogenesis, these drugs have not produced widespread or enduring clinical benefits in many patients, thereby openly exposing their limitations. Here, we describe tumor responses and existing cellular and molecular knowledge on the limitations of antiangiogenic therapies, including the current controversies in this field (Fig. 1). Furthermore, we propose the identification of biomarkers of response and suggest several strategies or approaches to overcome these limitations to increase the efficacy and extend the benefits of current antiangiogenic therapies.

Figure 1.

Limitations of antiangiogenic therapies and possible ways to overcome them. Sketch depicting different aspects that pose limitations (red type) to the efficacy and duration of antiangiogenic therapies (blue arrows). Several approaches have been proposed to overcome these limitations (green type), which could be used as combination treatments to impede the limitations and extend the long-term efficacy of antiangiogenics (green arrows). FGFR, fibroblast growth factor receptor; HIF, hypoxia-inducible factor.

Figure 1.

Limitations of antiangiogenic therapies and possible ways to overcome them. Sketch depicting different aspects that pose limitations (red type) to the efficacy and duration of antiangiogenic therapies (blue arrows). Several approaches have been proposed to overcome these limitations (green type), which could be used as combination treatments to impede the limitations and extend the long-term efficacy of antiangiogenics (green arrows). FGFR, fibroblast growth factor receptor; HIF, hypoxia-inducible factor.

Close modal

In the clinic, targeting VEGF ligand (i.e., with antibodies or receptor traps) or its receptors, VEGFRs [i.e., with small-molecule tyrosine kinase inhibitors (TKI)], shows effectiveness in different tumor types. Surprisingly, clinical benefits are achieved when ligand-blocking drugs are combined with chemotherapeutic agents or radiotherapy, but on the contrary, small-molecule inhibitors of the receptor are given as monotherapy and not combined (3, 4). This empirical (clinically validated) use of these kinds of drugs seems to be a limitation on its own, but has not yet been mechanistically explained. By ligand binding (i.e., bevacizumab) or trapping (i.e., aflibercept), or by inhibition of intracellular signaling pathways with small-molecule TKIs, these drugs achieve a significant inhibition of the VEGF–VEGFR signaling axis. But then, why is single-agent VEGF-targeted therapy efficacious in certain cancers, such as renal cell cancer (RCC) or hepatocellular carcinoma, but shows considerably less clinical benefit in others, for example in colorectal cancer, for which this therapy is administered in combination with chemotherapy?

TKIs Alone Only in a Subgroup of Tumors

Abnormalities of the tumor vasculature are dependent on endothelial cells as well as on other vascular cells such as perivascular cells or pericytes that are also affected by factors secreted by tumor cells. Tumor pericytes have a functionally relevant association with endothelial cells, which not only contributes to their barrier role but also can promote the direct survival of endothelial cells (5, 6). Indeed, these cells mediate the stabilization of vessels based on synthesis of new basement membrane and tight association of endothelial and pericyte junctions. Molecularly, a specific cross-talk between endothelial cells and pericytes that implicates VEGF and platelet-derived growth factor (PDGF) is key for vascular formation and maintenance and creates a crucial therapeutic opportunity that has been exploited (7). In preclinical studies, pericytes have been shown to be relevant targets for effective antiangiogenic therapies, and several preclinical studies have shown the increased efficacy of targeting both pericytes and endothelial cells (8–10). Molecularly, inhibition of the PDGF receptor (PDGFR) in pericytes with small-molecule TKIs enhances the benefit of anti-VEGFR antiangiogenics, as endothelial cells become more sensitive to apoptosis in the absence of pericyte coverage (7). Interestingly, a retrospective study compared the effects of sunitinib and bevacizumab (in combination with IFN-α) in advanced metastatic RCC and found sunitinib to be superior to bevacizumab plus IFN-α in terms of progression-free survival (PFS; ref. 11). As a consequence, PDGFR inhibition has been developed clinically in the context of combined inhibition of VEGFR and PDGFR with dual-specificity small-molecule inhibitors, such as sunitinib, sorafenib, pazopanib, and others, that have ultimately achieved significant and robust clinical efficacies in RCC and hepatocellular carcinoma, two typical VEGF-dependent tumor types (12, 13).

Key Concepts

By affecting blood vessel formation, antiangiogenic drugs produce vascular trimming and intratumoral hypoxia. However, the clinical benefit of antiangiogenic drugs is restricted because of intrinsic and acquired limitations:

  • Effective vascular trimming can also decrease the delivery of cytotoxic drugs to tumors, and hence reduce their efficacy.

  • Intrinsic resistance is characterized by tumor indifference to antiangiogenic therapy and continued tumor growth despite treatment.

  • Resistance to antiangiogenics stems from selection of resistant clones provided with alternative mechanisms that lead to activation of angiogenesis even when the target of the drug remains inhibited.

  • Antiangiogenic drugs can trigger adaptive mechanisms of resistance in individual tumor cells that modulate their features and render them therapy-insensitive.

  • Therapy-induced reduction of oxygen levels within the tumor induces the accumulation of tumor-initiating (stem) cells.

  • Antiangiogenic therapies can also increase tumor aggressiveness in some cancer types due to proinvasive mechanisms and increased dissemination and metastasis.

Although the preclinical and clinical development of this class of VEGFR TKI small molecules is expanding for the treatment of RCC and hepatocellular carcinoma, this class of drugs has not yet shown significant efficacy in other tumor types where combination with chemotherapy is necessary. What is the basis of this limitation? Several mechanistic studies have postulated that the deeper therapeutic impact that TKIs exert on vessels could be the cause of this incompatibility, which is not observed with anti-VEGF drugs. Specifically, TKIs induce distinct vessel alterations via the inhibition of both intracellular VEGFR2 and pericytes through PDGFR, which is not achieved by anti-VEGF ligand drugs (7). Nevertheless, several clinical studies testing various TKIs combined with chemotherapy have failed because of increased toxicity of these combination therapies. The mechanistic explanation could be based on off-target activity of small-molecule TKIs, which could add to the toxicities of chemotherapeutic drugs (14, 15). Indeed, many of the VEGFR2 TKIs show off-target inhibition of c-Kit, which plays an important role in bone marrow progenitor cell mobilization, and its inhibition could lead to myelosuppresion and exacerbate the bone marrow toxicity of chemotherapy (3, 16, 17). The ultimate cause for this “incompatibility” is not yet fully understood and warrants further studies to overcome this limitation of VEGFR TKI drugs.

Anti-VEGF Ligand and Chemotherapy: Always Together?

Although it has been known for several years that anti-VEGF ligand drugs can enhance the antitumor effects of cytotoxic drugs, the underlying mechanism has been far from clear since the early positive results of drug combination trials (18). The most widespread explanation for such a mechanism is the “vascular normalization” theory, initially postulated by Jain (19). The theory proposes that anti-VEGF/VEGFR therapies induce a structural and functional normalization of tumor blood vessels and, as a result, blood flow is increased and cytotoxic drugs can more easily reach into the tumor (20). Indeed, with antiangiogenic treatment, immature blood vessels are pruned and vessel tortuosity and dilation decrease, producing a reduction of blood vessel leakage, vascular permeability, and interstitial fluid pressure (IFP), which alleviates edema in patients with cancer and provides an important clinical benefit (21–23). Several antiangiogenic drugs have been shown to induce morphologic normalization of the tumor vasculature in both preclinical and clinical studies (24–26), and in some cases the functionality of individual surviving blood vessels has improved (22, 27, 28).

Nevertheless, a more recent report has clinically shown that angiogenesis inhibitors can decrease the delivery of cytotoxic drugs to tumors in patients, and hence hinder their therapeutic benefits (29). By means of a very sensitive positron emission tomography (PET) imaging method, the authors evaluated uptake and retention of a cytotoxic drug (radiolabeled docetaxel) in patients with advanced-stage non–small cell lung carcinoma (NSCLC) treated with bevacizumab. In these 10 patients, VEGF inhibition induced a fast and sustained decrease, rather than an increase, in the penetration of both water and docetaxel into the tumors. These results contrast with those of previous studies in patients with rectal cancer (21) and in patients with glioblastoma (24), showing that bevacizumab treatment induced vascular normalization and increased glucose uptake, which was used as a surrogate for cytotoxic drug uptake.

The discrepancies between the recent observations and previous results could be due to differences in blood vessel networks and response to angiogenesis inhibitors between the cancer types studied, as it is known that these agents can affect blood vessels in different ways in various tissues (24, 30). However, the finding that, at least in patients with NSCLC, antiangiogenic therapy does not improve but rather decreases cytotoxic drug delivery to tumors is a limitation and could be the cause of the modest benefits of these combination therapies in NSCLC and other tumor types (31).

To circumvent this limitation (Fig. 1), several studies have aimed to pinpoint the time interval of the normalization effects of antiangiogenics (“window of normalization”) to optimize the benefits of vascular normalization–enhanced tumor drug delivery (32). However, there is high variation in preclinical studies and clinical data, and noninvasive imaging techniques that monitor tumor blood flow need to be developed to better define the duration of the “window of normalization” in every tumor type. Moreover, chronic treatment with VEGF pathway inhibitors eventually reduces tumor blood perfusion and increases tumor hypoxia in experimental animal studies (33, 34), suggesting that uninterrupted treatment may not be optimal for tumor vascular normalization–enhanced combination chemotherapy. Therefore, intermittent antiangiogenesis treatment schedules need to be investigated to ascertain whether, and under which conditions, repeated cycles of antiangiogenics to normalize the vasculature and increase drug uptake can be achieved. The potential of such cycles of renormalization of the tumor vasculature to facilitate vascular recovery and prolong synergy with chemotherapeutics needs to be carefully considered. Overall, studies are needed to verify whether sequencing of chemotherapy followed by antiangiogenic drugs or intermittent antiangiogenesis treatment could reveal important benefits to circumvent the limitations of this kind of drug.

The initial hypothesis was that antiangiogenesis therapy would not induce resistance (“resistant to resistance”) because it targeted the genetically stable endothelial cells instead of the unstable tumor cells themselves (35). Nevertheless, clinical and experimental evidence indicates that resistance to antiangiogenic therapy does indeed occur (36, 37). Among tumor responses to therapy, it is essential to distinguish between refractoriness, sometimes called intrinsic resistance, and acquired resistance (ref. 38; Fig. 1). Intrinsic resistance is characterized by tumor indifference to antiangiogenic therapy and continued tumor growth despite treatment with antiangiogenics such as bevacizumab, sorafenib, or sunitinib (24, 39). On the other hand, acquired resistance to antiangiogenics seems to stem from tumor adaptations to therapy instead of mutations or gene amplifications that characterize acquired resistance to other therapeutic strategies. In this form of resistance, alternative mechanisms lead to activation of angiogenesis even when the target of the drug remains inhibited (36, 40–42). In fact, clinical evidence of this plasticity has been described in metastatic RCC treated repeatedly with VEGFR inhibitors (37).

Several molecular mechanisms of intrinsic or acquired resistance have been described. The most prominent one stems from therapy-induced vascular trimming and hypoxia, where neoplastic cells respond to hypoxia by becoming tolerant and modifying their metabolic characteristics to resist low oxygenation, or by upregulating multiple proangiogenic molecules, including VEGFs, fibroblast growth factors (FGF), and angiopoietins, which promote revascularization and eventual resistance (41, 43). In this case, both tumor cells and the stroma can contribute to therapy resistance through recruitment of infiltrating cells, such as cancer-associated fibroblasts (CAF; ref. 44) and tumor-associated macrophages (TAM; ref. 45), that aid in the production of alternative proangiogenic factors (46, 47). Alternatively, tumors can be refractory to anti-VEGF therapies because they do not depend on sprouting angiogenesis driven by VEGF, as in the case of co-option of preexisting normal vessels of the tissue or vascular mimicry, in which neoplastic cells can directly form vessel walls (48).

Interestingly, there are several similarities among the mechanisms that lead to intrinsic or acquired resistance. The difference lies in the intrinsic characteristics of each tumor, as in the acquired type, tumors require some time to generate these molecular changes and become resistant to therapy, whereas in intrinsic resistance, tumors are immune to this therapy, as they have upfront expression of these alternative factors (49, 50).

Strategies to overcome the limitations of resistance to antiangiogenic therapies are currently an intense area of research in the field. Several studies have proposed cotargeting of VEGF and FGF signaling pathways to improve efficacy and overcome adaptive resistance to VEGF inhibition in the RIP-Tag2 model of pancreatic neuroendocrine tumors (43, 51). Specifically, the use of the dual FGF receptor (FGFR)/VEGFR TKI, brivanib, shows antiangiogenic activity due to inhibition of VEGFR1–3 and disruption of FGFR1–3, which overcomes resistance to VEGF-selective therapy and blocks FGF-dependent tumor proliferation (51, 52). Clinically, brivanib had shown promising activity as a single agent in hepatocellular carcinoma and in combination with cetuximab in colorectal cancer (53), but in a more recent phase III study in liver cancer, it did not show any benefit over sorafenib, a more restricted VEGFR inhibitor (BRISK-FL trial). Other studies have proposed to aim at the root of tumor adaptation with alternative combination strategies designed to overcome the emergence of hypoxia-induced resistance. Along these lines, dual-targeted strategies have been tested in xenografts with the combination of bevacizumab and hypoxia-inducible factor-1 (HIF-1) or Sp1 inhibitors, which enhanced the therapeutic efficacy of antiangiogenic treatments in these preclinical studies (47, 54).

Furthermore, as resistance reflects reversible adaptations to angiogenic blockade, it has been suggested that subsequent treatment with therapy that does not target angiogenesis might resensitize patients to antiangiogenic strategies (55). Given this suggestion, sequential treatment with an antiangiogenic drug followed by a non-antiangiogenic drug (i.e., another targeted therapy or chemotherapy) could resensitize patients to another antiangiogenic drug as a third line of treatment. Obviously, many studies are warranted to unravel the preclinical basis and clinical potential of this hypothetical sequential treatment and to determine its clinical benefit for patients.

Tumor adaptation to antiangiogenic therapies may also involve alterations in signaling pathways in tumor cells and the accumulation of tumor-initiating cells or cancer stem cells (CSC), limiting treatment efficacy (ref. 56; Fig. 1). Studies in breast-cancer xenograft models treated with antiangiogenic agents (sunitinib and bevacizumab) showed accumulation of a tumor-initiating cell subpopulation that expressed the enzyme aldehyde dehydrogenase and initiated tumors when reimplanted in other mice. Similar cell populations have been described in tissue samples from patients with inflammatory breast cancer (57) and in glioblastoma treated with combination therapies in mice (58).

Several different mechanisms have been described to underlie the antiangiogenic-induced selection for cells with tumor-initiating (stemness) capacity. First and foremost, CSCs produce higher levels of VEGF in both normal and hypoxic conditions compared with their non-CSC counterparts (59), and they can recruit high amounts of endothelial cell precursors for revascularization and tumor growth or regrowth (60), which could make them less sensitive to anti-VEGF therapies. Another proposed mechanism involves antiangiogenic therapy–triggered vascular trimming and hypoxia. In this case, tumors treated with antiangiogenics activate not only a hypoxia-response program, but also the Akt/β-catenin signaling pathway, which regulates cell growth and adhesion between cells. This pathway has been previously implicated in the regulation of breast cancer progenitor cells (61). Furthermore, some intrinsic features of CSCs have been implicated in their resistance to chemo- and radiotherapy, and these same features could also be involved in their selection or insensitivity to antiangiogenic therapy. The most significant example is the low proliferative index that CSCs share with normal stem cells that could help them to survive under the conditions of nutrient and oxygen starvation generated by antiangiogenic therapies. The broad capacity of tumors to adapt to therapy and their intrinsic cellular plasticity initiates a rewiring of survival signals that triggers stemness and therapy resistance. Indeed, several reports have recently described the differential capacity of tumor-initiating cells to resist or survive after several treatment strategies, including not only chemo- and radiotherapy but also targeted therapies (62, 63).

How could this limitation of antiangiogenic therapies be overcome? One possibility would be to combine angiogenesis inhibitors with drugs that suppress the response of cancer cells to hypoxia and, as mentioned before in the case of acquired resistance, try to hit at the root or the cause of tumor adaptation. Another alternative could be the combination of antiangiogenics with inhibitors of the Akt/β-catenin pathway aimed at blocking the signaling pathways that mediate selection of stem-like cells and ultimately impeding tumor-initiating cell accumulation.

Tumor angiogenesis is intimately associated with systemic dissemination of tumor cells and the consequent development of distant metastases. The typically disorganized vascular tumor network, characterized by a loose association of basement membrane with endothelial cells and pericytes and by leaky blood vessels, serves as a permissive escape route for tumor cells (64). Moreover, the consequent formation of edema due to blood extravasation, together with the pushing force generated by the expansion of growing tumor cells and the absence of draining lymphatic vessels, converts the tumor microenvironment into an area with high IFP (20, 65). Under these conditions, the IFP inside the tumor could equalize with the microvascular pressure and generate a fluid convection to the surrounding tissue that may contribute to tumor-cell dissemination via the lymphatic and blood vessels, especially when considering the venous contribution to tumor-cell metastasis (65, 66). Thus, the aberrant and hyperpermeable tumor vasculature provides the push (higher IFP) and the opportunity (endothelial gaps and openings) for “passive” tumor cell intravasation and systemic dissemination (20). Therefore, blocking tumor angiogenesis by inhibition of the VEGF pathway should not only reduce tumor growth but also impede the formation of metastases. However, angiogenesis inhibitors have been shown to make some tumors more aggressive in several animal models, with potent angiogenesis inhibition altering the natural history of tumors by triggering resistance to therapy and increasing invasion and lymphatic or hematogenous metastasis (refs. 67, 68; Fig. 1). Here again, the mechanism of this more malignant phenotype is associated with tumor adaptation to therapy-induced hypoxia. Indeed, evidence linking hypoxia to a more aggressive metastatic cell behavior is well established (69), with HIF-1–dependent production of prometastatic proteins, the secretion of proteolytic enzymes, and alteration of adhesion molecules in tumor cells or in the extracellular matrix resulting in induction of epithelial-to-mesenchymal transition (70). Nevertheless, recent evidence suggests that other mechanisms, alone or in addition to hypoxia, could also be involved in the malignant response to these therapies, as in the case of activation of c-MET signaling leading to enhanced invasion and metastasis after anti-VEGF therapy (71–73).

Several studies have tried to address this issue in the clinical setting. Initially, a retrospective study analyzed tumor “rebound” after stopping antiangiogenic therapy in patients with metastatic cancer from five pooled phase III trials (74). The authors concluded that the disease developed with the same pattern of progression in bevacizumab- and placebo-treated patients, thus excluding an acceleration of tumor progression and an increase in mortality rates due to treatment. However, this analysis excluded a number of other clinical trials that reported somewhat discordant results (74, 75). More- recent clinical studies in the adjuvant setting (the AVANT trial and the NSABP-C08 trial) reported that patients in the bevacizumab-containing arm who relapsed after adjuvant treatment seemed to have a higher rate of death than those treated with chemotherapy alone (76, 77). Therefore, it will be crucial to evaluate tumor aggressiveness in future trials with antiangiogenic therapies.

How can this detrimental limitation be overcome? Similar to the emergence of acquired resistance and the accumulation of tumor-initiating cells, the possibility of restoring tumor oxygenation represents an appealing option to prevent tumor invasiveness/aggressiveness, as hypoxia is one of the drivers of neoplastic metastasis. Following Jain's (20) “vascular normalization” theory, several studies showed that controlled normalization of tumor oxygenation can be obtained by modulating the endothelial cell response to hypoxia with reduced activity of the oxygen sensor prolyl hydroxylase domain protein 2 (PHD2; refs. 78, 79). In animals heterozygous for PHD2, normalization of the endothelial lining restored perfusion and oxygen supply, and tumors were less invasive and developed fewer metastases than tumors with wild-type vessels. Similarly, studies in a transgenic model of pancreatic neuroendocrine tumors showed that restoration of tumor oxygenation levels through the normalization of endothelial cells impaired neoplastic invasion and dissemination (80). In this study, recombinant semaphorin-3A (Sema3A) was used as a therapeutic agent to inhibit both angiogenesis and invasive/metastatic behavior (80, 81). Nevertheless, these approaches should be taken with caution, as chronic exposure of cells to moderate hypoxia levels represents a well-recognized experimental method to induce adaptation to growth in hypoxic conditions and subsequent selection of resistant clones. In addition, these same intermediate levels of oxygen could increase the metastatic capability of cells (82). Therefore, intensity and potency of normalization of the tumor vasculature and its associated tumor reoxygenation should be carefully evaluated to avoid undesirable side effects.

Tumor responses to antiangiogenic therapies are everything but homogeneous. Although studies mostly report, and have to be based on, population (global) averages, the reality is that individual responses are very heterogeneous and variable. Indeed, results from preclinical models, case studies, and clinical trials report varying and often contradictory responses to antiangiogenic therapies, depending on the cancer type and the specific antiangiogenic therapy. The general agreement is that antiangiogenic treatments are more effective in terms of increasing PFS than prolonging overall survival, as in the case of VEGFR TKI for RCC (83, 84). In detail, anti-VEGF/R agents typically induce cavitation and loss of viable tumor mass, which exerts an impact on tumor growth, but this is not always associated with a significant alteration of tumor size and results in heterogeneity in individual Response Evaluation Criteria in Solid Tumors (RECIST) responses (38, 85). Indeed, the pattern of growth and the modifications induced at the site of tumor development are greatly influenced by the tumor type and in particular by its angiogenic features and proangiogenic capacity. A typical example is RCC, where angiogenesis is highly VEGF-dependent, in part due to frequent gene inactivation of the von Hippel-Lindau (VHL) tumor suppressor that drives dysregulated HIF-1 and VEGF overproduction (86). The same dependence on angiogenesis is thought to beimportant for the efficacy of antiangiogenic therapy in hepatocellular carcinoma, where tumors are highly angiogenic and displace the normal parenchyma. In contrast, the metastatic foci of colorectal cancer that grow in the liver often replace rather than displace the liver parenchyma, leading to co-option of existing blood vessels instead of dependence on sprouting angiogenesis (38, 87). This differential growth pattern, displacing or replacing the normal tissue, could be key in determining angiogenesis- and VEGF-dependency, and therefore response to antiangiogenic therapies. Furthermore, responses to antiangiogenic drugs also vary between primary tumors and their metastases (88). Interestingly, as the interplay between tumor cells and the tumor microenvironment is crucial for the development of neoplastic lesions, the same tumor–stromal cell collaboration is also involved in tumor responses to therapeutic inhibition of the VEGF pathway. Thus, together with tumor-cell characteristics, the stroma contributes to therapeutic inefficacy with a differential contribution in each cancer subtype, resulting in even more heterogeneity in responses to therapy.

With this broad spectrum of heterogeneity in tumor responses to antiangiogenics, it is logical that the different angiogenic features and vascular dependence of each type of tumor do indeed influence and limit the overall response to antiangiogenic therapy. But could this collection of limiting factors be managed and converted into predictors of sensitivity or response to therapy? Several years ago, hypertension was introduced as a putative biomarker for antiangiogenic treatments. Specifically, in a retrospective study in more than 500 patients with metastatic RCC, sunitinib treatment–induced hypertension was associated with an improved clinical outcome (89). These findings support the hypothesis that hypertension may be a viable “on-therapy biomarker” or a “monitoring biomarker” of antitumor efficacy in this patient population, although development of hypertension during sunitinib treatment was neither necessary nor sufficient for clinical benefit in all patients. Nevertheless, basal (pretreatment) hypertension in these patients did not associate with clinical benefit, and therefore blood pressure is not a useful predictive biomarker but rather an on-therapy monitor of treatment response. More recently, many studies have evaluated putative predictive factors of response to antiangiogenics, some stemming from tumor-cell adaptation to therapy (and its resulting hypoxia), and others initiating from the vascular structure and components (as it is the direct target of these therapies; Table 1). An example of the latter is a study where antiangiogenic-refractory tumors contained blood vessels with a prolific investment of pericytes expressing α-smooth muscle actin (α-SMA). Therefore, the occurrence of pericytes expressing α-SMA was postulated as a biomarker for tumors refractory to therapy (90). Obviously, further studies are warranted to validate this possibility. Another relevant example is the immune system and its intricate relationship with angiogenesis in cancer (91), as TAMs and myeloid-derived suppressor cells (MDSC) produce proinflammatory cytokines, endothelial growth factors, and proteases that drive neoangiogenesis (92, 93). Along these lines, preliminary studies suggest that several immune markers such as intratumoral MDSCs and interleukin-8 (IL-8), or peripheral regulatory T cells, may predict clinical response to antiangiogenic therapy. Nevertheless, there is not yet a recognized and validated biomarker of response to antiangiogenics (94).

Table 1.

Predictors of limitations to antiangiogenic therapies and avenues for intervention

Limitation to therapyBiomarkerTumor compartment contributionMethod of detectionInterventional therapyReferences
Alternative proangiogenic factors MSC Stroma IHC/flow cytometry: CXCR4+ Inhibition of VEGFR1+ (47, 97, 100) 
 TAM Stroma IHC/flow cytometry: M2 phenotype Interfere with recruitment by anti-PlGF (47, 97, 100) 
 TEM Stroma IHC/flow cytometry: Tie2+ Neutralization of ANG2 (101) 
 GR1+/CD11b+ Stroma IHC/flow cytometry: GR1+/CD11b+ Interfere with recruitment by anti-Bv8 (102) 
 FGF/PlGF/IL-8 Tumor cells/stroma Plasma/urine detection by ELISA Inhibition of FGF/FGFR, PlGF, etc. (36, 51, 95) 
 CSC Tumor cells Flow cytometry for specific markers (CD133, CD24, CD44, CD38, and CXCR4) Neutralization of CSCs by inhibition of specific targets (Notch, CXCR4, AKT-β-catenin pathway, etc.); ATRA (61–63) 
Independence from VEGF/VEGFR VEGF/VEGFR2 (low levels) Stroma Plasma/urine detection by ELISA Inhibit alternative proangiogenic factors (new targets) (76, 103) 
 SNP in VEGFR1 locus Stroma Genotype in blood Inhibition of VEGFR1 (72, 104) 
 Neuropilin Stroma NRP1 expression Inhibition of VEGF binding to NRP1 (97, 105) 
 Vascular cooption Tumor cells IHC: vWF-CD31 — (48) 
 Vascular mimicry Tumor cells IHC: vWF-CD31 — (48) 
Maintained vessel protection CAF Stroma IHC: α-SMA; S100A4 Neutralization of PDGF-C (46, 47, 97, 100) 
 CEC/CEP Stroma Flow cytometry in blood Simultaneous inhibition of VEGFR1 and VEGFR2 (106, 107) 
 Pericyte coverage Stroma IHC: NG2; α-SMA TKI (inhibition of PDGFR) (8, 10, 90) 
 Expression of HIF downstream pathways (PDGF, PlGF, etc.) Tumor cells/stroma HIF expression/expression of HIF-target genes (PDGF, PlGF, etc.) Inhibition of HIF-1; specific inhibition of HIF downstream genes (47, 54, 72, 108) 
Modification of EC features Proangiogenic miRNA Stroma miRNA expression in EC Regulation of miRNA expression (109) 
Access and distribution of the drug Hypoxia blood flow/permeability/IFP Stroma Tumor oxygen tension MRI/CT imaging/intratumoral needle Promote vascular normalization (inhibition of PHD2, administration of Sema3A, etc.) and new schedules. (20, 21, 47, 54, 65, 72, 79–81, 108, 110) 
Resistance to glucose starvation Glycolytic phenotype Tumor cells FDG uptake by PET imaging Inhibition of AMPK (111–113) 
 Expression of HIF target genes (GLUT1, LDHA, etc.) Tumor cells Expression of HIF target genes (GLUT1, LDHA, etc.); mutational profile (pVHL status) Inhibition of HIF-1; specific inhibition of HIF downstream genes (47, 54, 72, 86, 108) 
 Autophagy Tumor cells Autophagy markers (LC3, ATG16L, BNIP3, BNIP3L, and p62) Chemical inhibitors of autophagy; neutralization of p62 (114, 115) 
Resistant residual cells EGFR/c-KIT, etc. Tumor cells Expression of specific receptor Specific inhibition of the receptor (96, 116) 
 CSC Tumor cells Flow cytometry for specific markers (CD133, CD24, CD44, CD38, and CXCR4) Neutralization of CSCs by inhibition of specific targets (Notch, CXCR4, AKT-β-catenin pathway, etc.); ATRA (61–63) 
Metastatic potential CEC/CEP Stroma Flow cytometry in blood Simultaneous inhibition of VEGFR1 and VEGFR2 (106, 107) 
 CTC Tumor cells Blood CellSearch Technology Target molecules specifically expressed by CTCs (117–119) 
 CSC Tumor cells Flow cytometry for specific markers (CD133, CD24, CD44, CD38, and CXCR4) Neutralization of CSCs by inhibition of specific targets (Notch, CXCR4, AKT-β-catenin pathway, etc.); ATRA (61–63) 
 c-MET/EGFR Tumor cells Expression of specific receptor Specific inhibition of the receptor (72, 73) 
 Expression of HIF downstream pathways (CXCR4, SDF-1, MMP, etc.) Tumor cells/stroma Expression of HIF target genes (CXCR4, MMP, etc.); mutational profile (mTOR status) Inhibition of PHD2/HIF-1; specific inhibition of HIF downstream genes; inhibition of mTOR (47, 54, 72, 79, 108, 120) 
Limitation to therapyBiomarkerTumor compartment contributionMethod of detectionInterventional therapyReferences
Alternative proangiogenic factors MSC Stroma IHC/flow cytometry: CXCR4+ Inhibition of VEGFR1+ (47, 97, 100) 
 TAM Stroma IHC/flow cytometry: M2 phenotype Interfere with recruitment by anti-PlGF (47, 97, 100) 
 TEM Stroma IHC/flow cytometry: Tie2+ Neutralization of ANG2 (101) 
 GR1+/CD11b+ Stroma IHC/flow cytometry: GR1+/CD11b+ Interfere with recruitment by anti-Bv8 (102) 
 FGF/PlGF/IL-8 Tumor cells/stroma Plasma/urine detection by ELISA Inhibition of FGF/FGFR, PlGF, etc. (36, 51, 95) 
 CSC Tumor cells Flow cytometry for specific markers (CD133, CD24, CD44, CD38, and CXCR4) Neutralization of CSCs by inhibition of specific targets (Notch, CXCR4, AKT-β-catenin pathway, etc.); ATRA (61–63) 
Independence from VEGF/VEGFR VEGF/VEGFR2 (low levels) Stroma Plasma/urine detection by ELISA Inhibit alternative proangiogenic factors (new targets) (76, 103) 
 SNP in VEGFR1 locus Stroma Genotype in blood Inhibition of VEGFR1 (72, 104) 
 Neuropilin Stroma NRP1 expression Inhibition of VEGF binding to NRP1 (97, 105) 
 Vascular cooption Tumor cells IHC: vWF-CD31 — (48) 
 Vascular mimicry Tumor cells IHC: vWF-CD31 — (48) 
Maintained vessel protection CAF Stroma IHC: α-SMA; S100A4 Neutralization of PDGF-C (46, 47, 97, 100) 
 CEC/CEP Stroma Flow cytometry in blood Simultaneous inhibition of VEGFR1 and VEGFR2 (106, 107) 
 Pericyte coverage Stroma IHC: NG2; α-SMA TKI (inhibition of PDGFR) (8, 10, 90) 
 Expression of HIF downstream pathways (PDGF, PlGF, etc.) Tumor cells/stroma HIF expression/expression of HIF-target genes (PDGF, PlGF, etc.) Inhibition of HIF-1; specific inhibition of HIF downstream genes (47, 54, 72, 108) 
Modification of EC features Proangiogenic miRNA Stroma miRNA expression in EC Regulation of miRNA expression (109) 
Access and distribution of the drug Hypoxia blood flow/permeability/IFP Stroma Tumor oxygen tension MRI/CT imaging/intratumoral needle Promote vascular normalization (inhibition of PHD2, administration of Sema3A, etc.) and new schedules. (20, 21, 47, 54, 65, 72, 79–81, 108, 110) 
Resistance to glucose starvation Glycolytic phenotype Tumor cells FDG uptake by PET imaging Inhibition of AMPK (111–113) 
 Expression of HIF target genes (GLUT1, LDHA, etc.) Tumor cells Expression of HIF target genes (GLUT1, LDHA, etc.); mutational profile (pVHL status) Inhibition of HIF-1; specific inhibition of HIF downstream genes (47, 54, 72, 86, 108) 
 Autophagy Tumor cells Autophagy markers (LC3, ATG16L, BNIP3, BNIP3L, and p62) Chemical inhibitors of autophagy; neutralization of p62 (114, 115) 
Resistant residual cells EGFR/c-KIT, etc. Tumor cells Expression of specific receptor Specific inhibition of the receptor (96, 116) 
 CSC Tumor cells Flow cytometry for specific markers (CD133, CD24, CD44, CD38, and CXCR4) Neutralization of CSCs by inhibition of specific targets (Notch, CXCR4, AKT-β-catenin pathway, etc.); ATRA (61–63) 
Metastatic potential CEC/CEP Stroma Flow cytometry in blood Simultaneous inhibition of VEGFR1 and VEGFR2 (106, 107) 
 CTC Tumor cells Blood CellSearch Technology Target molecules specifically expressed by CTCs (117–119) 
 CSC Tumor cells Flow cytometry for specific markers (CD133, CD24, CD44, CD38, and CXCR4) Neutralization of CSCs by inhibition of specific targets (Notch, CXCR4, AKT-β-catenin pathway, etc.); ATRA (61–63) 
 c-MET/EGFR Tumor cells Expression of specific receptor Specific inhibition of the receptor (72, 73) 
 Expression of HIF downstream pathways (CXCR4, SDF-1, MMP, etc.) Tumor cells/stroma Expression of HIF target genes (CXCR4, MMP, etc.); mutational profile (mTOR status) Inhibition of PHD2/HIF-1; specific inhibition of HIF downstream genes; inhibition of mTOR (47, 54, 72, 79, 108, 120) 

NOTE: Basic mechanisms behind the limitations to antiangiogenic therapies with their predictive biomarkers and related methods of detection, together with opportunities for interventional therapy to overcome them.

Abbreviations: AMPK, AMP-activated protein kinase; ANG2, angiopoietin-2; ATRA, all-trans retinoic acid; CEC, circulating endothelial cell; CEP, circulating endothelial progenitor cell; CTC, circulating tumor cell; CT, computed tomographic scan; EC, endothelial cell; FDG, 2[18F]fluoro-2-deoxy-d-glucose; GLUT1, glucose transporter-1; IHC, immunohistochemistry; LDHA, lactate dehydrogenase A; miRNA, microRNA; MMP, matrix metalloproteinase; MSC, mesenchymal stem cell; NRP1, neuropilin-1; PlGF, placental growth factor; pVHL, VHL protein; SDF-1, stromal cell–derived factor-1; SNP, single-nucleotide polymorphism; TEM, Tie2-expressing monocyte; vWF, von Willebrand factor.

To be successful in the search for the elusive biomarkers of antiangiogenics, our view is that in the era of targeted/personalized therapy, patient populations have to be carefully studied to find the most appropriate marker or characteristic that defines dependence on tumor angiogenesis, and more specifically VEGF. Several morphologic, histologic, and molecular features in tumors are indeed known to be strongly associated with tumor vessel density, angiogenesis, and, therefore, VEGF signaling. However, not only VEGF is implicated in this process and, depending on the tumor, other proangiogenic factors overtake its function to become the main promoters of neovascularization (95). Therefore, due to their profound influence on therapeutic efficacy, we need to develop markers that quantify dependence on these factors to define subgroups of patients who will respond best to the specific blockade of the VEGF signaling pathway, angiogenesis, or other specific targets (36, 51).

Another important avenue with regard to biomarkers is the development of factors that predict the limitations of antiangiogenic therapies, because if we could predict the emergence of resistance or drawbacks of therapies, we would be able to develop strategies to overcome these limitations. The prediction of the limitations of antiangiogenics has to be based on the characteristics of each tumor at the tumor-cell and microenvironment levels. Indeed, tumor cells themselves can exhibit or switch on a repertoire of features that insure their survival under therapeutic selection, such as the expression of prosurvival receptors like EGF receptor (EGFR) and c-KIT or the activation of autophagic protection (96). Moreover, they can produce factors that modify the surrounding stroma, for example those related to HIF downstream genes. In addition, the stroma itself can also contribute to the failure of therapy if we consider the role of CAFs in vessel maintenance (97). With all these mechanisms and molecules described, we envision developing different biomarkers of response (or of failure) based on the detection of key molecules that critically contribute to each of the limitations to antiangiogenic therapies (Table 1).

On the other hand, the limitations to antiangiogenic therapies also exhibit very heterogeneous responses. In fact, preclinical data reveal that the same treatment elicits different and often contrasting responses depending on the tumor cell type and its microenvironment, varying from local adaptation to systemic dissemination, as mechanisms of escape from therapy (36, 98). A relevant example in the case of enhanced aggressiveness after antiangiogenics is the fact that sunitinib increases metastasis in orthotopic mouse models of breast and colon cancer, whereas it does not promote metastatic behavior in lung cancer, suggesting a role of intrinsic tumor-cell characteristics in this heterogeneous response to therapy (73). Could the different angiogenic features of each type of tumor also influence the escape behavior of tumor cells after antiangiogenic therapy? And does reduction of the oxygen and nutrient supplies due to inhibition of vessel growth more efficiently mobilize the angiogenesis-dependent tumor cells? In this case, and similar to the other limitations to antiangiogenic therapies previously described, the angiogenic features of the tumor and its angiogenesis dependence not only may be predictors of response to therapy but also may help to foresee the limitations that will limit the long-term benefits of antiangiogenic therapies.

Overall, the current knowledge in the field emphasizes the need for a carefully balanced evaluation of the benefits and limitations of antiangiogenic therapies. In addition, the causes and mechanisms of these limitations have to be unraveled in order to therapeutically overcome them (Fig. 1). We now know that the molecular profile of tumor cells as well as the microenvironmental features of the tumor can deeply affect the efficacy of therapy. Thus, the combination of antiangiogenic therapy with treatments targeting the tumor- signaling pathways or specific stromal cells implicated in both intrinsic and acquired limitations could prevent or delay treatment failure (Table 1). Therefore, inhibitors of these limiting pathways should also be taken into consideration in the development of new antiangiogenic therapeutic approaches, where maximal benefits could be achieved with upfront combinatorial or multitargeted therapies that block tumor angiogenesis and overcome intrinsic limitations. Alternatively, these treatments could be improved by sequential scheduling of antiangiogenic drugs followed by additive combinations with other drugs that target acquired limitations. This is particularly the case with acquired resistance that accounts for tumor desensitization to treatment. Hence, targeting the alternative pathways triggered or selected by previous antiangiogenic drugs could result in a marked improvement of the duration of benefit.

Nevertheless, not all the mechanistic predictions result in effective and clinically beneficial combinations. In the clinic, even in the presence of well-documented molecular pathways and targets of resistance, combinatorial/multitargeted therapies still fail in some patients, as in the case of erlotinib and bevacizumab in patients with breast cancer (99). Altogether, predictive factors of response but also predictors of the limitations are urgently needed as they could ultimately allow for categorization of subgroups of patients or even individualization of combination treatments to extend the benefits of antiangiogenic therapies for a wider population of patients. It should be our aim as translational researchers to convert the limitations of antiangiogenic therapies into therapeutic benefits for the future.

O. Casanovas has received honoraria from the Speakers Bureaus of Ipsen and Pfizer and is a consultant/advisory board member of Teva and Ipsen. No potential conflicts of interest were disclosed by the other authors.

Conception and design: L. Moserle, G. Jiménez-Valerio, O. Casanovas

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): O. Casanovas

Writing, review, and/or revision of the manuscript: L. Moserle, G. Jiménez-Valerio, O. Casanovas

Study supervision: O. Casanovas

The authors thank Mariona Graupera and Francesc Viñals for critical reading of the article and helpful suggestions.

The authors' work is supported by research grants from FP7-European Union (ERC-StG-281830), MICINN-Spain (SAF2009-08375 and SAF2012-36575), and AGAUR-Catalonia (SGR681).

1.
Carmeliet
P
. 
VEGF as a key mediator of angiogenesis in cancer
.
Oncology
2005
;
69
(
Suppl 3
):
4
10
.
2.
Ebos
JM
,
Kerbel
RS
. 
Antiangiogenic therapy: impact on invasion, disease progression, and metastasis
.
Nat Rev Clin Oncol
2011
;
8
:
210
21
.
3.
Motzer
RJ
,
Hutson
TE
,
Tomczak
P
,
Michaelson
MD
,
Bukowski
RM
,
Rixe
O
, et al
Sunitinib versus interferon alfa in metastatic renal-cell carcinoma
.
N Engl J Med
2007
;
356
:
115
24
.
4.
Escudier
B
,
Pluzanska
A
,
Koralewski
P
,
Ravaud
A
,
Bracarda
S
,
Szczylik
C
, et al
Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial
.
Lancet
2007
;
370
:
2103
11
.
5.
Gerhardt
H
,
Semb
H
. 
Pericytes: gatekeepers in tumour cell metastasis?
J Mol Med
2008
;
86
:
135
44
.
6.
Xian
X
,
Hakansson
J
,
Stahlberg
A
,
Lindblom
P
,
Betsholtz
C
,
Gerhardt
H
, et al
Pericytes limit tumor cell metastasis
.
J Clin Invest
2006
;
116
:
642
51
.
7.
Franco
M
,
Roswall
P
,
Cortez
E
,
Hanahan
D
,
Pietras
K
. 
Pericytes promote endothelial cell survival through induction of autocrine VEGF-A signaling and Bcl-w expression
.
Blood
2011
;
118
:
2906
17
.
8.
Bergers
G
,
Song
S
,
Meyer-Morse
N
,
Bergsland
E
,
Hanahan
D
. 
Benefits of targeting both pericytes and endothelial cells in the tumor vasculature with kinase inhibitors
.
J Clin Invest
2003
;
111
:
1287
95
.
9.
Pietras
K
,
Hanahan
D
. 
A multitargeted, metronomic, and maximum-tolerated dose “chemo-switch” regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cancer
.
J Clin Oncol
2005
;
23
:
939
52
.
10.
Saharinen
P
,
Alitalo
K
. 
Double target for tumor mass destruction
.
J Clin Invest
2003
;
111
:
1277
80
.
11.
Thompson Coon
JS
,
Liu
Z
,
Hoyle
M
,
Rogers
G
,
Green
C
,
Moxham
T
, et al
Sunitinib and bevacizumab for first-line treatment of metastatic renal cell carcinoma: a systematic review and indirect comparison of clinical effectiveness
.
Br J Cancer
2009
;
101
:
238
43
.
12.
Rini
BI
. 
Vascular endothelial growth factor-targeted therapy in metastatic renal cell carcinoma
.
Cancer
2009
;
115
:
2306
12
.
13.
Bukowski
RM
. 
Third generation tyrosine kinase inhibitors and their development in advanced renal cell carcinoma
.
Front Oncol
2012
;
2
:
13
.
14.
Liebler
DC
,
Guengerich
FP
. 
Elucidating mechanisms of drug-induced toxicity
.
Nat Rev Drug Discov
2005
;
4
:
410
20
.
15.
Hasinoff
BB
,
Patel
D
,
O'Hara
KA
. 
Mechanisms of myocyte cytotoxicity induced by the multiple receptor tyrosine kinase inhibitor sunitinib
.
Mol Pharmacol
2008
;
74
:
1722
8
.
16.
Motzer
RJ
,
Michaelson
MD
,
Redman
BG
,
Hudes
GR
,
Wilding
G
,
Figlin
RA
, et al
Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma
.
J Clin Oncol
2006
;
24
:
16
24
.
17.
Bhojani
N
,
Jeldres
C
,
Patard
JJ
,
Perrotte
P
,
Suardi
N
,
Hutterer
G
, et al
Toxicities associated with the administration of sorafenib, sunitinib, and temsirolimus and their management in patients with metastatic renal cell carcinoma
.
Eur Urol
2008
;
53
:
917
30
.
18.
Hurwitz
HI
,
Fehrenbacher
L
,
Hainsworth
JD
,
Heim
W
,
Berlin
J
,
Holmgren
E
, et al
Bevacizumab in combination with fluorouracil and leucovorin: an active regimen for first-line metastatic colorectal cancer
.
J Clin Oncol
2005
;
23
:
3502
8
.
19.
Jain
RK
. 
Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy
.
Nat Med
2001
;
7
:
987
9
.
20.
Jain
RK
. 
Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy
.
Science
2005
;
307
:
58
62
.
21.
Willett
CG
,
Boucher
Y
,
di Tomaso
E
,
Duda
DG
,
Munn
LL
,
Tong
RT
, et al
Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer
.
Nat Med
2004
;
10
:
145
7
.
22.
Huber
PE
,
Bischof
M
,
Jenne
J
,
Heiland
S
,
Peschke
P
,
Saffrich
R
, et al
Trimodal cancer treatment: beneficial effects of combined antiangiogenesis, radiation, and chemotherapy
.
Cancer Res
2005
;
65
:
3643
55
.
23.
Tong
RT
,
Boucher
Y
,
Kozin
SV
,
Winkler
F
,
Hicklin
DJ
,
Jain
RK
. 
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
6
.
24.
Batchelor
TT
,
Sorensen
AG
,
di Tomaso
E
,
Zhang
WT
,
Duda
DG
,
Cohen
KS
, 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
.
25.
Inai
T
,
Mancuso
M
,
Hashizume
H
,
Baffert
F
,
Haskell
A
,
Baluk
P
, et al
Inhibition of vascular endothelial growth factor (VEGF) signaling in cancer causes loss of endothelial fenestrations, regression of tumor vessels, and appearance of basement membrane ghosts
.
Am J Pathol
2004
;
165
:
35
52
.
26.
Nakamura
K
,
Taguchi
E
,
Miura
T
,
Yamamoto
A
,
Takahashi
K
,
Bichat
F
, et al
KRN951, a highly potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, has antitumor activities and affects functional vascular properties
.
Cancer Res
2006
;
66
:
9134
42
.
27.
Nakahara
T
,
Norberg
SM
,
Shalinsky
DR
,
Hu-Lowe
DD
,
McDonald
DM
. 
Effect of inhibition of vascular endothelial growth factor signaling on distribution of extravasated antibodies in tumors
.
Cancer Res
2006
;
66
:
1434
45
.
28.
Kan
Z
,
Kobayashi
S
,
Phongkitkarun
S
,
Charnsangavej
C
. 
Functional CT quantification of tumor perfusion after transhepatic arterial embolization in a rat model
.
Radiology
2005
;
237
:
144
50
.
29.
Van der Veldt
AA
,
Lubberink
M
,
Bahce
I
,
Walraven
M
,
de Boer
MP
,
Greuter
HN
, et al
Rapid decrease in delivery of chemotherapy to tumors after anti-VEGF therapy: implications for scheduling of anti-angiogenic drugs
.
Cancer Cell
2012
;
21
:
82
91
.
30.
Kamba
T
,
Tam
BY
,
Hashizume
H
,
Haskell
A
,
Sennino
B
,
Mancuso
MR
, et al
VEGF-dependent plasticity of fenestrated capillaries in the normal adult microvasculature
.
Am J Physiol Heart Circ Physiol
2006
;
290
:
H560
76
.
31.
Sandler
A
,
Gray
R
,
Perry
MC
,
Brahmer
J
,
Schiller
JH
,
Dowlati
A
, et al
Paclitaxel–carboplatin alone or with bevacizumab for non–small-cell lung cancer
.
N Engl J Med
2006
;
355
:
2542
50
.
32.
Weissleder
R
. 
Scaling down imaging: molecular mapping of cancer in mice
.
Nat Rev Cancer
2002
;
2
:
11
8
.
33.
Franco
M
,
Man
S
,
Chen
L
,
Emmenegger
U
,
Shaked
Y
,
Cheung
AM
, et al
Targeted anti-vascular endothelial growth factor receptor-2 therapy leads to short-term and long-term impairment of vascular function and increase in tumor hypoxia
.
Cancer Res
2006
;
66
:
3639
48
.
34.
Dings
RP
,
Loren
M
,
Heun
H
,
McNiel
E
,
Griffioen
AW
,
Mayo
KH
, et al
Scheduling of radiation with angiogenesis inhibitors anginex and Avastin improves therapeutic outcome via vessel normalization
.
Clin Cancer Res
2007
;
13
:
3395
402
.
35.
Boehm
T
,
Folkman
J
,
Browder
T
,
O'Reilly
MS
. 
Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance
.
Nature
1997
;
390
:
404
7
.
36.
Bergers
G
,
Hanahan
D
. 
Modes of resistance to anti-angiogenic therapy
.
Nat Rev Cancer
2008
;
8
:
592
603
.
37.
Rini
BI
,
Atkins
MB
. 
Resistance to targeted therapy in renal-cell carcinoma
.
Lancet Oncol
2009
;
10
:
992
1000
.
38.
Ellis
LM
,
Hicklin
DJ
. 
VEGF-targeted therapy: mechanisms of anti-tumour activity
.
Nat Rev Cancer
2008
;
8
:
579
91
.
39.
Kindler
HL
. 
Pancreatic cancer: an update
.
Curr Oncol Rep
2007
;
9
:
170
6
.
40.
Kerbel
RS
. 
Therapeutic implications of intrinsic or induced angiogenic growth factor redundancy in tumors revealed
.
Cancer Cell
2005
;
8
:
269
71
.
41.
Carmeliet
P
. 
Angiogenesis in life, disease and medicine
.
Nature
2005
;
438
:
932
6
.
42.
Kerbel
RS
. 
Molecular and physiologic mechanisms of drug resistance in cancer: an overview
.
Cancer Metastasis Rev
2001
;
20
:
1
2
.
43.
Casanovas
O
,
Hicklin
DJ
,
Bergers
G
,
Hanahan
D
. 
Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors
.
Cancer Cell
2005
;
8
:
299
309
.
44.
Kalluri
R
,
Zeisberg
M
. 
Fibroblasts in cancer
.
Nat Rev Cancer
2006
;
6
:
392
401
.
45.
Mantovani
A
,
Allavena
P
,
Sica
A
,
Balkwill
F
. 
Cancer-related inflammation
.
Nature
2008
;
454
:
436
44
.
46.
Azam
F
,
Mehta
S
,
Harris
AL
. 
Mechanisms of resistance to antiangiogenesis therapy
.
Eur J Cancer
2010
;
46
:
1323
32
.
47.
Rapisarda
A
,
Melillo
G
. 
Role of the hypoxic tumor microenvironment in the resistance to anti-angiogenic therapies
.
Drug Resist Updat
2009
;
12
:
74
80
.
48.
Spannuth
WA
,
Sood
AK
,
Coleman
RL
. 
Angiogenesis as a strategic target for ovarian cancer therapy
.
Nat Clin Pract Oncol
2008
;
5
:
194
204
.
49.
Oberg
K
,
Casanovas
O
,
Castano
JP
,
Chung
D
,
Delle Fave
G
,
Denefle
P
, et al
Molecular pathogenesis of neuroendocrine tumors: implications for current and future therapeutic approaches
.
Clin Cancer Res
2013
;
19
:
2842
9
.
50.
Jimenez-Valerio
G
,
Casanovas
O
. 
Molecular pathways and mechanisms of resistance to antiangiogenics.
In:
Galceran
JC
, editor. 
Integral atlas of renal cancer [in Spanish].
Madrid, Spain
:
You and Us
; 
2012
. p.
75
81
.
51.
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
310
.
52.
Chou
T
,
Finn
RS
. 
Brivanib: a review of development
.
Future Oncol
2012
;
8
:
1083
90
.
53.
Diaz-Padilla
I
,
Siu
LL
. 
Brivanib alaninate for cancer
.
Expert Opin Investig Drugs
2011
;
20
:
577
86
.
54.
Jia
Z
,
Zhang
J
,
Wei
D
,
Wang
L
,
Yuan
P
,
Le
X
, et al
Molecular basis of the synergistic antiangiogenic activity of bevacizumab and mithramycin A
.
Cancer Res
2007
;
67
:
4878
85
.
55.
Teule
A
,
Casanovas
O
. 
Relevance of angiogenesis in neuroendocrine tumors
.
Target Oncol
2012
;
7
:
93
8
.
56.
Conley
SJ
,
Gheordunescu
E
,
Kakarala
P
,
Newman
B
,
Korkaya
H
,
Heath
AN
, et al
Antiangiogenic agents increase breast cancer stem cells via the generation of tumor hypoxia
.
Proc Natl Acad Sci U S A
2012
;
109
:
2784
9
.
57.
Charafe-Jauffret
E
,
Ginestier
C
,
Iovino
F
,
Tarpin
C
,
Diebel
M
,
Esterni
B
, et al
Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer
.
Clin Cancer Res
2010
;
16
:
45
55
.
58.
Folkins
C
,
Man
S
,
Xu
P
,
Shaked
Y
,
Hicklin
DJ
,
Kerbel
RS
. 
Anticancer therapies combining antiangiogenic and tumor cell cytotoxic effects reduce the tumor stem-like cell fraction in glioma xenograft tumors
.
Cancer Res
2007
;
67
:
3560
4
.
59.
Bao
S
,
Wu
Q
,
Sathornsumetee
S
,
Hao
Y
,
Li
Z
,
Hjelmeland
AB
, et al
Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor
.
Cancer Res
2006
;
66
:
7843
8
.
60.
Folkins
C
,
Shaked
Y
,
Man
S
,
Tang
T
,
Lee
CR
,
Zhu
Z
, et al
Glioma tumor stem-like cells promote tumor angiogenesis and vasculogenesis via vascular endothelial growth factor and stromal-derived factor 1
.
Cancer Res
2009
;
69
:
7243
51
.
61.
Korkaya
H
,
Paulson
A
,
Charafe-Jauffret
E
,
Ginestier
C
,
Brown
M
,
Dutcher
J
, et al
Regulation of mammary stem/progenitor cells by PTEN/Akt/beta-catenin signaling
.
PLoS Biol
2009
;
7
:
e1000121
.
62.
Ribatti
D
. 
Cancer stem cells and tumor angiogenesis
.
Cancer Lett
2012
;
321
:
13
7
.
63.
Moserle
L
,
Ghisi
M
,
Amadori
A
,
Indraccolo
S
. 
Side population and cancer stem cells: therapeutic implications
.
Cancer Lett
2010
;
288
:
1
9
.
64.
Madsen
CD
,
Sahai
E
. 
Cancer dissemination—lessons from leukocytes
.
Dev Cell
2010
;
19
:
13
26
.
65.
Jain
RK
,
Tong
RT
,
Munn
LL
. 
Effect of vascular normalization by antiangiogenic therapy on interstitial hypertension, peritumor edema, and lymphatic metastasis: insights from a mathematical model
.
Cancer Res
2007
;
67
:
2729
35
.
66.
Chambers
AF
,
Groom
AC
,
MacDonald
IC
. 
Dissemination and growth of cancer cells in metastatic sites
.
Nat Rev Cancer
2002
;
2
:
563
72
.
67.
Paez-Ribes
M
,
Allen
E
,
Hudock
J
,
Takeda
T
,
Okuyama
H
,
Vinals
F
, et al
Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis
.
Cancer Cell
2009
;
15
:
220
31
.
68.
Ebos
JM
,
Lee
CR
,
Cruz-Munoz
W
,
Bjarnason
GA
,
Christensen
JG
,
Kerbel
RS
. 
Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis
.
Cancer Cell
2009
;
15
:
232
9
.
69.
Demir
R
,
Naschberger
L
,
Demir
I
,
Melling
N
,
Dimmler
A
,
Papadopoulus
T
, et al
Hypoxia generates a more invasive phenotype of tumour cells: an in vivo experimental setup based on the chorioallantoic membrane
.
Pathol Oncol Res
2009
;
15
:
417
22
.
70.
Lu
X
,
Kang
Y
. 
Hypoxia and hypoxia-inducible factors: master regulators of metastasis
.
Clin Cancer Res
2010
;
16
:
5928
35
.
71.
Lu
KV
,
Chang
JP
,
Parachoniak
CA
,
Pandika
MM
,
Aghi
MK
,
Meyronet
D
, et al
VEGF inhibits tumor cell invasion and mesenchymal transition through a MET/VEGFR2 complex
.
Cancer Cell
2012
;
22
:
21
35
.
72.
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
.
73.
Shojaei
F
,
Simmons
BH
,
Lee
JH
,
Lappin
PB
,
Christensen
JG
. 
HGF/c-Met pathway is one of the mediators of sunitinib-induced tumor cell type–dependent metastasis
.
Cancer Lett
2012
;
320
:
48
55
.
74.
Miles
D
,
Harbeck
N
,
Escudier
B
,
Hurwitz
H
,
Saltz
L
,
Van Cutsem
E
, et al
Disease course patterns after discontinuation of bevacizumab: pooled analysis of randomized phase III trials
.
J Clin Oncol
2010
;
29
:
83
8
.
75.
Potemski
P
. 
Is the postprogression survival time really not shortened in the bevacizumab-containing arms of phase III clinical trials?
J Clin Oncol
2011
;
29
:
e384
5
.
76.
de Gramont
A
,
Van Cutsem
E
,
Schmoll
HJ
,
Tabernero
J
,
Clarke
S
,
Moore
MJ
, et al
Bevacizumab plus oxaliplatin-based chemotherapy as adjuvant treatment for colon cancer (AVANT): a phase 3 randomised controlled trial
.
Lancet Oncol
2012
;
13
:
1225
33
.
77.
Allegra
CJ
,
Yothers
G
,
O'Connell
MJ
,
Sharif
S
,
Petrelli
NJ
,
Lopa
SH
, et al
Bevacizumab in stage II–III colon cancer: 5-year update of the National Surgical Adjuvant Breast and Bowel Project C-08 trial.
J Clin Oncol
2013
;
31
:
359
64
.
78.
Bautch
VL
. 
Endothelial cells form a phalanx to block tumor metastasis
.
Cell
2009
;
136
:
810
2
.
79.
Mazzone
M
,
Dettori
D
,
Leite de Oliveira
R
,
Loges
S
,
Schmidt
T
,
Jonckx
B
, et al
Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization
.
Cell
2009
;
136
:
839
51
.
80.
Maione
F
,
Capano
S
,
Regano
D
,
Zentilin
L
,
Giacca
M
,
Casanovas
O
, et al
Semaphorin 3A overcomes cancer hypoxia and metastatic dissemination induced by antiangiogenic treatment in mice
.
J Clin Invest
2012
;
122
:
1832
48
.
81.
Maione
F
,
Molla
F
,
Meda
C
,
Latini
R
,
Zentilin
L
,
Giacca
M
, et al
Semaphorin 3A is an endogenous angiogenesis inhibitor that blocks tumor growth and normalizes tumor vasculature in transgenic mouse models
.
J Clin Invest
2009
;
119
:
3356
72
.
82.
Subarsky
P
,
Hill
RP
. 
Graded hypoxia modulates the invasive potential of HT1080 fibrosarcoma and MDA MB231 carcinoma cells
.
Clin Exp Metastasis
2008
;
25
:
253
64
.
83.
Faris
JE
,
Michaelson
MD
. 
Targeted therapies: sunitinib versus interferon-alpha in metastatic RCC
.
Nat Rev Clin Oncol
2009
;
7
:
7
8
.
84.
Hotte
SJ
,
Bjarnason
GA
,
Heng
DY
,
Jewett
MA
,
Kapoor
A
,
Kollmannsberger
C
, et al
Progression-free survival as a clinical trial endpoint in advanced renal cell carcinoma
.
Curr Oncol
2011
;
18
(
Suppl 2
):
S11
9
.
85.
Faivre
S
,
Demetri
G
,
Sargent
W
,
Raymond
E
. 
Molecular basis for sunitinib efficacy and future clinical development
.
Nat Rev Drug Discov
2007
;
6
:
734
45
.
86.
Rosner
I
,
Bratslavsky
G
,
Pinto
PA
,
Linehan
WM
. 
The clinical implications of the genetics of renal cell carcinoma
.
Urol Oncol
2009
;
27
:
131
6
.
87.
Yoong
KF
,
Afford
SC
,
Randhawa
S
,
Hubscher
SG
,
Adams
DH
. 
Fas/Fas ligand interaction in human colorectal hepatic metastases: a mechanism of hepatocyte destruction to facilitate local tumor invasion
.
Am J Pathol
1999
;
154
:
693
703
.
88.
Jin
K
,
Lan
H
,
Cao
F
,
Han
N
,
Xu
Z
,
Li
G
, et al
Differential response to EGFR- and VEGF-targeted therapies in patient-derived tumor tissue xenograft models of colon carcinoma and related metastases
.
Int J Oncol
2012
;
41
:
583
8
.
89.
Rini
BI
,
Cohen
DP
,
Lu
DR
,
Chen
I
,
Hariharan
S
,
Gore
ME
, et al
Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib
.
J Natl Cancer Inst
2011
;
103
:
763
73
.
90.
Franco
M
,
Paez-Ribes
M
,
Cortez
E
,
Casanovas
O
,
Pietras
K
. 
Use of a mouse model of pancreatic neuroendocrine tumors to find pericyte biomarkers of resistance to anti-angiogenic therapy
.
Horm Metab Res
2011
;
43
:
884
9
.
91.
de Visser
KE
,
Eichten
A
,
Coussens
LM
. 
Paradoxical roles of the immune system during cancer development
.
Nat Rev Cancer
2006
;
6
:
24
37
.
92.
Grunewald
M
,
Avraham
I
,
Dor
Y
,
Bachar-Lustig
E
,
Itin
A
,
Jung
S
, et al
VEGF-induced adult neovascularization: recruitment, retention, and role of accessory cells
.
Cell
2006
;
124
:
175
89
.
93.
Du
R
,
Lu
KV
,
Petritsch
C
,
Liu
P
,
Ganss
R
,
Passegue
E
, et al
HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion
.
Cancer Cell
2008
;
13
:
206
20
.
94.
Maru
DM
,
Venook
AP
,
Ellis
LM
. 
Predictive biomarkers for bevacizumab: are we there yet?
Clin Cancer Res
2013
;
19
:
2824
7
.
95.
Folkman
J
. 
Angiogenesis: an organizing principle for drug discovery?
Nat Rev Drug Discov
2007
;
6
:
273
86
.
96.
Herbst
RS
,
Johnson
DH
,
Mininberg
E
,
Carbone
DP
,
Henderson
T
,
Kim
ES
, et al
Phase I/II trial evaluating the anti-vascular endothelial growth factor monoclonal antibody bevacizumab in combination with the HER-1/epidermal growth factor receptor tyrosine kinase inhibitor erlotinib for patients with recurrent non–small-cell lung cancer
.
J Clin Oncol
2005
;
23
:
2544
55
.
97.
Loges
S
,
Schmidt
T
,
Tjwa
M
,
van Geyte
K
,
Lievens
D
,
Lutgens
E
, et al
Malignant cells fuel tumor growth by educating infiltrating leukocytes to produce the mitogen Gas6
.
Blood
2010
;
115
:
2264
73
.
98.
Moserle
L
,
Casanovas
O
. 
Anti-angiogenesis and metastasis: a tumour and stromal cell alliance
.
J Intern Med
2012
;
273
:
128
37
.
99.
Dickler
MN
,
Rugo
HS
,
Eberle
CA
,
Brogi
E
,
Caravelli
JF
,
Panageas
KS
, et al
A phase II trial of erlotinib in combination with bevacizumab in patients with metastatic breast cancer
.
Clin Cancer Res
2008
;
14
:
7878
83
.
100.
Ebos
JM
,
Lee
CR
,
Kerbel
RS
. 
Tumor and host-mediated pathways of resistance and disease progression in response to antiangiogenic therapy
.
Clin Cancer Res
2009
;
15
:
5020
5
.
101.
De Palma
M
,
Lewis
CE
. 
Cancer: macrophages limit chemotherapy
.
Nature
2011
;
472
:
303
4
.
102.
Bronte
V
,
Apolloni
E
,
Cabrelle
A
,
Ronca
R
,
Serafini
P
,
Zamboni
P
, et al
Identification of a CD11b(+)/Gr-1(+)/CD31(+) myeloid progenitor capable of activating or suppressing CD8(+) T cells
.
Blood
2000
;
96
:
3838
46
.
103.
Jurgensmeier
JM
,
Schmoll
HJ
,
Robertson
JD
,
Brooks
L
,
Taboada
M
,
Morgan
SR
, et al
Prognostic and predictive value of VEGF, sVEGFR-2 and CEA in mCRC studies comparing cediranib, bevacizumab and chemotherapy
.
Br J Cancer
2013
;
108
:
1316
23
.
104.
Lambrechts
D
,
Claes
B
,
Delmar
P
,
Reumers
J
,
Mazzone
M
,
Yesilyurt
BT
, et al
VEGF pathway genetic variants as biomarkers of treatment outcome with bevacizumab: an analysis of data from the AViTA and AVOREN randomised trials
.
Lancet Oncol
2012
;
13
:
724
33
.
105.
Bielenberg
DR
,
Pettaway
CA
,
Takashima
S
,
Klagsbrun
M
. 
Neuropilins in neoplasms: expression, regulation, and function
.
Exp Cell Res
2006
;
312
:
584
93
.
106.
Rafii
S
,
Lyden
D
,
Benezra
R
,
Hattori
K
,
Heissig
B
. 
Vascular and haematopoietic stem cells: novel targets for anti-angiogenesis therapy?
Nat Rev Cancer
2002
;
2
:
826
35
.
107.
Ronzoni
M
,
Manzoni
M
,
Mariucci
S
,
Loupakis
F
,
Brugnatelli
S
,
Bencardino
K
, et al
Circulating endothelial cells and endothelial progenitors as predictive markers of clinical response to bevacizumab-based first-line treatment in advanced colorectal cancer patients
.
Ann Oncol
2010
;
21
:
2382
9
.
108.
Le
QT
,
Denko
NC
,
Giaccia
AJ
. 
Hypoxic gene expression and metastasis
.
Cancer Metastasis Rev
2004
;
23
:
293
310
.
109.
Landskroner-Eiger
S
,
Moneke
I
,
Sessa
WC
. 
miRNAs as modulators of angiogenesis
.
Cold Spring Harb Perspect Med
2013
;
3
:
a006643
.
110.
Willett
CG
,
Kozin
SV
,
Duda
DG
,
di Tomaso
E
,
Kozak
KR
,
Boucher
Y
, et al
Combined vascular endothelial growth factor–targeted therapy and radiotherapy for rectal cancer: theory and clinical practice
.
Semin Oncol
2006
;
33
:
S35
40
.
111.
Favaro
E
,
Nardo
G
,
Persano
L
,
Masiero
M
,
Moserle
L
,
Zamarchi
R
, et al
Hypoxia inducible factor-1alpha inactivation unveils a link between tumor cell metabolism and hypoxia-induced cell death
.
Am J Pathol
2008
;
173
:
1186
201
.
112.
Plathow
C
,
Weber
WA
. 
Tumor cell metabolism imaging
.
J Nucl Med
2008
;
49
(
Suppl 2
):
43S
63S
.
113.
Nardo
G
,
Favaro
E
,
Curtarello
M
,
Moserle
L
,
Zulato
E
,
Persano
L
, et al
Glycolytic phenotype and AMP kinase modify the pathologic response of tumor xenografts to VEGF neutralization
.
Cancer Res
2011
;
71
:
4214
25
.
114.
Mathew
R
,
Karp
CM
,
Beaudoin
B
,
Vuong
N
,
Chen
G
,
Chen
HY
, et al
Autophagy suppresses tumorigenesis through elimination of p62
.
Cell
2009
;
137
:
1062
75
.
115.
Mathew
R
,
White
E
. 
Autophagy in tumorigenesis and energy metabolism: friend by day, foe by night
.
Curr Opin Genet Dev
2011
;
21
:
113
9
.
116.
Zhang
YW
,
Staal
B
,
Essenburg
C
,
Lewis
S
,
Kaufman
D
,
Vande Woude
GF
. 
Strengthening context-dependent anticancer effects on non–small cell lung carcinoma by inhibition of both MET and EGFR
.
Mol Cancer Ther
2013
;
12
:
1429
41
.
117.
Cristofanilli
M
,
Budd
GT
,
Ellis
MJ
,
Stopeck
A
,
Matera
J
,
Miller
MC
, et al
Circulating tumor cells, disease progression, and survival in metastatic breast cancer
.
N Engl J Med
2004
;
351
:
781
91
.
118.
Danila
DC
,
Heller
G
,
Gignac
GA
,
Gonzalez-Espinoza
R
,
Anand
A
,
Tanaka
E
, et al
Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer
.
Clin Cancer Res
2007
;
13
:
7053
8
.
119.
Amadori
A
,
Rossi
E
,
Zamarchi
R
,
Carli
P
,
Pastorelli
D
,
Jirillo
A
. 
Circulating and disseminated tumor cells in the clinical management of breast cancer patients: unanswered questions
.
Oncology
2009
;
76
:
375
86
.
120.
Pantuck
AJ
,
Seligson
DB
,
Klatte
T
,
Yu
H
,
Leppert
JT
,
Moore
L
, et al
Prognostic relevance of the mTOR pathway in renal cell carcinoma: implications for molecular patient selection for targeted therapy
.
Cancer
2007
;
109
:
2257
67
.