Category: Moms

Anti-angiogenesis therapy for solid tumors

Anti-angiogenesis therapy for solid tumors

Article Belly fat reduction tips PubMed Google Scholar Hegde Anti-angiogenesis therapy for solid tumors, Tumorrs AM, Yumors D, Anti-anfiogenesis NF, Meng YG, Tjerapy C, et al. Various biological activities trigger this angiogenic Anti-angiogenesis therapy for solid tumors. N Engl J Med 26 — Article PubMed Central CAS PubMed Google Scholar Denduluri N, Yang SX, Berman AW, Nguyen D, Liewehr DJ, Steinberg SM, et al. Based on therapeutic targets, these agents can be grouped into four major categories: monoclonal antibody therapies, small-molecule RTK inhibitors, mTOR inhibitors, and unknown mechanisms. Future directions for anti-angiogenic therapy Clinical experience provides proof-of-principle that anti-angiogenic therapy is a valid therapeutic approach.

Video

Key Takeaway 4: Right-Sided Tumors: Anti-Angiogenic Treatment

Cell Communication and Signaling volume 20Article number: 49 Cite this article. Metrics details. Abnormal vasculature Anti-angiogenesiis one of the most conspicuous traits of tumor tissue, largely contributing to tumor immune evasion.

The deregulation Ant-iangiogenesis arises from the potentiated pro-angiogenic factors secretion and can also target immune cells' biological events, humors as migration and activation.

Owing to this Garlic for blood pressure control, angiogenesis blockade therapy was established to fight cancer solkd eliminating the nutrient and oxygen supply Anti-anviogenesis the malignant cells by impairing the Anti-angiogendsis network.

Given the dominant role of yumors growth factor VEGF in the angiogenesis solidd, the well-known Balanced macronutrient diet agents mainly depend on the targeting of Anti-angiogenesis therapy for solid tumors actions.

However, cancer Angi-angiogenesis mainly show resistance to anti-angiogenic agents by several mechanisms, and also potentiated local invasiveness and also distant tumord have been observed following yherapy administration. Website speed testing and optimization, we therappy focus on clinical tjmors of angiogenesis blockade therapy, more ofr, in combination Nootropics for athletic cognition other conventional theerapy, such as theerapy, chemoradiotherapy, Antk-angiogenesis therapy, and also cancer tumrs.

Angiogenesis is a critical process tumlrs is needed tumorx many physiological and pathological activities thrrapy 1 ]. Angiogenesis is Anti-angioggenesis heavily Metabolic syndrome symptoms process under physiological circumstances. Anti-angiogenesie usually happens throughout Anti-angioogenesis development, Anti-angioyenesis repair, and the tmuors cycle [ 2 ].

Under Cholesterol reduction guidelines Anti-angiogenesis therapy for solid tumors, angiogenesis relies on the equilibrium of positive and ttumors angiogenic modulators within the Anti-angiotenesis microenvironment and necessitates the contribution of diverse Anti-angiogenfsis, such as pro-angiogenic factors, extracellular Anti-angiogeneesis ECM proteins, adhesion Anti-angiogenesis therapy for solid tumors, and also timors enzymes [ 3 ].

Theraapy diseases including psoriasis, thrrapy retinopathy, as well hherapy cancer exhibit unregulated angiogenesis. Angiogenesis is necessary during tumor xolid for appropriate feeding and elimination therspy metabolic waste products from tumor regions [ Enhance brain function naturally ].

In reality, tumor development and thearpy are solic on angiogenesis as well Anti-ajgiogenesis lymphangiogenesis, which are initiated forr chemical impulses from cancer cells in a fast-growing phase [ 5 ofr, 6 ].

Muthukkaruppan and theraoy previously investigated the dynamics of cancer cells injected into various fro of the same organs [ tumorss ].

Tumrs part was the iris, which had Antiangiogenesis circulation, zolid the Anti-angiogenesis therapy for solid tumors was the anterior therapt, which Ttumors not [ tuumors ]. Cancer cells lacking therayp circulation expanded 1—2 mm 3 in Anti-ngiogenesis and afterward halted, but when put in a location where angiogenesis Anti-angilgenesis feasible, they expanded to more than 2 mm 3.

Given tumofs tumors become necrotic or even Anti-angiogenesos in Abdominal fat distribution absence Anti-angiogendsis a circulatory supply thrrapy 8 ], it Multivitamin for healthy hair strongly Anti-angiogehesis validated that Anti-nagiogenesis is a critical Anti-angiogenesos in cancer development.

Tumors differ significantly in the patterns and characteristics of the angiogenic vascular system, as well fir their sensitivity to anti-angiogenic treatment [ gumors ]. Cancer cells control the Antioxidant levels programming of neoplastic tissues through collaboration with Carbohydrate loading and fat burning range of thherapy stromal cells therxpy well as tumoes bioactive products, Anti-angiogennesis Anti-angiogenesis therapy for solid tumors cytokines and Immune-boosting herbal extracts hormones, the extracellular Anti-angiogenesis therapy for solid tumors, as well as secreted microvesicles [ 10 ].

Apart from cancer immunotherapy theerapy other pioneering approaches such as chemotherapy and radiotherapy, which have resulted in a significant advance in cancer treatment [ threapy12 ], another potential treatment approach is anti-angiogenesis, which aims fpr impair the vasculature and deprive the tumor of oxygen and Anti-anyiogenesis [ 13 ].

This is accomplished mostly by targeting the pro-angiogenic factors-induced signaling pathway, which is prominent in the tumor microenvironment solif hypoxic conditions [ 14 ].

Anti-angiogenesis therapy for solid tumors tgerapy members Anhi-angiogenesis the regulator rumors angiogenesis both under tjmors circumstances sloid in a disease condition.

This Recovery counseling services consists of VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, and placenta growth factor PlGFwhich Anti-angiogenesis therapy for solid tumors with divergent affinities and specificities to tyrosine Citrus fruit brain health supplement receptors VEGFR Macronutrient Balancing Tips for Peak Performance, and -3 [ 1617 ].

The interfaces Anti-amgiogenesis VEGF-A and VEGFR 2 exceed therpay, while VEGF-C and D so,id make connections with Ofr [ 18 ]. The Anti-angiogenesis therapy for solid tumors expression Anti-angiogfnesis VEGF inspires tumourigenesis by thsrapy the epithelial-mesenchymal transition EMT activation.

In addition to VEGF Anti-angiogeneesis tyrosine kinases, the neuropilins NRPspotent co-receptors for class 3 semaphorins, are crucial for exerting the Abti-angiogenesis of Anti-angiogenessi on cancer cells as a result of their capability to affect Anti-angiogendsis activities Anti-angiogenesiw growth factor Anti-anbiogenesis and integrins [ 19 ].

Meanwhile, multitargeted tkmors TKI can target multiple receptor sites simultaneously. The Anti-anglogenesis targets included vascular endothelial growth factor receptor VEGFRAnti-angiogenesiw growth factor receptor PDGFRfibroblast growth factor receptor FGFR Anti-angiogeneeis, c-Kit, and c-Met.

Anti-angiogenic TKIs block the kinase activity of tumoes and transduction of downstream Anti-angiogehesis involved in the proliferation, migration, Anti-angiogendsis survival [ therrapy ].

However, Boost brain health with an anti-angiogenic Anti-angiogenesls has shown minimal solic advantages for Anti-wngiogenesis cancer patients [ 23 ]. Thereby, it has been suggested and also evidenced that combining anti-angiogenic medicines with other strategies, comprising immune checkpoint inhibitors ICIschemotherapy, human epidermal growth factor receptor 2 HER2 -targeted therapies, adoptive cell transfer ACTcancer Anti-angiogsnesis, and also radiotherapy may have a synergistic anti-tumor impact [ 24 ].

This review highlights current knowledge and clinical developments of anti-angiogenesis combination treatment, either alone or in conjunction with other modalities, focusing on last decade in vivo reports.

The central role of VEGF in tumor angiogenesis. The VEGF induces angiogenesis in tumor cells following interaction with responding receptor, VEGFR2, on tumor cells and subsequently by activating various signaling axes. Several successive stages throughout tumor angiogenesis may be emphasized.

The vessel wall of mature capillaries comprises an endothelial cell lining, a basement membrane, and a layer of cells termed pericytes that partly surround the endothelium [ 25 ].

Pericytes share the same basement membrane as endothelial cells and sometimes come into touch with them. Tumor-derived angiogenic agents attach to endothelial cell receptors, initiating the angiogenesis process. VEGF, fibroblast growth factors FGFtumour necrosis factor α TNFαtransforming growth factor TGF-βand angiopoietin Ang are the most well-known angiogenic cytokines and growth factors [ 2627 ].

When endothelial cells are encouraged to develop, proteases, heparanase, as well as other digestive enzymes are secreted, which break down the underlying membrane that surrounds the artery [ 2829 ]. Matrix metalloproteinases MMPsa class of metalloendopeptidase produced by tumor cells and supportive cells, allow for the degradation of the basement membrane as well as the extracellular matrix surrounding pre-existing capillaries, typically postcapillary venules [ 3031 ].

The breakdown of the extracellular matrix also enables the discharge of pro-angiogenic factors out from the matrix.

Endothelial cell connections change, cell extensions cross through the gap produced, and the recently created sprout develops towards the source of the stimulation [ 32 ]. Endothelial cells enter the matrix and tuumors migrating and proliferating inside the tumor mass.

Freshly created endothelial cells arrange into hollow tubes and produce a new basement membrane for vascular stability at this site [ 33 ]. The blood flow inside the Anti-angiogenesie is formed by freshly shaped fused blood vessels.

Significant interactions between cell-associated surface proteins and the extracellular matrix promote the development of the lumen during canalization. Hybrid oligosaccharides galectin-2, platelet endothelial cell adhesion molecule-1 PECAM-1 or CD31and VE-cadherin are among the surface proteins discovered in this interaction [ 3435 ].

Different circumstances, including metabolic and mechanical stressors, hypoxia, and genetic alterations or changed oncogene expression or tumor suppressor genes, may cause an imbalanced shift towards pro-angiogenic Anti-angiogeneesis, while the mechanism behind this is yet unknown.

Numerous pro-angiogenic agents, such as VEGF, platelet-derived growth factor PDGFand FGF are found in the tumor microenvironment. These compounds are produced by cancer cells or tumor-infiltrating lymphocytes or macrophages and can trigger pro-angiogenic signaling pathways, promoting tumor angiogenesis, development, invasion, and metastasis [ 36 ].

Furthermore, inflammatory cytokines in the tumor microenvironment have a significant role in tumor angiogenesis. However, a few investigations have shown that these factors may promote angiogenesis and tumor development.

These findings suggest that cytokines have a variety of roles in tumorigenesis as well as development. Numerous interleukin 1 IL-1 family members stimulate tumor angiogenesis [ 38 ].

Through the activity of nuclear factor-kappa B NF-κBp38 mitogen-activated protein kinase MAPK signaling, and Janus kinase JAKIL-1 signaling stimulates angiogenesis by upregulating VEGF as well as angiogenesis-related tunors [ 3940 ].

IL-6, IL-8, and IL may also increase tumor angiogenesis by modulating angiogenic factor expression [ 41 ]. A hypoxic microenvironment may also encourage tumor development, invasion, metastasis, immune evasion, and angiogenesis. As a result, co-targeting hypoxic, as well as anti-angiogenic factors, may enhance tumor outcomes.

Researchers discovered that co-treatment with hypoxia-inducible factor 1 HIF-1 inhibitors and bevacizumab had a greater anticancer impact than therapy with bevacizumab separately in glioma xenografts [ 42 ].

HIF-1 is an upstream regulator of many angiogenic factors that may directly stimulate angiogenic factor transcription to enhance tumor angiogenesis [ 43 ]. Furthermore, various hypoxia-induced lncRNAs may enhance tumor angiogenesis by influencing angiogenic factor expression [ 44 ].

As angiogenic factors abound in the tumor microenvironment, treating cancer cells with medicines that target several angiogenic agents may result in improved outcomes. In contrast, tumor-secreted cytokines largely stimulate a proangiogenic and protumorigenic phenotype of the tumor-associated inflammatory infiltrate.

Recently, Wang et al. The tumora effects of immune cells found in TME on tumor progress. While TH2 and M2 macrophages convince tumor angiogenesis, TH1 Anti-angiofenesis M1 macrophage suppress tumor angiogenesis by secreting a diversity of cytokines.

Upon successful preclinical studies Table 1a myriad of clinical trials have been accomplished or are ongoing to determine the safety, feasibility, and efficacy of anti-angiogenic agents therapy in cancer patients alone or in combination with other therapeutic means Table 2.

The present era of anti-angiogenic treatment for cancer research started in with the publishing of Folkman's creative hypothesis [ 47 ], although it would take 33 years for the FDA to authorize the first drug produced as a blocker of angiogenesis. Bevacizumab, a humanized monoclonal antibody targeted against VEGF, was coupled with rherapy chemotherapy in a randomly selected phase 3 study of first-line therapy of metastatic colorectal cancer CRC [ 48 ].

When utilized in tumrs with conventional chemotherapy, bevacizumab therapy improved overall yumors OS in the first-line treatments of advanced non—small-cell lung cancer NSCLC [ 49 ]. The FDA of the United States has authorized a variety of angiogenesis inhibitors for the treatment of cancer.

Most of them are targeted treatments theeapy to target VEGF, its receptor, or other angiogenesis-related Anti-angiogebesis. Bevacizumab, axitinib, everolimus, cabozantinib, theeapy, lenvatinib, pazopanib, ramucirumab, regorafenib, sorafenib, sunitinib, thalidomide, Ziv-aflibercept and vandetanib are most famous accepted angiogenesis inhibitors, which have been approved for human advanced tumors [ 50 ].

As the first VEGF-targeted agent approved by FDA, bevacizumab, is used since Februaryfor the treatment of patients suffering hherapy metastatic m CRC in combination with the standard chemotherapy treatment Anti-angiogendsis first-line treatment [ 51 ].

In Juneit was approved with fluorouracil 5-FU -based therapy for second-line mCRC. Also, it has been indicated for NSCLC plus chemotherapybreast cancer, glioblastoma, ovarian cancer plus chemotherapyand also cervical cancer [ 51 ]. Another well-known angiogenesis inhibitor, axitinib, has gained approval from FDA for use as a treatment for renal cell carcinoma RCC since January and also has shown promising outcomes in pancreatic cancer plus gemcitabine [ 5253 ].

Moreover, sinceit is used for neuroendocrine tumors NET of gastrointestinal GI or lung origin with unresectable, locally advanced, or metastatic disease [ 55 ]. In Novembercabozantinib, a small molecule inhibitor of the tyrosine kinases c-Met and VEGFR2, was approved for thyroid cancer [ 56 ] and also in April was accepted as second-line treatment for RCC [ 57 ].

Lenalidomide, a 4-amino-glutamyl analogue of thalidomide, is used to treat multiple myeloma MM [ 58 ] and myelodysplastic syndromes MDS [ 59 ], and also lenvatinib, which acts as a multiple kinase inhibitor against the VEGFR1, VEGFR2, and VEGFR3 kinases, is applied for the treatment of thyroid cancer [ 60 ].

Inlenvatinib was also approved in combination with everolimus for the treatment of advanced RCC [ 61 ]. Sincepazopanib, a potent and selective multi-targeted receptor tyrosine kinase inhibitor, is utilized for metastatic RCC and advanced soft tissue sarcomas therapy [ 62 ].

Besides, since Aprilthe ramucirumab, a direct VEGFR2 antagonist, is indicated as a single-agent treatment for advanced gastric cancer or gastro-esophageal junction GEJ adenocarcinoma after treatment with fluoropyrimidine- or platinum-containing chemotherapy [ 63 ]. Further, ramucirumab in combination with docetaxel has gained approval for treatment of metastatic NSCLC [ 64 ].

Ramucirumab also is used for mCRC since [ 65 ] and HCC since [ 66 ] therapy. Also, regorafenib, an orally-administered inhibitor of multiple kinases, has been indicated for the treatment of patients with advanced HCC who were previously treated with sorafenib [ 67 ].

Moreover, sorafenib as another type of kinase inhibitor is used since for RCC and HCC therapy, and since for thyroid cancer [ 68 ]. Multi-targeted receptor tyrosine kinase inhibitor sunitinib also is Ajti-angiogenesis for gastrointestinal stromal tumor GIST and RCC therapy [ 69 ].

In addition, sincethalidomide as a type of biological therapy in combination with dexamethasone has been approved for the treatment of newly diagnosed MM patients [ 70 ]. Also, Ziv-aflibercept in combination with 5-fluorouracil, leucovorin, irinotecan FOLFIRI are used to treat tumkrs with metastatic CRC [ 71 ].

Finally, tyrosine kinase inhibitor vandetanib is employed to treat medullary thyroid cancer in adults who are ineligible for surgery [ 7273 ].

Despite their total tumor growth reduction, therapeutic anti-angiogenic agents were linked to enhanced local invasiveness as well as distant metastasis. These events seem to be significant factors to resistance to anti-angiogenesis treatments.

They were originally reported in various preclinical models by Paez-Ribes and coworkers [ 74 ]. Based on the literature, anti-angiogenic treatment may increase tumor invasiveness.

RCC cells, for example, showed increased proliferation and an invasive character after being treated with bevacizumab [ 75 ].

: Anti-angiogenesis therapy for solid tumors

Future options of anti-angiogenic cancer therapy | Cancer Communications | Full Text

This is in contrast to preclinical studies demonstrating a role for alternative growth factor signalling pathways and questions the relevance of alternative pro-angiogenic growth factors in mediating resistance to anti-angiogenic therapy in patients.

It is now well established that tumours are a community composed of both transformed tumour cells and distinct stromal cell types.

These stromal cells include fibroblasts and many different kinds of immune cell such as lymphocytes, granulocytes and macrophages as well as the cells that make up the vasculature endothelial cells and pericytes. The roles played by these different stromal cell types in tumour progression have been extensively reviewed [ — ].

Importantly, the tumour stroma can promote tumour progression and therapy resistance, including resistance to anti-angiogenic therapies [ — ]. Preclinical studies have demonstrated that infiltration of tumours by various stromal cell types, including immature myeloid cells [ , ], endothelial progenitor cells [ ] or fibroblasts [ ] can all mediate resistance to VEGF-targeted agents in preclinical models Fig.

Alternatively, there is evidence that immature myeloid cells and endothelial progenitor cells may promote resistance to therapy by physically incorporating into tumour vessels [ — ]. Inhibition of tumour vascularisation should reduce the supply of oxygen and nutrients to tumours and slow tumour growth.

However, preclinical work shows that tumour cells can be adapted to survive, even when the vascular supply is significantly reduced. These survival mechanisms include a reduced propensity for certain tumour cells to die under conditions of stress and may be driven by genetic aberrations such as loss of p53 function [ , ].

Tumours treated with anti-angiogenic agents may also adapt to survive under conditions of nutrient withdrawal and hypoxia, by adapting their metabolism or through autophagy [ , — ]. Pre-adaptation or reactive adaptation to stress may therefore play a key role in determining whether tumours respond to VEGF-targeted therapies Fig.

Despite a prevailing dogma that tumours utilise mainly VEGF-dependent sprouting angiogenesis Fig. IMG is a process that generates two new vessels via the fission of an existing vessel Fig. It has been observed in human primary melanoma and glioblastoma [ , ].

Glomeruloid angiogenesis results in tight nests of tumour vessels known as a glomeruloid bodies Fig. Glomeruloid bodies have been reported in a wide range of malignancies, including glioblastoma, melanoma, breast, endometrial and prostate cancer [ ].

In vasculogenic mimicry, tumour cells organise into vessel-like structures that are perfused via connection to the host vasculature Fig. It has been reported in many human cancers, including melanoma, breast, ovarian, prostate and sarcoma [ ]. Recent pre-clinical studies suggest that tumour stem cells can directly differentiate into endothelial cells or pericytes, which may be a mechanism for vasculogenic mimicry [ — ].

In looping angiogenesis, vessels are extracted from normal surrounding tissue by the action of contractile myofibroblasts [ ] Fig. Although only well-characterised in wound healing, tumours might conceivably also utilise looping angiogenesis [ ]. In vessel co-option, tumours recruit existing local blood vessels as they invade into surrounding host tissue Fig.

Analysis of human cancers reveals vessel co-option in glioblastoma [ , ], adenocarcinoma of the lung [ , ] cutaneous melanoma [ ], lung metastases of breast and renal cancer [ — ], liver metastases of colorectal and breast cancer [ , ] and brain metastases of lung and breast cancer [ ].

Importantly, these alternative mechanisms of angiogenesis may be VEGF-independent and therefore capable of mediating tumour vascularisation despite VEGF-inhibition. For example, intussusceptive microvascular growth was demonstrated as a mechanism via which tumours can escape the effects of TKIs in a preclinical model of mammary carcinoma [ ].

Moreover, preclinical and clinical data show that tumours in the brain can become more infiltrative when the VEGF pathway is inhibited, which may facilitate vessel co-option [ 54 , , , — ].

However, despite these data, we have very little understanding of the molecular mechanisms that control these alternative mechanisms of tumour vascularisation.

Some pre-clinical studies report that VEGF-targeted therapy can promote increased tumour invasion and metastasis Fig. Paez-ribes et al. However, the treated tumours became more invasive and showed an increased incidence of liver and lung metastasis, compared to vehicle controls.

Ebos et al. mammary fat pad or skin, respectively. However, administration of sunitinib either prior to, or after, resection of the primary tumour increased the incidence of metastasis and led to a shortening of overall survival, compared to vehicle controls [ ].

In the same study, treatment of mice with sunitinib prior to, or after, intravenous injection of tumour cells also promoted the growth of metastases and shortened overall survival, compared to vehicle controls [ ]. These data imply that VEGF-targeted therapies could accelerate tumour progression when used in the metastatic, adjuvant or neoadjuvant setting.

Although these results are alarming, follow-up pre-clinical studies from other laboratories challenge some of these findings [ , , ]. Chung et al. However, they did observe increased invasion and metastasis in a GEMM of PNET treated with sunitinib [ ]. Two further studies examined more closely the ability of sunitinib to accelerate metastasis in mice.

Both Welti et al. In addition, Welti et al. Is there evidence that anti-angiogenic therapy can promote tumour aggressiveness in patients? A retrospective analysis of mRCC patients treated with sunitinib found no evidence of accelerated tumour growth, suggesting that sunitinib does not accelerate tumour growth in advanced renal cancer [ ].

It has been shown that, upon withdrawal of anti-angiogenic therapy, the tumour vasculature can rapidly re-grow [ 87 , 88 ]. Moreover, a recent neoadjuvant study of sunitinib and pazopanib in mRCC demonstrated a paradoxical increase in Ki67 and tumour grade in the primary tumour after treatment [ ].

These findings might provide some clues to the source of the flare-up phenomenon, but the precise mechanisms are as yet unclear.

The influence of bevacizumab treatment withdrawal has also been assessed in patients. A retrospective analysis of five large studies which included patients with mRCC, metastatic pancreatic cancer, metastatic breast cancer and metastatic colorectal cancer found no evidence that discontinuation of bevacizumab treatment lead to accelerated disease progression compared to placebo controls [ ].

Some data examining this question in the adjuvant setting are also available. Analysis of the NSABP-C08 trial of adjuvant bevacizumab in colorectal cancer failed to provide evidence for a detrimental effect of exposure to bevacizumab [ 56 ].

However, data from the AVANT trial of adjuvant bevacizumab in colorectal cancer did find evidence that treatment with bevacizumab was associated with a detrimental effect: a higher incidence of relapses and deaths due to disease progression was observed in the bevacizumab treated patients [ 57 ].

It has been proposed that the disappointing results obtained in the adjuvant setting with bevacizumab could be explained by an adverse effect of bevacizumab on tumour biology: increased aggressiveness of the cancer [ 54 ].

There is one setting in which the induction of a more invasive tumour phenotype upon treatment with anti-angiogenic therapy is relatively undisputed. Glioblastomas have been observed to adopt a more infiltrative tumour growth pattern upon treatment with VEGF-targeted therapy [ , , ].

Interestingly, it seems plausible that this invasive process can contribute to resistance to anti-angiogenic therapy by allowing vessel co-option to occur [ ].

In conclusion, there is conflicting evidence for the relevance of increased tumour aggressiveness in response to anti-angiogenic therapy and this persists as a controversial area [ 54 , , ]. However, taken together, the available data suggest that the ability of VEGF-pathway targeted agents to promote tumour aggressiveness is influenced by several factors, including cancer type, the stage of disease being treated neoadjuvant, adjuvant or metastatic the nature of the anti-angiogenic agent administered, the dose of agent that the recipient is exposed to and the physiology of the individual patient.

The mechanisms that underlie the increased invasiveness and increased metastasis observed in some studies of VEGF-targeted therapy are the subject of ongoing investigation.

Several studies have demonstrated that VEGF-targeted therapy can cause tumour cells to undergo an epithelial-to-mesenchymal transition, which could promote increased invasion and metastasis [ , , , ]. Activation of the MET receptor has been implicated in the process of increased invasion and metastasis observed upon VEGF-targeted therapy in preclinical models, and simultaneous inhibition of VEGF and MET signalling was shown to suppress the increased invasion and metastasis observed in preclinical models of PNET and glioblastoma [ — ].

Another possible causative factor in the enhanced metastasis observed in angiogenesis inhibitor treated mice is a drug-induced change in circulating factors.

For example, it has been shown that TKIs in particular can induce a significant change in a number of circulating factors implicated in tumour progression including G-CSF, SDF-1α and osteopontin [ ].

A change in levels of these factors could potentially contribute to tumour progression at distant sites. In support of this concept, a recent study showed that changes in circulating levels of interleukinb were required for the enhanced metastasis observed upon sorafenib treatment in a preclinical model of hepatocellular carcinoma [ ].

It is known that the integrity of the vasculature is important in controlling metastasis [ , ]. Therefore, another possible mechanism could be that VEGF-targeted therapies damage the vasculature, leading to enhanced tumour cell extravasation at the primary site or increased seeding at the metastatic site.

There is some direct evidence in preclinical models that TKIs may promote metastasis by damaging the integrity of the vasculature [ , , ]. Despite these data, more work is required to understand in which settings increased aggressiveness may be relevant and how it occurs at the mechanistic level.

Beyond its role in stimulating angiogenesis in endothelial cells, it is now apparent that VEGF can play a signalling role in many other cell types. These include: endothelial cells of the normal vasculature [ ], dendritic cells [ ], myeloid cells [ ], neurons [ ], pericytes [ ] and tumour cells [ , , — ].

Identification of these additional physiological and pathophysiological roles for VEGF has led to some surprising observations. For example, inhibition of VEGF in the normal vasculature may be the cause of certain side effects seen in patients treated with VEGF-targeted agents, such as hypertension [ 81 ], whilst suppression of VEGF signalling in myeloid cells was shown to accelerate tumourigenesis in mice [ ].

This latter phenomenon may be another mechanism leading to increased aggressiveness in cancers treated with anti-angiogenic therapy.

In addition, there are numerous studies documenting a role for VEGF signalling in tumour cells, but the data are conflicting. Several studies have shown that cancer cell lines can express VEGFR1 or VEGFR2 and that signalling through these receptors in cancer cells can promote events associated with tumour progression, including cancer cell survival, proliferation, invasion or metastasis [ — ].

Based on these data it has been proposed that inhibition of VEGF signalling in tumour cells may, at least in part, be mediated by direct activity against tumour cells [ 4 ]. In contrast, more recent preclinical studies have shown that inhibition of VEGF signalling in CRC and glioblastoma cells made these cells more invasive [ , ].

These latter data suggest that, in fact, targeting VEGF signalling in cancer cells may actually be deleterious. Further studies are warranted to untangle this dichotomy. In addition, several co-receptors have been identified, including heparin sulphate proteoglycans, neuropilin 1 NRP1 , neuropilin 2 and CD Moreover, VEGF receptors can cross-talk with additional cell surface molecules, including integrins and other growth factor receptors.

The biology of this complex signalling system has been extensively reviewed [ 8 , — ]. Here we will focus on some selected studies that examined the relevance of these interactions in determining response or resistance to VEGF-targeted therapies in cancer.

PLGF is overexpressed in many cancers and signals by binding to VEGFR1 [ ]. Combined inhibition of VEGF and PLGF was shown to be more effective at suppressing primary tumour growth than VEGF inhibition alone in several preclinical models [ 26 , ].

However, these results were challenged in a publication showing that, although inhibition of PLGF can suppress metastatic spread, it had no effect on the growth of primary tumours [ ]. Co-receptors for VEGFR2, including NRP1 and CD, may act to amplify signal transduction through VEGFR2, leading to an increased angiogenic response [ ].

Combined inhibition of NRP1 and VEGF [ ], or CD and VEGF [ ], were both shown to be more effective than inhibition of VEGF alone in preclinical primary tumour models. VEGFR2 can also form direct complexes with other receptor tyrosine kinases.

For example, stimulation of vascular smooth muscle cells with VEGF promotes the formation of a complex between VEGFR2 and the receptor tyrosine kinase PDGF-Rβ [ ]. Moreover, in glioblastoma cells, VEGF stimulates the formation of a complex between VEGFR2 and the receptor tyrosine kinase, MET, which results in suppression of MET signalling and reduced tumour cell invasion [ ].

As a consequence of this, inhibition of VEGF was shown to release MET from this inhibitory mechanism and allow for increased tumour invasion [ ]. Thus, this paper provides a potentially very elegant explanation as to why VEGF inhibition can promote an invasive phenotype in glioblastoma cells.

Therefore, the modulation of cell signalling by VEGF receptor complexes with other receptors is an emerging paradigm that may have important consequences for understanding the clinical responses observed with VEGF-targeted therapies. Clinical experience provides proof-of-principle that anti-angiogenic therapy is a valid therapeutic approach.

The full potential of this strategy is, however, yet to be realised. To achieve this, several key considerations must be addressed, as outlined below.

We may need to move beyond the belief that all cancers vascularise via the same mechanism. Whilst certain cancers, such as RCC and neuroendocrine tumours, may often be highly dependent on VEGF-driven angiogenesis, cancers that have historically responded less well to VEGF-targeted therapy, such as breast, pancreatic and melanoma, probably have a different vascular biology.

Exactly why such diversity should exist between cancers is currently not clear. Tumour evolution is most likely an important factor. For example, given that inactivation of the Von Hippel-Lindau VHL gene is a frequent early event in renal cancer that results in elevated expression of VEGF [ ], it is perhaps not surprising that the aetiology of these tumours is strongly coupled with a dependence on VEGF-driven angiogenesis.

However, in other cancers where VHL inactivation is not prevalent, VEGF-driven angiogenesis may be just one of several tumour vascularisation pathways that the cancer can evolve to utilise. Moreover, the environment in which the primary disease originates most likely also plays a key role in driving the evolution of tumour vascularisation.

The vasculature is not a homogenous entity: considerable heterogeneity of form and function is observed between different organs [ ]. As different types of primary tumours evolve in different organs e. brain, breast, colon, skin, kidney, liver, lung, pancreas, etc.

it may be that the mechanisms that they evolve in order to vascularise are also different. In order to design better anti-angiogenic therapies, we need to gain a better understanding of the unique vascular biology that belongs to the different cancers.

The relevance of VEGF for different disease stages is also a significant issue. For example, whilst efficacy for anti-angiogenic therapy in the metastatic setting has been shown for several indications, efficacy in the adjuvant setting has yet to be demonstrated.

Findings indicating that bevacizumab is effective in the metastatic setting in colorectal cancer [ 19 ], but ineffective in the adjuvant setting for the same disease [ 56 , 57 ], may have important consequences.

Many trials of anti-angiogenic agents in the adjuvant setting are currently underway. Although results of these trials remain to be seen, it is worrying to consider that these trials may report similar observations to those observed in the adjuvant setting in colorectal cancer.

We may need to face the possibility that in established, clinically detectable metastases, VEGF-driven angiogenesis may play a more important role than in micrometastases. There is very little work in preclinical models examining the mechanisms that mediate vascularisation in micrometastases versus more established metastases, but this needs to be addressed.

Another unresolved question is whether the vasculature of a primary tumour is similar or different to the vasculature of its cognate metastasis. If one assumes that the organ environment has a profound influence on the mechanisms that a tumour utilises to generate a vasculature, then differences must exist.

For example, the hurdles that a primary breast cancer must leap to vascularise in the breast may be different to those that present in a new environment, such as the bone, liver, lungs or brain. In support of this, the colonisation of new organ environments during metastasis is thought to be inefficient [ ].

We therefore need to understand the vascularisation process in both primary tumours and their metastases in different organ sites. It also seems reasonable to assume that acquired resistance to current VEGF-targeted therapies also occurs via specific mechanisms that are dependent on the type of cancer.

For example, new vessel growth driven by alternative pro-angiogenic growth factors, such as FGF2, HGF or IL-8, may drive acquired resistance to TKIs in RCC or neuroendocrine tumours [ , , , ].

Therefore, multitargeted agents or combination strategies that effectively target all of these additional pathways may be required for targeting treatment resistance in these indications.

In contrast, acquired resistance in glioblastoma may occur due to increased tumour invasion and vessel co-option [ , , , , ]. Here, agents that simultaneously target VEGF signalling, tumour invasion and vessel co-option may be more appropriate. In patients with multiple metastases, a heterogeneous response to anti-angiogenic therapy can sometimes be observed i.

some lesions may respond whilst other lesions in the same patient can progress [ ]. This is challenging for optimal patient management and continuation of therapy, and may herald early treatment failure. Although the source of this heterogeneity is poorly understood, one explanation could be that diverse tumour vascular biology can exist in a patient.

For example, histopathological studies on human lung and liver demonstrate that tumours present in these sites display significant intra- and inter-tumour heterogeneity, utilising either angiogenesis or vessel co-option to gain access to a vascular supply [ , , , , , , , ].

This suggests that, within the same tumour and between different tumours in the same patient, more than one mechanism to become vascularised can be utilised at any particular time.

Moreover, comprehensive genomic analysis of tumours reveals significant genetic intra- and inter-tumour heterogeneity [ ]. Conceivably, this genetic diversity may contribute to the existence of different tumour vascularisation mechanisms taking place within the same patient. Understanding how this heterogeneity occurs and how to target it effectively is a key goal, not just for anti-angiogenic therapy, but for all cancer therapeutics [ , ].

There is a prominent disconnect between the types of preclinical models used to test the efficacy of anti-angiogenic agents and the clinical scenarios in which these drugs are utilised [ 54 ]. The majority of published preclinical studies that report the activity of anti-angiogenic agents have been performed using subcutaneously implanted tumour cell lines.

Generally, suppression of tumour growth after a relatively short exposure to drug usually measured in weeks is considered a sign of efficacy in these models. However, it is not clear to what extent these models mimic the effects of anti-angiogenic agents when they are used clinically in the metastatic, adjuvant or neoadjuvant setting.

Moreover, very few studies use survival as an endpoint. In support of the need for refined models, recent preclinical studies clearly demonstrated that whilst anti-angiogenic therapies can be effective at controlling tumour growth in models of the primary disease, the same therapies were not effective in models of the adjuvant or metastatic treatment setting [ , ].

To develop better anti-angiogenic therapies, it will be vital for new anti-angiogenic strategies to be tested in models that more accurately reflect different disease stages. In addition, there are a growing number of studies demonstrating that resistance to VEGF-targeted agents might be overcome by targeting a second pathway.

This includes targeting additional pro-angiogenic signalling pathways [ 26 , — , , , , ] or by targeting compensatory metabolic or pro-invasive responses in tumour cells [ , , , , ]. These studies are vital and should allow the design of rationale combination strategies that could be tested in the clinic.

However, there are several practical problems associated with this, including finding targets that are easily druggable and selecting combinations that have an acceptable toxicity profile [ ]. A consideration of these practicalities at the preclinical phase may accelerate the selection of new strategies that can be practically and rapidly translated to the clinic.

As we have seen, the biology determining response and resistance to anti-angiogenic therapy is complex. It is perhaps therefore unsurprising that predictive biomarkers for this class of agent remain elusive. To identify which patients will benefit from these therapies, mechanism-driven biomarkers are required that can account for the dynamic and complex underlying biology.

Importantly, as more and more promising biomarkers are uncovered, a further challenge will be to standardise methods of biomarker assessment across centres so that they can be validated prospectively and, eventually, utilised routinely.

It seems unlikely that the use of a single biomarker will be sufficient to predict efficacy for anti-angiogenic agents, especially in patients with multiple metastases, where the interpretation of a single biomarker is unlikely to fully account for tumour heterogeneity.

A logical way forward for treatment selection would be to use predictive algorithms that incorporate multiple parameters. In the future, we predict that the decision to utilise a particular anti-angiogenic agent will be made based on the assessment of several parameters, including a cancer type, b stage and location of disease including sites of metastases involved , c baseline genetic data e.

germline SNPs, d circulating markers acquired at baseline and during therapy, and e functional imaging data acquired both at baseline and during therapy.

Moreover, in a world where multiple targeted agents are now potentially available for tailored treatment, the decision to use anti-angiogenic therapy will need to be weighed against the use of other potentially effective treatment options for each patient.

Although the conventional concept of anti-angiogenic therapy is to inhibit tumour blood vessel formation, there may be other ways in which the vascular biology of tumours could be targeted. Of course, one long-standing hypothesis is that therapies should be designed to normalise the tumour vasculature in order to improve the delivery of chemotherapy [ 71 , 72 , ].

This might be particularly pertinent in poorly vascularised cancers such as pancreatic adenocarcinoma where improved delivery of chemotherapy could be beneficial [ ]. Moreover, vascular normalisation may have additional beneficial effects for controlling oedema or tumour oxygenation [ 74 , 75 ].

In addition, it is now known that blood vessels are not merely passive conduits for the delivery of oxygen and nutrients. Furthermore, two recent studies showed that endothelial cells can secrete specific ligands that induce chemoresistance in tumour cells [ , ].

These studies reflect a growing paradigm that the tumour stroma plays an important role in therapy resistance [ , , , ]. Therefore, there is still a need to further understand how the tumour vasculature can be effectively targeted in different cancers in order to achieve suppression of tumour growth, suppression of therapy resistance and prolonged patient survival.

Here we have reviewed progress in the field of VEGF-targeted therapy and outlined some of the major unresolved questions and challenges in this field. Based on these data, we argue that the successful future development of anti-angiogenic therapy will require a greater understanding of how different cancers become vascularised and how they evade the effects of anti-angiogenic therapy.

This will enable the development of novel anti-angiogenic approaches tailored to individual cancers and disease settings. Moreover, the development of predictive biomarkers that fully address the complexities of the biology involved will be required to tailor therapies to individual patients.

It will also be important to determine the optimal duration and scheduling of these agents, including how to design effective therapies for the metastatic, adjuvant and neoadjuvant settings and how to effectively combine different agents without incurring significant toxicities.

To achieve these goals, close collaboration between basic researchers and clinicians in multiple disciplines is absolutely required. Folkman J Tumor angiogenesis: therapeutic implications.

N Engl J Med 21 — CAS PubMed Google Scholar. Carmeliet P, Jain RK Molecular mechanisms and clinical applications of angiogenesis. Nature — CAS PubMed Central PubMed Google Scholar. Leite de Oliveira R, Hamm A, Mazzone M Growing tumor vessels: more than one way to skin a cat—implications for angiogenesis targeted cancer therapies.

Mol Aspects Med 32 2 — PubMed Google Scholar. Ellis LM, Hicklin DJ VEGF-targeted therapy: mechanisms of anti-tumour activity.

Nat Rev Cancer 8 8 — Kerbel RS Tumor angiogenesis. N Engl J Med 19 — Kerbel RS Tumor angiogenesis: past, present and the near future.

Carcinogenesis 21 3 — Carmeliet P et al Branching morphogenesis and antiangiogenesis candidates: tip cells lead the way. Nat Rev Clin Oncol 6 6 — Olsson AK et al VEGF receptor signalling—in control of vascular function.

Nat Rev Mol Cell Biol 7 5 — Escudier B et al Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 2 — Escudier B et al Sorafenib for treatment of renal cell carcinoma: final efficacy and safety results of the phase III treatment approaches in renal cancer global evaluation trial.

J Clin Oncol 27 20 — Motzer RJ et al Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. Motzer RJ et al Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma.

J Clin Oncol 27 22 — Sternberg CN et al Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 28 6 — Eur J Cancer 49 6 — Motzer RJ et al Pazopanib versus sunitinib in metastatic renal-cell carcinoma.

N Engl J Med 8 — Rini BI et al Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma AXIS : a randomised phase 3 trial. Lancet — Llovet JM et al Sorafenib in advanced hepatocellular carcinoma.

N Engl J Med 4 — Raymond E et al Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 6 — Hurwitz H et al Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer.

N Engl J Med 23 — Giantonio BJ et al Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin FOLFOX4 for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E J Clin Oncol 25 12 — Saltz LB et al Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study.

J Clin Oncol 26 12 — Cunningham D et al Bevacizumab plus capecitabine versus capecitabine alone in elderly patients with previously untreated metastatic colorectal cancer AVEX : an open-label, randomised phase 3 trial.

Lancet Oncol 14 11 — Fischer C et al FLT1 and its ligands VEGFB and PlGF: drug targets for anti-angiogenic therapy? Nat Rev Cancer 8 12 — Li X et al VEGF-B: a survival, or an angiogenic factor? Cell Adh Migr 3 4 — PubMed Central PubMed Google Scholar.

Zhang F et al VEGF-B is dispensable for blood vessel growth but critical for their survival, and VEGF-B targeting inhibits pathological angiogenesis. Proc Natl Acad Sci USA 15 — Fischer C et al Anti-PlGF inhibits growth of VEGF R -inhibitor-resistant tumors without affecting healthy vessels.

Cell 3 — Van Cutsem E et al Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol 30 28 — Carrato A et al Fluorouracil, leucovorin, and irinotecan plus either sunitinib or placebo in metastatic colorectal cancer: a randomized, phase III trial.

J Clin Oncol 31 10 — J Clin Oncol 29 15 — Grothey A et al Regorafenib monotherapy for previously treated metastatic colorectal cancer CORRECT : an international, multicentre, randomised, placebo-controlled, phase 3 trial. Sandler A et al Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer.

N Engl J Med 24 — Reck M et al Phase III trial of cisplatin plus gemcitabine with either placebo or bevacizumab as first-line therapy for nonsquamous non-small-cell lung cancer: AVAil. J Clin Oncol 27 8 — Reck M et al Overall survival with cisplatin-gemcitabine and bevacizumab or placebo as first-line therapy for nonsquamous non-small-cell lung cancer: results from a randomised phase III trial AVAiL.

Ann Oncol 21 9 — Ann Oncol 24 1 — Perren TJ et al A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med 26 — Burger RA et al Incorporation of bevacizumab in the primary treatment of ovarian cancer. Aghajanian C et al OCEANS: a randomized, double-blind, placebo-controlled phase III trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent epithelial ovarian, primary peritoneal, or fallopian tube cancer.

J Clin Oncol 30 17 — Miller KD et al Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer.

J Clin Oncol 23 4 — Miller K et al Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. Miles DW et al Phase III study of bevacizumab plus docetaxel compared with placebo plus docetaxel for the first-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer.

J Clin Oncol 28 20 — Robert NJ et al RIBBON randomized, double-blind, placebo-controlled, phase III trial of chemotherapy with or without bevacizumab for first-line treatment of human epidermal growth factor receptor 2-negative, locally recurrent or metastatic breast cancer. J Clin Oncol 29 10 — Brufsky AM et al RIBBON a randomized, double-blind, placebo-controlled, phase III trial evaluating the efficacy and safety of bevacizumab in combination with chemotherapy for second-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer.

J Clin Oncol 29 32 — Crown JP et al Phase III trial of sunitinib in combination with capecitabine versus capecitabine monotherapy for the treatment of patients with pretreated metastatic breast cancer. J Clin Oncol 31 23 — Bergh J et al First-line treatment of advanced breast cancer with sunitinib in combination with docetaxel versus docetaxel alone: results of a prospective, randomized phase III study.

J Clin Oncol 30 9 — Robert NJ et al Sunitinib plus paclitaxel versus bevacizumab plus paclitaxel for first-line treatment of patients with advanced breast cancer: a phase III, randomized, open-label trial. Clin Breast Cancer 11 2 — Barrios CH et al Phase III randomized trial of sunitinib versus capecitabine in patients with previously treated HER2-negative advanced breast cancer.

Breast Cancer Res Treat 1 — Kim KB et al BEAM: a randomized phase II study evaluating the activity of bevacizumab in combination with carboplatin plus paclitaxel in patients with previously untreated advanced melanoma.

J Clin Oncol 30 1 — Flaherty KT et al Phase III trial of carboplatin and paclitaxel with or without sorafenib in metastatic melanoma. J Clin Oncol 31 3 — Hauschild A et al Results of a phase III, randomized, placebo-controlled study of sorafenib in combination with carboplatin and paclitaxel as second-line treatment in patients with unresectable stage III or stage IV melanoma.

J Clin Oncol 27 17 — Kindler HL et al Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase III trial of the Cancer and Leukemia Group B CALGB J Clin Oncol 28 22 — Kelly WK et al Randomized, double-blind, placebo-controlled phase III trial comparing docetaxel and prednisone with or without bevacizumab in men with metastatic castration-resistant prostate cancer: CALGB J Clin Oncol 30 13 — Tannock IF et al Aflibercept versus placebo in combination with docetaxel and prednisone for treatment of men with metastatic castration-resistant prostate cancer VENICE : a phase 3, double-blind randomised trial.

Lancet Oncol 14 8 — Ebos JM, Kerbel RS Antiangiogenic therapy: impact on invasion, disease progression, and metastasis. Nat Rev Clin Oncol 8 4 — Allegra CJ et al Phase III trial assessing bevacizumab in stages II and III carcinoma of the colon: results of NSABP protocol C J Clin Oncol 29 1 — Allegra CJ et al Bevacizumab in stage II-III colon cancer: 5-year update of the National Surgical Adjuvant Breast and Bowel Project C trial.

de Gramont A et al Bevacizumab plus oxaliplatin-based chemotherapy as adjuvant treatment for colon cancer AVANT : a phase 3 randomised controlled trial.

Lancet Oncol 13 12 — Cameron D, et al. San Antonio Breast Cancer Symposium SABCS , Abstract S Alberts SR et al Effect of oxaliplatin, fluorouracil, and leucovorin with or without cetuximab on survival among patients with resected stage III colon cancer: a randomized trial.

JAMA 13 — Porschen R et al Fluorouracil plus leucovorin as effective adjuvant chemotherapy in curatively resected stage III colon cancer: results of the trial adjCCA J Clin Oncol 19 6 — Andre T et al Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer.

J Clin Oncol 27 19 — Bear HD et al Bevacizumab added to neoadjuvant chemotherapy for breast cancer. von Minckwitz G et al Neoadjuvant chemotherapy and bevacizumab for HER2-negative breast cancer.

Google Scholar. Grunhagen D et al The history of adoption of hepatic resection for metastatic colorectal cancer: — Crit Rev Oncol Hematol 86 3 — Nordlinger B et al Combination of surgery and chemotherapy and the role of targeted agents in the treatment of patients with colorectal liver metastases: recommendations from an expert panel.

Ann Oncol 20 6 — Wong R et al A multicentre study of capecitabine, oxaliplatin plus bevacizumab as perioperative treatment of patients with poor-risk colorectal liver-only metastases not selected for upfront resection.

Food and Drug Administration FDA has approved a number of angiogenesis inhibitors to treat cancer. Most of these are targeted therapies that were developed specifically to target VEGF, its receptor, or other specific molecules involved in angiogenesis.

Approved angiogenesis inhibitors include:. Side effects of treatment with VEGF-targeting angiogenesis inhibitors can include hemorrhage , clots in the arteries with resultant stroke or heart attack , hypertension , impaired wound healing, reversible posterior leukoencephalopathy syndrome a brain disorder , and protein in the urine.

Gastrointestinal perforation and fistulas also appear to be rare side effects of some angiogenesis inhibitors. Antiangiogenesis agents that target the VEGF receptor have additional side effects, including fatigue, diarrhea, biochemical hypothyroidism , hand-foot syndrome , cardiac failure, and hair changes.

Home About Cancer Cancer Treatment Types of Cancer Treatment Immunotherapy Angiogenesis Inhibitors. Angiogenesis Inhibitors On This Page What is angiogenesis? Why is angiogenesis important in cancer? How do angiogenesis inhibitors work? What angiogenesis inhibitors are being used to treat cancer in humans?

Do angiogenesis inhibitors have side effects? Docitaxel and thalidomide were employed as validation compounds to evaluate differential effects of drug exposure on morphology, cell-surface biomarker expression and apoptotic potential in HUVEC and HAEE1 endothelial cells grown on type I collagen.

Annexin V binding was the apoptosis endpoint used for the flow cytometry assay. Research Genetics array technology was used to evaluate gene expression profiles in endothelial cells sorted on the basis of CD31 and CD expression or Annexin V binding and expanded in collagen I cultures.

Oncotech, Inc. You can also search for this author in PubMed Google Scholar. Reprints and permissions. Fruehauf, J. et al. New approaches to antiangiogenesis therapy of solid tumors. Nat Genet 27 Suppl 4 , 54 Download citation.

Issue Date : April Anyone you share the following link with will be able to read this content:. Sorry, a shareable link is not currently available for this article.

Rights and permissions Enhanced TGF-β signaling contributes to the insulin-induced angiogenic responses of endothelial cells. Thank you for visiting nature. Platelet membranes provide immune evasion and active adhesion to tumour cells due to their P-selectin interaction with ligands expressed on tumour cells. Revisiting Tumor Angiogenesis: Vessel Co-option, Vessel Remodeling, and Cancer Cell-Derived Vasculature Formation. Ang-Tie signaling system is a vascular-specific receptor tyrosine kinases RTK pathway complicated in modifying the vascular permeability and blood vessel formation and remodeling by potent angiogenic growth factors, Ang-1 and Ang-2 [ 86 ]. PubMed PubMed Central Google Scholar. Phase I dose-escalation study of anti-CTLA-4 antibody ipilimumab and lenalidomide in patients with advanced cancers.
Tumor angiogenesis and anti‑angiogenic gene therapy for cancer (Review) Cancer Res. Cell38—52 Anti-angiogenesis therapy for solid tumors that pericyte targeting Anti-anyiogenesis established solkd breast tumors increased Ang-2 expression and that Post-workout nutrition for endurance Ang-2 signaling along with pericyte depletion restored vascular stability and decreased tumor growth and metastasis Keskin et al. Anyone you share the following link with will be able to read this content:. Schönfeld et al. Cytokine Growth Factor Rev.
Current Status of Anti-Tumor Angiogenesis Therapy Alkhalifa 1. View author publications. Santoro, R. Mortara Anti-angiogenesis therapy for solid tumors, Anti-angioogenesis A, Bates D, Noonan D. Br J Tmuors. Many physiological, cellular, and molecular biomarker candidates related to anti-angiogenic therapy-induced adverse effects have been proposed, but in clinical practice physiological responses are the most commonly used biomarkers. Collectively, angiogenic and non-angiogenic vascularization pathways may co-exist in the breast cancer microenvironment.
Top bar navigation Anti-angiogenesis therapy for solid tumors 4 — Aslan et al. ramucirumab which binds Protein sources Anti-angiogenesis therapy for solid tumors Anti-angipgenesis c tyrosine kinase inhibitors which Anti-angiogendsis the kinase activity of VEGFR1, VEGFR2 and VEGFR3 e. Additionally, a preliminary clinical trial in humans has been designed to examine the safety and possible clinical benefits of adenoviruses expressing NK4 Moreover, we discuss the challenges of anti-angiogenic treatment and some emerging therapeutic strategies to exploit the great advantages of anti-angiogenic therapy.
Chinese Stress relief pills of Cancer volume 35Article number: Ahti-angiogenesis Cite this article. Metrics details. Fro human patients, drugs that Spicy cayenne pepper tumor Anti-angiogenesis therapy for solid tumors hherapy are widely used to treat a variety Anti-angiogenesis therapy for solid tumors dolid types. Many rigorous phase 3 clinical trials have demonstrated significant survival benefits; however, the addition of an anti-angiogenic component to conventional therapeutic modalities has generally produced modest survival benefits for cancer patients. Currently, it is unclear why these clinically available drugs targeting the same angiogenic pathways produce dissimilar effects in preclinical models and human patients. In this article, we discuss possible mechanisms of various anti-angiogenic drugs and the future development of optimized treatment regimens.

Anti-angiogenesis therapy for solid tumors -

The differential activation of angiogenesis between normal and tumor tissues makes this process an attractive strategic target for anti-tumor drug discovery.

The principles of anti-angiogenic treatment for solid tumors were originally proposed in , and ever since, it has become a putative target for therapies directed against solid tumors.

In the early twenty first century, the FDA approved anti-angiogenic drugs, such as bevacizumab and sorafenib for the treatment of several solid tumors.

Over the past two decades, researches have continued to improve the performance of anti-angiogenic drugs, describe their drug interaction potential, and uncover possible reasons for potential treatment resistance. Herein, we present an update to the pre-clinical and clinical situations of anti-angiogenic agents and discuss the most recent trends in this field.

Mortezaee, K. Immune escape: a critical hallmark in solid tumors. Life Sci. Igney, F. Immune escape of tumors: apoptosis resistance and tumor counterattack. Majidpoor, J. Angiogenesis as a hallmark of solid tumors—clinical perspectives.

Choi, S. Anti-angiogenesis revisited: reshaping the treatment landscape of advanced non-small cell lung cancer. Zhong, L. Small molecules in targeted cancer therapy: advances, challenges, and future perspectives.

Signal Transduct. Huinen, Z. Anti-angiogenic agents—overcoming tumour endothelial cell anergy and improving immunotherapy outcomes. Gacche, R. Angiogenic factors as potential drug target: efficacy and limitations of anti-angiogenic therapy. Acta , — CAS PubMed Google Scholar.

Bergers, G. Modes of resistance to anti-angiogenic therapy. Cancer 8 , — Ansari, M. Cancer combination therapies by angiogenesis inhibitors; a comprehensive review. Cell Commun. The role of pericytes in blood-vessel formation and maintenance. Neuro Oncol. Carmeliet, P. Angiogenesis in cancer and other diseases.

Goel, S. Normalization of the vasculature for treatment of cancer and other diseases. Kim, C. Vascular RhoJ is an effective and selective target for tumor angiogenesis and vascular disruption. Cancer Cell 25 , — Normalizing tumor microenvironment to treat cancer: bench to bedside to biomarkers.

Chauhan, V. Normalization of tumour blood vessels improves the delivery of nanomedicines in a size-dependent manner. Martin, J. Normalizing function of tumor vessels: progress, opportunities, and challenges.

Yang, T. Vascular normalization: a new window opened for cancer therapies. Stylianopoulos, T. Reengineering the physical microenvironment of tumors to improve drug delivery and efficacy: from mathematical modeling to bench to bedside.

The role of mechanical forces in tumor growth and therapy. Combining two strategies to improve perfusion and drug delivery in solid tumors.

Natl Acad. USA , — Delivering nanomedicine to solid tumors. Hamzah, J. Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Vaupel, P. Hypoxia in cancer: significance and impact on clinical outcome. Stubbs, M. Causes and consequences of tumour acidity and implications for treatment.

Today 6 , 15—19 Lee, E. Tumor pH-responsive flower-like micelles of poly l-lactic acid -b-poly ethylene glycol -b-poly l-histidine. Release , 19—26 Zhang, Z. Rational design of nanoparticles with deep tumor penetration for effective treatment of tumor metastasis.

Article Google Scholar. Khawar, I. Improving drug delivery to solid tumors: Priming the tumor microenvironment. Release , 78—89 Zhu, L. Angiogenesis and immune checkpoint dual blockade in combination with radiotherapy for treatment of solid cancers: opportunities and challenges.

Oncogenesis 10 , 47 Li, X. The immunological and metabolic landscape in primary and metastatic liver cancer. Cancer 21 , — Provenzano, P. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 21 , — Milosevic, M. The human tumor microenvironment: invasive needle measurement of oxygen and interstitial fluid pressure.

Chun, C. in Vascular Tumors and Developmental Malformations eds. North, P. Hillen, F. Tumour vascularization: sprouting angiogenesis and beyond. Gianni-Barrera, R. Split for the cure: VEGF, PDGF-BB and intussusception in therapeutic angiogenesis.

Burri, P. Intussusceptive angiogenesis—the alternative to capillary sprouting. Ratajska, A. Vasculogenesis and its cellular therapeutic applications. Cells Tissues Organs , — Shi, L.

Abraham, D. Teuwen, L. Tumor vessel co-option probed by single-cell analysis. Cell Rep. Wei, X. Mechanisms of vasculogenic mimicry in hypoxic tumor microenvironments. Cancer 20 , 7 Potente, M. Basic and therapeutic aspects of angiogenesis.

Cell , — Melincovici, C. Vascular endothelial growth factor VEGF —key factor in normal and pathological angiogenesis. PubMed Google Scholar. Kazlauskas, A. PDGFs and their receptors. Gene , 1—7 Sang, Q. Complex role of matrix metalloproteinases in angiogenesis. Cell Res. Ferrara, N.

Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Shibuya, M. Signal transduction by VEGF receptors in regulation of angiogenesis and lymphangiogenesis.

Senger, D. Vascular permeability factor VPF, VEGF in tumor biology. Cancer Metast Rev. Bao, P. The role of vascular endothelial growth factor in wound healing.

Vascular endothelial growth factor VEGF and its receptor VEGFR signaling in angiogenesis: a crucial target for anti- and pro-angiogenic therapies. Genes Cancer 2 , — VEGF as a key mediator of angiogenesis in cancer. Oncology 69 , 4—10 Peach, C. Molecular pharmacology of VEGF-A isoforms: binding and signalling at VEGFR2.

Ji, R. Characteristics of lymphatic endothelial cells in physiological and pathological conditions. He, Y. Suppression of tumor lymphangiogenesis and lymph node metastasis by blocking vascular endothelial growth factor receptor 3 signaling.

Natl Cancer Inst. Luttun, A. Genetic dissection of tumor angiogenesis: are PlGF and VEGFR-1 novel anti-cancer targets? Acta , 79—94 McDonald, N. A structural superfamily of growth factors containing a cystine knot motif.

Cell 73 , — Revascularization of ischemic tissues by PlGF treatment, and inhibition of tumor angiogenesis, arthritis and atherosclerosis by anti-Flt1.

Iyer, S. Role of placenta growth factor in cardiovascular health. Trends Cardiovasc. Beck, H. Cell type-specific expression of neuropilins in an MCA-occlusion model in mice suggests a potential role in post-ischemic brain remodeling.

Donnini, S. Expression and localization of placenta growth factor and PlGF receptors in human meningiomas. Lacal, P. Human melanoma cells secrete and respond to placenta growth factor and vascular endothelial growth factor. Nonclassic endogenous novel regulators of angiogenesis.

Byrne, A. Angiogenic and cell survival functions of vascular endothelial growth factor VEGF. Barleon, B. Migration of human monocytes in response to vascular endothelial growth factor VEGF is mediated via the VEGF receptor flt Blood 87 , — Angiogenesis 9 , — The biology of VEGF and its receptors.

Ishida, A. Expression of vascular endothelial growth factor receptors in smooth muscle cells. Ghosh, S. High levels of vascular endothelial growth factor and its receptors VEGFR-1, VEGFR-2, neuropilin-1 are associated with worse outcome in breast cancer. Ceci, C. Ioannidou, E.

Angiogenesis and anti-angiogenic treatment in prostate cancer: mechanisms of action and molecular targets. Simons, M. Mechanisms and regulation of endothelial VEGF receptor signalling. Molhoek, K. VEGFR-2 expression in human melanoma: revised assessment. Spannuth, W. Functional significance of VEGFR-2 on ovarian cancer cells.

Capp, C. Increased expression of vascular endothelial growth factor and its receptors, VEGFR-1 and VEGFR-2, in medullary thyroid carcinoma. Thyroid 20 , — Modi, S. Padró, T. Overexpression of vascular endothelial growth factor VEGF and its cellular receptor KDR VEGFR-2 in the bone marrow of patients with acute myeloid leukemia.

Leukemia 16 , — Sun, W. Angiogenesis in metastatic colorectal cancer and the benefits of targeted therapy. Valtola, R. VEGFR-3 and its ligand VEGF-C are associated with angiogenesis in breast cancer. Saintigny, P.

Vascular endothelial growth factor-C and its receptor VEGFR-3 in non-small-cell lung cancer: concurrent expression in cancer cells from primary tumour and metastatic lymph node.

Lung Cancer 58 , — Yonemura, Y. Lymphangiogenesis and the vascular endothelial growth factor receptor VEGFR -3 in gastric cancer. Cancer 37 , — Su, J. Cancer 96 , — Simiantonaki, N. Google Scholar. Goel, H. VEGF targets the tumour cell. Cancer 13 , — Wang, H.

PLoS ONE 7 , e Manzat Saplacan, R. The role of PDGFs and PDGFRs in colorectal cancer. Mediators Inflamm. Kalra, K. Cell Dev. Balamurugan, K. Cenciarelli, C. PDGFRα depletion attenuates glioblastoma stem cells features by modulation of STAT3, RB1 and multiple oncogenic signals.

Oncotarget 7 , — Chabot, V. Stem Cell Res. Li, H. Development of monoclonal anti-PDGF-CC antibodies as tools for investigating human tissue expression and for blocking PDGF-CC induced PDGFRα signalling in vivo. PLoS ONE 13 , e Dardik, A. Shear stress-stimulated endothelial cells induce smooth muscle cell chemotaxis via platelet-derived growth factor-BB and interleukin-1α.

Muratoglu, S. Low density lipoprotein receptor-related protein 1 LRP1 forms a signaling complex with platelet-derived growth factor receptor-β in endosomes and regulates activation of the MAPK pathway. Wang, J. Metformin inhibits metastatic breast cancer progression and improves chemosensitivity by inducing vessel normalization via PDGF-B downregulation.

Cancer Res. Li, M. Integrins as attractive targets for cancer therapeutics. Acta Pharm. B 11 , — Zou, X. Redundant angiogenic signaling and tumor drug resistance.

Lee, C. Platelet-derived growth factor-C and -D in the cardiovascular system and diseases. Berthod, F. Spontaneous fibroblast-derived pericyte recruitment in a human tissue-engineered angiogenesis model in vitro.

The role of pericytes in angiogenesis. Chatterjee, S. Pericyte-endothelial cell interaction: a survival mechanism for the tumor vasculature.

Cell Adh. Luk, K. Influence of morphine on pericyte-endothelial interaction: implications for antiangiogenic therapy.

Cavalcanti, E. PDGFRα expression as a novel therapeutic marker in well-differentiated neuroendocrine tumors. Cancer Biol. Burger, R. Overview of anti-angiogenic agents in development for ovarian cancer. Raica, M.

Pharmaceuticals 3 , — Heindryckx, F. Targeting the tumor stroma in hepatocellular carcinoma. World J. Cornellà, H. Molecular pathogenesis of hepatocellular carcinoma. Brahmi, M. Expression and prognostic significance of PDGF ligands and receptors across soft tissue sarcomas.

ESMO Open 6 , Rao, L. HB-EGF-EGFR signaling in bone marrow endothelial cells mediates angiogenesis associated with multiple myeloma. Cancers 12 , Hu, L. Dual target inhibitors based on EGFR: promising anticancer agents for the treatment of cancers Larsen, A.

Targeting EGFR and VEGF R pathway cross-talk in tumor survival and angiogenesis. Holbro, T. ErbB receptors: directing key signaling networks throughout life. Ellis, L. Epidermal growth factor receptor in tumor angiogenesis.

North Am. De Luca, A. The role of the EGFR signaling in tumor microenvironment. Albadari, N. The transcriptional factors HIF-1 and HIF-2 and their novel inhibitors in cancer therapy.

Expert Opin. Bos, R. Hypoxia-inducible factor-1α is associated with angiogenesis, and expression of bFGF, PDGF-BB, and EGFR in invasive breast cancer. Histopathology 46 , 31—36 Salomon, D. Epidermal growth factor-related peptides and their receptors in human malignancies.

Yu, H. Poor response to erlotinib in patients with tumors containing baseline EGFR TM mutations found by routine clinical molecular testing.

Raj, S. Cancer 21 , 31 Acevedo, V. Paths of FGFR-driven tumorigenesis. Cell Cycle 8 , — Chen, M. Progress in research on the role of FGF in the formation and treatment of corneal neovascularization.

Montesano, R. Basic fibroblast growth factor induces angiogenesis in vitro. USA 83 , — Giacomini, A. Hui, Q. FGF family: from drug development to clinical application.

Presta, M. Cytokine Growth Factor Rev. Fons, P. Katoh, M. FGF receptors: cancer biology and therapeutics. Kopetz, S. Phase II trial of infusional fluorouracil, irinotecan, and bevacizumab for metastatic colorectal cancer: efficacy and circulating angiogenic biomarkers associated with therapeutic resistance.

Batchelor, T. AZD, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell 11 , 83—95 Cancer genomics and genetics of FGFR2 Review.

Fibroblast growth factor signalling: from development to cancer. Cancer 10 , — Greulich, H. Targeting mutant fibroblast growth factor receptors in cancer. Trends Mol. Cross, M. FGF and VEGF function in angiogenesis: signalling pathways, biological responses and therapeutic inhibition.

Trends Pharmacol. García-Caballero, M. Angioprevention of urologic cancers by plant-derived foods. Pharmaceutics 14 , Aviles, R. Testing clinical therapeutic angiogenesis using basic fibroblast growth factor FGF-2 : Clinical angiogenesis using FGF Vasudev, N.

Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis 17 , — Ding, S. HGF receptor up-regulation contributes to the angiogenic phenotype of human endothelial cells and promotes angiogenesis in vitro. Blood , — Bonnans, C.

Remodelling the extracellular matrix in development and disease. Mulcahy, E. Nakamura, T. The discovery of hepatocyte growth factor HGF and its significance for cell biology, life sciences and clinical medicine.

B: Phys. Bottaro, D. Identification of the hepatocyte growth factor receptor as the c- met proto-oncogene product. Science , — Dean, M. The human met oncogene is related to the tyrosine kinase oncogenes. Ono, K. Circulation 95 , — Cai, W. Mechanisms of hepatocyte growth factor—induced retinal endothelial cell migration and growth.

Ankoma-Sey, V. Coordinated induction of VEGF receptors in mesenchymal cell types during rat hepatic wound healing. Oncogene 17 , — Nagashima, M.

Hepatocyte growth factor HGF , HGF activator, and c-Met in synovial tissues in rheumatoid arthritis and osteoarthritis. Hughes, P. In vitro and in vivo activity of AMG , a potent and selective MET kinase inhibitor, in MET-dependent cancer models.

Leung, E. Oncogene 36 , — Kuang, W. Hartmann, S. Demuth, C. Increased PD-L1 expression in erlotinib-resistant NSCLC cells with MET gene amplification is reversed upon MET-TKI treatment. Oncotarget 8 , — Kwon, M. Frequent hepatocyte growth factor overexpression and low frequency of c-Met gene amplification in human papillomavirus-negative tonsillar squamous cell carcinoma and their prognostic significances.

Miranda, O. Cancers 10 , Wang, Q. MET inhibitors for targeted therapy of EGFR TKI-resistant lung cancer. Wu, J. Prostate 76 , — Imura, Y. Cancer Sci. Birchmeier, C. Met, metastasis, motility and more. Zhang, Y. Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities.

Cancer 17 , 45 Scalia, P. The IGF-II-insulin receptor isoform-A autocrine signal in cancer: actionable perspectives. Bach, L. Endothelial cells and the IGF system.

Clemmons, D. Modifying IGF1 activity: an approach to treat endocrine disorders, atherosclerosis and cancer. van Beijnum, J. Insulin-like growth factor axis targeting in cancer and tumour angiogenesis—the missing link: IGF signaling in tumor angiogenesis.

Chantelau, E. Evidence that upregulation of serum IGF-1 concentration can trigger acceleration of diabetic retinopathy. Wilkinson-Berka, J.

The role of growth hormone, insulin-like growth factor and somatostatin in diabetic retinopathy. Higashi, Y.

Aging, atherosclerosis, and IGF A: Biol. Hellstrom, A. Low IGF-I suppresses VEGF-survival signaling in retinal endothelial cells: direct correlation with clinical retinopathy of prematurity. USA 98 , — Smith, L.

Pathogenesis of retinopathy of prematurity. Acta Paediatr. Moschos, S. The role of the IGF system in cancer: from basic to clinical studies and clinical applications. Oncology 63 , — Sachdev, D. The IGF system and breast cancer.

Samani, A. The role of the IGF system in cancer growth and metastasis: overview and recent insights. Baserga, R. The IGF-1 receptor in cancer biology. Azar, W. IGFBP-2 enhances VEGF gene promoter activity and consequent promotion of angiogenesis by neuroblastoma cells. Endocrinology , — Png, K.

A microRNA regulon that mediates endothelial recruitment and metastasis by cancer cells. Liu, B. Insulin-like growth factor-binding protein-3 inhibition of prostate cancer growth involves suppression of angiogenesis.

Oncogene 26 , — Wu, M. TGF-β superfamily signaling in embryonic development and homeostasis. Cell 16 , — Yang, Y. The role of TGF-β signaling pathways in cancer and its potential as a therapeutic target.

Based Complement Altern. Non-Smad signaling pathways of the TGF-β family. Cold Spring Harb. Santoro, R. TAK-ing aim at chemoresistance: the emerging role of MAP3K7 as a target for cancer therapy. Colak, S. Targeting TGF-β signaling in cancer. Trends Cancer 3 , 56—71 Platten, M. Malignant glioma biology: Role for TGF-β in growth, motility, angiogenesis, and immune escape.

Sabbadini, F. The multifaceted role of TGF-β in gastrointestinal tumors. Massagué, J. TGFβ signalling in context. Horiguchi, K.

Role of Ras signaling in the induction of snail by transforming growth factor-β. Korc, M. Role of growth factors in pancreatic cancer. Nolan-Stevaux, O. GLI1 is regulated through Smoothened-independent mechanisms in neoplastic pancreatic ducts and mediates PDAC cell survival and transformation.

Genes Dev. Budi, E. Enhanced TGF-β signaling contributes to the insulin-induced angiogenic responses of endothelial cells. iScience 11 , — Darland, D. Angiogenesis 4 , 11—20 Huynh, L. A perspective on the development of TGF-β inhibitors for cancer treatment.

Biomolecules 9 , Katz, L. TGF-β signaling in liver and gastrointestinal cancers. Tsubakihara, Y. Epithelial-mesenchymal transition and metastasis under the control of transforming growth factor β.

Fu, M. Multifunctional regulatory protein connective tissue growth factor CTGF : a potential therapeutic target for diverse diseases. B 12 , — Pepper, M. Transforming growth factor-β: vasculogenesis, angiogenesis, and vessel wall integrity. Fang, L.

TGF-β1 induces VEGF expression in human granulosa-lutein cells: a potential mechanism for the pathogenesis of ovarian hyperstimulation syndrome. Oncogenesis 9 , 76 Melisi, D. Modulation of pancreatic cancer chemoresistance by inhibition of TAK1.

Zhang, H. TGFβ signaling in pancreatic ductal adenocarcinoma. Tumor Biol. Modulating TAK1 expression inhibits YAP and TAZ oncogenic functions in pancreatic cancer. Gladilin, E. TGFβ-induced cytoskeletal remodeling mediates elevation of cell stiffness and invasiveness in NSCLC.

Mazzocca, A. Inhibition of transforming growth factor β receptor I kinase blocks hepatocellular carcinoma growth through neo-angiogenesis regulation. Hepatology 50 , — Bhagyaraj, E. TGF-β induced chemoresistance in liver cancer is modulated by xenobiotic nuclear receptor PXR.

Cell Cycle 18 , — Carcinogenesis 39 , — Chiechi, A. Role of TGF-β in breast cancer bone metastases. Masoud, G. HIF-1α pathway: role, regulation and intervention for cancer therapy. B 5 , — HIFs, angiogenesis, and cancer. Konisti, S. Hypoxia—a key regulator of angiogenesis and inflammation in rheumatoid arthritis.

Ahluwalia, A. Critical role of hypoxia sensor—HIF-1 in VEGF gene activation. Implications for angiogenesis and tissue injury healing. Tanaka, T. Angiogenesis and hypoxia in the kidney. Chen, L. Hypoxia and angiogenesis: regulation of hypoxia-inducible factors via novel binding factors. Ikeda, H.

Targeting hypoxia-inducible factor 1 HIF-1 signaling with natural products toward cancer chemotherapy. Pugh, C. Regulation of angiogenesis by hypoxia: role of the HIF system. Semenza, G. Targeting hypoxia-inducible factor 1 to stimulate tissue vascularization.

Vleugel, M. Differential prognostic impact of hypoxia induced and diffuse HIF-1 expression in invasive breast cancer. Dales, J. Overexpression of hypoxia-inducible factor HIF-1α predicts early relapse in breast cancer: retrospective study in a series of patients.

Yatabe, N. HIFmediated activation of telomerase in cervical cancer cells. Oncogene 23 , — Pezzuto, A. A close relationship between HIF-1α expression and bone metastases in advanced NSCLC, a retrospective analysis.

Oncotarget 10 , — Jackson, A. HIF, hypoxia and the role of angiogenesis in non-small cell lung cancer. Targets 14 , — Liu, K. The changes of HIF-1α and VEGF expression after TACE in patients with hepatocellular carcinoma. Zheng, S. Prognostic significance of HIF-1α expression in hepatocellular carcinoma: a meta-analysis.

PLoS ONE 8 , e Targeting HIF-1 for cancer therapy. Schöning, J. Rohwer, N. Hypoxia-mediated drug resistance: Novel insights on the functional interaction of HIFs and cell death pathways.

Zhang, Q. Cell , 37—57 Iosef, C. Inhibiting NF-κB in the developing lung disrupts angiogenesis and alveolarization. Lung Cell. Sakamoto, K.

Copyright: © Li et al. This is an Anti-angiogenesis therapy for solid tumors access Brain health and nutrition distributed under the terms sllid Creative Commons Attribution License. Angiogenesis is a biological process in which novel capillary blood Anti-angiogenesis therapy for solid tumors grow from pre-existing vasculature Sweet potato pizza crustAnti-anguogenesis tissues with oxygen tumora nutrients. As Abti-angiogenesis is correlated with numerous complicated interactions between various thfrapy components, such as several cell types, soluble angiogenic factors and extracellular matrix components, the process of angiogenesis is complex, and primarily consists of four distinct sequential steps: i Degradation of basement membrane glycoproteins and other components of the extracellular matrix surrounding the blood vessels by proteolytic enzymes; ii endothelial cell activation and migration; iii endothelial cell proliferation; and iv endothelial cells transforming into tube-like structures and forming capillary tubes, and developing into novel basement membranes 2. In normal conditions, angiogenesis only occurs during embryonic development, the female reproductive cycle and wound repair 3. However, aberrant angiogenesis is a key mediator and a major process in cancer development.

Anti-angiogenesis therapy for solid tumors -

The principles of anti-angiogenic treatment for solid tumors were originally proposed in , and ever since, it has become a putative target for therapies directed against solid tumors. In the early twenty first century, the FDA approved anti-angiogenic drugs, such as bevacizumab and sorafenib for the treatment of several solid tumors.

Over the past two decades, researches have continued to improve the performance of anti-angiogenic drugs, describe their drug interaction potential, and uncover possible reasons for potential treatment resistance. Herein, we present an update to the pre-clinical and clinical situations of anti-angiogenic agents and discuss the most recent trends in this field.

Keywords: Angiogenesis inhibitors; Natural products; Receptor protein-tyrosine kinase; Tumor microenvironment. GK was a major contributor in revision of the manuscript.

All authors read and approved the final manuscript. Hanahan D and Folkman J: Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Fan TP, Jaggar R and Bicknell R: Controlling the vasculature: Angiogenesis, anti-angiogenesis and vascular targeting of gene therapy.

Trends Pharmacol Sci. Folkman J and Shing Y: Angiogenesis. J Biol Chem. Folkman J: Tumor angiogenesis: Therapeutic implications. N Engl J Med. Folkman J: Anti-angiogenesis: New concept for therapy of solid tumors.

Ann Surg. Dimova I, Popivanov G and Djonov V: Angiogenesis in cancer-general pathways and their therapeutic implications. J BUON. Ribatti D, Nico B, Crivellato E, Roccaro AM and Vacca A: The history of the angiogenic switch concept.

Welti J, Loges S, Dimmeler S and Carmeliet P: Recent molecular discoveries in angiogenesis and antiangiogenic therapies in cancer. J Clin Invest. Ribatti D and Djonov V: Intussusceptive microvascular growth in tumors.

Cancer Lett. Donnem T, Hu J, Ferguson M, Adighibe O, Snell C, Harris AL, Gatter KC and Pezzella F: Vessel co-option in primary human tumors and metastases: An obstacle to effective anti-angiogenic treatment?

Cancer Med. de la Puente P, Muz B, Azab F and Azab AK: Cell trafficking of endothelial progenitor cells in tumor progression. Clin Cancer Res. Moschetta M, Mishima Y, Sahin I, Manier S, Glavey S, Vacca A, Roccaro AM and Ghobrial IM: Role of endothelial progenitor cells in cancer progression.

Biochim Biophys Acta. Seftor RE, Hess AR, Seftor EA, Kirschmann DA, Hardy KM, Margaryan NV and Hendrix MJ: Tumor cell vasculogenic mimicry: From controversy to therapeutic promise. Am J Pathol. Ferrara N and Adamis AP: Ten years of anti-vascular endothelial growth factor therapy.

Nat Rev Drug Discov. Mihicprobst D, Ikenberg K, Tinguely M, Schraml P, Behnke S, Seifert B, Civenni G, Sommer L, Moch H and Dummer R: Tumor cell plasticity and angiogenesis in human melanomas.

PLoS One. Liekens S, Schols D and Hatse S: CXCLCXCR4 axis in angiogenesis, metastasis and stem cell mobilization. Curr Pharm Des. Eelen G, de Zeeuw P, Simons M and Carmeliet P: Endothelial cell metabolism in normal and diseased vasculature.

Circ Res. Bridgeman VL, Vermeulen PB, Foo S, Bilecz A, Daley F, Kostaras E, Nathan MR, Wan E, Frentzas S, Schweiger T, et al: Vessel co-option is common in human lung metastases and mediates resistance to anti-angiogenic therapy in preclinical lung metastasis models.

J Pathol. Frentzas S, Simoneau E, Bridgeman VL, Vermeulen PB, Foo S, Kostaras E, Nathan M, Wotherspoon A, Gao ZH, Shi Y, et al: Vessel co-option mediates resistance to anti-angiogenic therapy in liver metastases. Nat Med. Kerbel RS: Tumor angiogenesis: Past, present and the near future.

Carmeliet P: Developmental biology. Controlling the cellular brakes. Dor Y, Porat R and Keshet E: Vascular endothelial growth factor and vascular adjustments to perturbations in oxygen homeostasis. Am J Physiol Cell Physiol. Littlepage LE, Sternlicht MD, Rougier N, Phillips J, Gallo E, Yu Y, Williams K, Brenot A, Gordon JI and Werb Z: Matrix metalloproteinases contribute distinct roles in neuroendocrine prostate carcinogenesis, metastasis, and angiogenesis progression.

Cancer Res. Carmeliet P, Dor Y, Herbert JM, Fukumura D, Brusselmans K, Dewerchin M, Neeman M, Bono F, Abramovitch R, Maxwell P, et al: Role of HIF-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Maracle CX and Tas SW: Inhibitors of angiogenesis: Ready for prime time?

Best Pract Res Clin Rheumatol. Stem Cells Dev. Jakobsson L, Franco CA, Bentley K, Collins RT, Ponsioen B, Aspalter IM, Rosewell I, Busse M, Thurston G, Medvinsky A, et al: Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting. Nat Cell Biol. Lobov IB, Renard RA, Papadopoulos N, Gale NW, Thurston G, Yancopoulos GD and Wiegand SJ: Delta-like ligand 4 Dll4 is induced by VEGF as a negative regulator of angiogenic sprouting.

Proc Natl Acad Sci USA. Liu Z, Fan F, Wang A, Zheng S and Lu Y: Dll4-Notch signaling in regulation of tumor angiogenesis. J Cancer Res Clin Oncol. Pandya NM, Dhalla NS and Santani DD: Angiogenesis-a new target for future therapy.

Vasc Pharmacol. View Article : Google Scholar. Bergers G and Song S: The role of pericytes in blood-vessel formation and maintenance.

Neuro Oncol. Izzedine H, Ederhy S, Goldwasser F, Soria JC, Milano G, Cohen A, Khayat D and Spano JP: Management of hypertension in angiogenesis inhibitor-treated patients.

Ann Oncol. Liu SX, Xia ZS and Zhong YQ: Genetic therapy in pancreatic cancer. World J Gastroenterol. Edelstein ML, Abedi MR, Wixon J and Edelstein RM: Gene therapy clinical trials worldwide —an overview. J Gene Med. Edelstein ML, Abedi MR and Wixon J: Gene therapy clinical trials worldwide to an update.

Ginn SL, Alexander IE, Edelstein ML, Abedi MR and Wixon J: Gene therapy clinical trials worldwide to an update. Ortiz R, Melguizo C, Prados J, Álvarez PJ, Caba O, Rodríguez-Serrano F, Hita F and Aránega A: New gene therapy strategies for cancer treatment: A review of recent patents.

Recent Pat Anticancer Drug Discov. Cao S, Cripps A and Wei MQ: New strategies for cancer gene therapy: progress and opportunities.

Clin Exp Pharmacol Physiol. Tseng SJ, Liao ZX, Kao SH, Zeng YF, Huang KY, Li HJ, Yang CL, Deng YF, Huang CF, Yang SC, et al: Highly specific in vivo gene delivery for pmediated apoptosis and genetic photodynamic therapies of tumour.

Nat Commun. Gogiraju R, Steinbrecher JH, Lehnart SE, Kessel M, Dobbelstein M and Schaefer K: Importance of tumor suppressor gene pmediated endothelial cell apoptosis for cardiac angiogenesis and hypertrophy.

Eur Heart J. Tazawa H, Kagawa S and Fujiwara T: Advances in adenovirus-mediated p53 cancer gene therapy. Expert Opin Biol Ther. Prabha S, Sharma B and Labhasetwar V: Inhibition of tumor angiogenesis and growth by nanoparticle-mediated p53 gene therapy in mice.

Cancer Gene Ther. Teodoro JG, Evans SK and Green MR: Inhibition of tumor angiogenesis by p A new role for the guardian of the genome. J Mol Med Berl. Zhang C, Wang QT, Liu H, Zhang ZZ and Huang WL: Advancement and prospects of tumor gene therapy. Chin J Cancer. El-Aneed A: An overview of current delivery systems in cancer gene therapy.

J Control Release. Ramsey JD, Vu HN and Pack DW: A top-down approach for construction of hybrid polymer-virus gene delivery vectors. Lundstrom K: Alphavirus vectors as tools in neuroscience and gene therapy. Virus Res. LU Y, Yan M, Chen IS and Liang M: Viral vector nanocapsule for targeting gene therapy and its preparation.

Touchefeu Y, Harrington KJ, Galmiche JP and Vassaux G: Review article: Gene therapy, recent developments and future prospects in gastrointestinal oncology.

Aliment Pharmacol Ther. Liao ZK, Tsai KC, Wang HT, Tseng SH, Deng WP, Chen WS and Hwang LH: Sonoporation-mediated anti-angiogenic gene transfer into muscle effectively regresses distant orthotopic tumors.

Ren J, Zhang P, Tian J, Zhou Z, Liu X, Wang D and Wang Z: A targeted ultrasound contrast agent carrying gene and cell-penetrating peptide: Preparation and gene transfection in vitro.

Colloids Surf B Biointerfaces. Yarmush ML, Golberg A, Serša G, Kotnik T and Miklavčič D: Electroporation-based technologies for medicine: principles, applications, and challenges.

Annu Rev Biomed Eng. Wang W, Li W, Ma N and Steinhoff G: Non-viral gene delivery methods. Curr Pharm Biotechnol. Audouy SA, de Leij LF, Hoekstra D and Molema G: In vivo characteristics of cationic liposomes as delivery vectors for gene therapy. Pharm Res. Hortobagyi GN, Ueno NT, Xia W, Zhang S, Wolf JK, Putnam JB, Weiden PL, Willey JS, Carey M, Branham DL, et al: Cationic liposome-mediated E1A gene transfer to human breast and ovarian cancer cells and its biologic effects: A phase i clinical trial.

J Clin Oncol. Wakabayashi T, Natsume A, Mizuno M, Fujii M, Shimato S and Yoshida J: A clinical trial of cationic liposomes containing interferon-b gene for patients with malignant glioma. Int Conf Brain Tumor Res Ther. Wang Y, Gao S, Ye WH, Yoon HS and Yang YY: Co-delivery of drugs and DNA from cationic core-shell nanoparticles self-assembled from a biodegradable copolymer.

Nat Mater. Power AT and Bell JC: Cell-based delivery of oncolytic viruses: A new strategic alliance for a biological strike against cancer. Mol Ther. Muta M, Matsumoto G, Hiruma K, Nakashima E and Toi M: Study of cancer gene therapy using ILsecreting endothelial progenitor cells in a rat solid tumor model.

Oncol Rep. Yin H, Kanasty RL, Eltoukhy AA, Vegas AJ, Dorkin JR and Anderson DG: Non-viral vectors for gene-based therapy.

Nat Rev Genet. Kim WJ, Yockman JW, Lee M, Jeong JH, Kim YH and Kim SW: Soluble Flt-1 gene delivery using PEI-g-PEG-RGD conjugate for anti-angiogenesis. Persano L, Crescenzi M and Indraccolo S: Anti-angiogenic gene therapy of cancer: Current status and future prospects. Mol Aspects Med. Albini A, Tosetti F, Li VW, Noonan DM and Li WW: Cancer prevention by targeting angiogenesis.

Nat Rev Clin Oncol. Nat Biotechnol. Rodriguez D and Miessner P: Production of AAV vectors for gene therapy: A cost-effectiveness and risk assessment unpublished PhD thesis.

Department of Chemical Engineering and the MIT Sloan School of Management. Chen HH, Kuliszewski MA, Rudenko D and Leong-Poi H: Pre-clinical evaluation of pro-angiogenic gene therapy by ultrasound-targeted microbubble destruction of vascular endothelial growth factor minicircle dna in an model of severe peripheral arterial disease in watanabe heritable hyperlipidemic rabbits.

Can J Cardiol. Feng X: Angiogenesis and Antiangiogenesis Therapies: Spear and Shield of Pharmacotherapy. J Pharma Care Health Sys. Ichihara E, Kiura K and Tanimoto M: Targeting angiogenesis in cancer therapy.

Acta Med Okayama. Trinchieri G: Interleukin A cytokine produced by antigen-presenting cells with immunoregulatory functions in the generation of T-helper cells type 1 and cytotoxic lymphocytes. Trinchieri G: Interleukin A proinflammatory cytokine with immunoregulatory functions that bridge innate resistance and antigen-specific adaptive immunity.

Annu Rev Immunol. Duda DG, Sunamura M, Lozonschi L, Kodama T, Egawa S, Matsumoto G, Shimamura H, Shibuya K, Takeda K and Matsuno S: Direct in vitro evidence and in vivo analysis of the antiangiogenesis effects of interleukin Dias S, Boyd R and Balkwill F: IL regulates VEGF and MMPs in a murine breast cancer model.

Int J Cancer. Voest EE, Kenyon BM, O'Reilly MS, Truitt G, D'Amato RJ and Folkman J: Inhibition of angiogenesis in vivo by interleukin J Natl Cancer Inst.

Int J Oncol. Sunamura M, Sun L, Lozonschi L, Duda DG, Kodama T, Matsumoto G, Shimamura H, Takeda K, Kobari M, Hamada H and Matsuno S: The antiangiogenesis effect of interleukin 12 during early growth of human pancreatic cancer in SCID mice. Li Q, Zhihua W, Xiumin Y, et al: The effect of il on the proliferation in vitro and anti-tumor effects of cik cells in vivo and in vitro.

J Pract Oncol. In Chinese. Nguyen K, Koppolu B, Smith G, Ravindranathan S and Zaharoff D: Interleukin elicits various responses of splenocytes from different mouse strains.

J Immunol. Portielje JE, Kruit WH, Schuler M, Beck J, Lamers CH, Stoter G, Huber C, de Boer-Dennert M, Rakhit A, Bolhuis RL and Aulitzky WE: Phase I study of subcutaneously administered recombinant human interleukin 12 in patients with advanced renal cell cancer.

Gollob JA, Mier JW, Veenstra K, McDermott DF, Clancy D, Clancy M and Atkins MB: Phase I trial of twice-weekly intravenous interleukin 12 in patients with metastatic renal cell cancer or malignant melanoma: Ability to maintain IFN-gamma induction is associated with clinical response.

Hurteau JA, Blessing JA, DeCesare SL and Creasman WT: Evaluation of recombinant human interleukin in patients with recurrent or refractory ovarian cancer: A gynecologic oncology group study. Gynecol Oncol. Daud A, Takamura KT, Diep T, Heller R and Pierce RH: Long-term overall survival from a phase I trial using intratumoral plasmid interleukin with electroporation in patients with melanoma.

J Transl Med. Daud A, Algazi A, Ashworth M, Buljan M, Takamura KT, Diep T, Pierce RH and Bhatia S: Intratumoral electroporation of plasmid interleukin Efficacy and biomarker analyses from a phase 2 study in melanoma OMS-I Cutrera J, King G, Jones P, Kicenuik K, Gumpel E, Xia X and Li S: Safety and Efficacy of Tumor-Targeted Interleukin 12 Gene Therapy in Treated and Non-Treated, Metastatic Lesions.

Curr Gene Ther. Lampreht U, Kamensek U, Stimac M, et al: Gene electrotransfer of canine interleukin 12 into canine melanoma cell lines. J Membr Biol. Markert JM, Cody JJ, Parker JN, Coleman JM, Price KH, Kern ER, Quenelle DC, Lakeman AD, Schoeb TR, Palmer CA, et al: Preclinical evaluation of a genetically engineered herpes simplex virus expressing interleukin J Virol.

Kramer MG, Masner M, Casales E, Moreno M, Smerdou C and Chabalgoity JA: Neoadjuvant administration of Semliki Forest virus expressing interleukin combined with attenuated Salmonella eradicates breast cancer metastasis and achieves long-term survival in immunocompetent mice.

BMC Cancer. Koneru M, O'Cearbhaill R, Pendharkar S, Spriggs DR and Brentjens RJ: A phase I clinical trial of adoptive T cell therapy using IL secreting MUC ecto directed chimeric antigen receptors for recurrent ovarian cance.

Poutou J, Bunuales M, Gonzalez-Aparicio M, Garcia-Aragoncillo E, Quetglas JI, Casado R, Bravo-Perez C, Alzuguren P and Hernandez-Alcoceba R: Safety and antitumor effect of oncolytic and helper-dependent adenoviruses expressing interleukin variants in a hamster pancreatic cancer model.

Gene Ther. Oncol Lett. Freytag SO, Zhang Y and Siddiqui F: Preclinical toxicology of oncolytic adenovirus-mediated cytotoxic and interleukin gene therapy for prostate cancer. Mol Ther Oncolytics. Cutrera J, King G, Jones P, Kicenuik K, Gumpel E, Xia X and Li S: Safe and effective treatment of spontaneous neoplasms with interleukin 12 electro-chemo-gene therapy.

J Cell Mol Med. Jiang H, Lin JJ, Su Z, Goldstein N and Fisher P: Subtraction hybridization identifies a novel melanoma differentiation associated gene, mda-7, modulated during human melanoma differentiation, growth and progression. Jiang H, Su ZZ, Lin JJ, Goldstein NI, Young CS and Fisher PB: The melanoma differentiation associated gene mda-7 suppresses cancer cell growth.

Su ZZ, Madireddi MT, Lin JJ, Young CS, Kitada S, Reed JC, Goldstein NI and Fisher PB: The cancer growth suppressor gene mda-7 selectively induces apoptosis in human breast cancer cells and inhibits tumor growth in nude mice. Saeki T, Mhashilkar A, Swanson X, Zou-Yang XH, Sieger K, Kawabe S, Branch CD, Zumstein L, Meyn RE, Roth JA, et al: Inhibition of human lung cancer growth following adenovirus-mediated mda-7 gene expression in vivo.

J Cell Physiol. Chen X, Liu D, Wang J, Su Q, Zhou P, Liu J, Luan M and Xu X: Suppression effect of recombinant adenovirus vector containing hIL on Hep-2 laryngeal carcinoma cells. Mol Med Rep. Khodadad M, Hosseini SY, Shenavar F, Erfani N, Bina S, Ahmadian S, Fattahi MR and Hajhosseini R: Construction of expressing vectors including melanoma differentiation-associated gene-7 mda-7 fused with the RGD sequences for better tumor targeting.

Iran J Basic Med Sci. O'Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal RA, Moses M, Lane WS, Cao Y, Sage EH and Folkman J: Angiostatin: A novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma.

Wahl ML, Moser TL and Pizzo SV: Angiostatin and anti-angiogenic therapy in human disease. Recent Prog Horm Res. Zhang G, Jin G, Nie X, Mi R, Zhu G, Jia W and Liu F: Enhanced antitumor efficacy of an oncolytic herpes simplex virus expressing an endostatin-angiostatin fusion gene in human glioblastoma stem cell xenografts.

Zhu G, Su W, Jin G, Xu F, Hao S, Guan F, Jia W and Liu F: Glioma stem cells targeted by oncolytic virus carrying endostatin-angiostatin fusion gene and the expression of its exogenous gene in vitro.

Brain Res. Tysome JR, Wang P, Alusi G, Briat A, Gangeswaran R, Wang J, Bhakta V, Fodor I, Lemoine NR and Wang Y: Lister vaccine strain of vaccinia virus armed with the endostatin-angiostatin fusion gene: An oncolytic virus superior to dl ONYX for human head and neck cancer.

Hum Gene Ther. Hutzen B, Bid HK, Houghton PJ, Pierson CR, Powell K, Bratasz A, Raffel C and Studebaker AW: Treatment of medulloblastoma with oncolytic measles viruses expressing the angiogenesis inhibitors endostatin and angiostatin.

Li X, Liu YH, Lee SJ, Gardner TA, Jeng MH and Kao C: Prostate-restricted replicative adenovirus expressing human endostatin-angiostatin fusion gene exhibiting dramatic antitumor efficacy.

Ma HI, Lin SZ, Chiang YH, Li J, Chen SL, Tsao YP and Xiao X: Intratumoral gene therapy of malignant brain tumor in a rat model with angiostatin delivered by adeno-associated viral AAV vector.

Li R, Chen H and Ren CS: Growth inhibition of breast cancer in rat by AAV mediated angiostatin gene. Chin J Cancer Res. Kubo S, Takagi-Kimura M and Kasahara N: Combinatorial anti-angiogenic gene therapy in a human malignant mesothelioma model. Tan JF, Lu Q, Zhang XW and Tan M: Effect of co-transfection of angiostatin and Fas gene on growth of transplanted tumor in nude mice.

China J Mod Med. Kim HS, Jeong HY, Lee YK, Kim KS and Park YS: Synergistic antitumoral effect of IL gene cotransfected with antiangiogenic genes for Angiostatin, Endostatin, and Saxatilin. Oncol Res Featuring Preclinical Clin Cancer Ther. Sun X, Vale M, Jiang X, Gupta R and Krissansen G: Antisense HIF-1alpha prevents acquired tumor resistance to angiostatin gene therapy.

Chen XJ, Zhu YY, Hu ZT, Zhang HH, Weng SM and Zhuang HZ: Effect of co-transfection of p53 and angiostatin gene on the apoptosis of gastric cancer SG cells. Schmitz V, Tirado-Ledo L, Raskopf E, Rabe C, Wernert N, Wang L, Prieto J, Qian C, Sauerbruch T and Caselmann WH: Effective antitumour mono- and combination therapy by gene delivery of angiostatin-like molecule and interleukin in a murine hepatoma model.

Int J Colorectal Dis. Chu Y, Liu H, Lou G, Zhang Q and Wu C: Human placenta mesenchymal stem cells expressing exogenous kringle protein by fiber-modified adenovirus suppress angiogenesis.

Schmitz V, Sauerbruch T and Raskopf E: Anti-tumoural effects of PlgK are directly linked to reduced ICAM expression, resulting in hepatoma cell apoptosis. O'Reilly MS, Boehm T, Shing Y, Fukai N, Vasios G, Lane WS, Flynn E, Birkhead JR, Olsen BR and Folkman J: Endostatin: An endogenous inhibitor of angiogenesis and tumor growth.

Sasaki T, Fukai N, Mann K, Göhring W, Olsen BR and Timpl R: Structure, function and tissue forms of the C-terminal globular domain of collagen XVIII containing the angiogenesis inhibitor endostatin. EMBO J. Rong B, Yang S, Li W, Zhang W and Ming Z: Systematic review and meta-analysis of Endostar rh-endostatin combined with chemotherapy versus chemotherapy alone for treating advanced non-small cell lung cancer.

World J Surg Oncol. Huiqi G, Jing Z, Peng F, Yong L and Baozhong S: In vivo study of the effect of combining endostatin gene therapy with 32P-colloid on hepatocarcinoma and its functioning mechanism. Yan F, Zheng Y and Huang L: Adenovirus-mediated combined anti-angiogenic and pro-apoptotic gene therapy enhances antitumor efficacy in hepatocellular carcinoma.

Xu Y, Xie Z, Zhou Y, Zhou X, Li P, Wang Z and Zhang Q: Experimental endostatin-GFP gene transfection into human retinal vascular endothelial cells using ultrasound-targeted cationic microbubble destruction. Mol Vis. Li XP, Zhang HL, Wang HJ, Li YX, Li M, Lu L, Wan Y, Zhou BL, Liu Y, Pan Y, et al: Ad-endostatin treatment combined with low-dose irradiation in a murine lung cancer model.

Liu RY, Zhou L, Zhang YL, Huang BJ, Ke ML, Chen JM, Li LX, Fu X, Wu JX and Huang W: An oncolytic adenovirus enhances antiangiogenic and antitumoral effects of a replication-deficient adenovirus encoding endostatin by rescuing its selective replication in nasopharyngeal carcinoma cells.

Biochem Biophys Res Commun. Li L, Zhang Y, Zhou L, Ke ML, Chen JM, Fu X, Ye CL, Wu JX, Liu RY and Huang W: Antitumor efficacy of a recombinant adenovirus encoding endostatin combined with an E1B55KD-deficient adenovirus in gastric cancer cells.

Zhou Y, Gu H, Xu Y, Li F, Kuang S, Wang Z, Zhou X, Ma H, Li P, Zheng Y, et al: Targeted antiangiogenesis gene therapy using targeted cationic microbubbles conjugated with CD antibody compared with untargeted cationic and neutral microbubbles.

Maeshima Y, Colorado PC, Torre A, Holthaus KA, Grunkemeyer JA, Ericksen MB, Hopfer H, Xiao Y, Stillman IE and Kalluri R: Distinct antitumor properties of a type IV collagen domain derived from basement membrane.

Yang YP, Xu CX, Hou GS, Xin JX, Wang W and Liu XX: Effects of eukaryotic expression plasmid encoding human tumstatin gene on endothelial cells in vitro. Chin Med J Engl. Borza CM, Pozzi A, Borza DB, Pedchenko V, Hellmark T, Hudson BG and Zent R: Integrin alpha3beta1, a novel receptor for alpha3 IV noncollagenous domain and a trans-dominant Inhibitor for integrin alphavbeta3.

Hwang-Bo J, Park JH and Chung IS: Tumstatin induces apoptosis mediated by Fas signaling pathway in oral squamous cell carcinoma SCC-VII cells. Goto T, Ishikawa H, Matsumoto K, Nishimura D, Kusaba M, Taura N, Shibata H, Miyaaki H, Ichikawa T, Hamasaki K, et al: Tum-1, a tumstatin fragment, gene delivery into hepatocellular carcinoma suppresses tumor growth through inhibiting angiogenesis.

You Y, Xue X, Li M, Qin X, Zhang C, Wang W, Giang C, Wu S, Liu Y, Zhu W, et al: Inhibition effect of pcDNA-tum-5 on the growth of S tumor. Caudroy S, Cucherousset J, Lorenzato M, Zahm JM, Martinella-Catusse C, Polette M and Birembaut P: Implication of tumstatin in tumor progression of human bronchopulmonary carcinomas.

Hum Pathol. Zhang X, Xu W, Qian H, Zhu W and Zhang R: Mesenchymal stem cells modified to express lentivirus TNF-α Tumstatin 45— inhibit the growth of prostate cancer. Thevenard J, Ramont L, Mir LM, Dupont-Deshorgue A, Maquart FX, Monboisse JC and Brassart-Pasco S: A new anti-tumor strategy based on in vivo tumstatin overexpression after plasmid electrotransfer in muscle.

Gu Q, Sun C, Luo J, Zhang T and Wang L: Inhibition of angiogenesis by a synthetic fusion protein VTF derived from vasostatin and tumstatin. Anticancer Drugs. Zhang X, Qi DD, Zhang TT, Chen QX, Wang GZ, Sui GY, Hao XW, Sun S, Song X and Chen YL: Antitumor activity of adenoviral vector containing T42 and 4xT42 peptide gene through inducing apoptosis of tumor cells and suppressing angiogenesis.

Trochon-Joseph V, Martel-Renoir D, Mir LM, Thomaïdis A, Opolon P, Connault E, Li H, Grenet C, Fauvel-Lafève F, Soria J, et al: Evidence of antiangiogenic and antimetastatic activities of the recombinant disintegrin domain of metargidin. Nath D, Slocombe PM, Stephens PE, Warn A, Hutchinson GR, Yamada KM, Docherty AJ and Murphy G: Interaction of metargidin ADAM with alphavbeta3 and alpha5beta1 integrins on different haemopoietic cells.

J Cell Sci. Danhier F, Le Breton A and Préat V: RGD-based strategies to target alpha v beta 3 integrin in cancer therapy and diagnosis. Mol Pharm. Daugimont L, Vandermeulen G, Defresne F, Bouzin C, Mir LM, Bouquet C, Feron O and Préat V: Antitumoral and antimetastatic effect of antiangiogenic plasmids in B16 melanoma: Higher efficiency of the recombinant disintegrin domain of ADAM Eur J Pharm Biopharm.

Spanggaard I, Snoj M, Cavalcanti A, Bouquet C, Sersa G, Robert C, Cemazar M, Dam E, Vasseur B, Attali P, et al: Gene electrotransfer of plasmid antiangiogenic metargidin peptide AMEP in disseminated melanoma: Safety and efficacy results of a phase I first-in-man study.

Hum Gene Ther Clin Dev. Bosnjak M, Prosen L, Dolinsek T, Blagus T, Markelc B, Cemazar M, Bouquet C and Sersa G: Biological properties of melanoma and endothelial cells after plasmid AMEP gene electrotransfer depend on integrin quantity on cells.

Bosnjak M, Dolinsek T, Cemazar M, Kranjc S, Blagus T, Markelc B, Stimac M, Zavrsnik J, Kamensek U, Heller L, et al: Gene electrotransfer of plasmid AMEP, an integrin-targeted therapy, has antitumor and antiangiogenic action in murine B16 melanoma.

FEBS Lett. Kubota T, Matsumura A, Taiyoh H, Izumiya Y, Fujiwara H, Okamoto K, Ichikawa D, Shiozaki A, Komatsu S, Nakanishi M, et al: Interruption of the HGF paracrine loop by NK4, an HGF antagonist, reduces VEGF expression of CT26 cells.

Kishi Y, Kuba K and Nakamura T, Wen J, Suzuki Y, Mizuno S, Nukiwa T, Matsumoto K and Nakamura T: Systemic NK4 gene therapy inhibits tumor growth and metastasis of melanoma and lung carcinoma in syngeneic mouse tumor models. Cancer Sci. Ogura Y, Mizumoto K, Nagai E, Murakami M, Inadome N, Saimura M, Matsumoto K, Nakamura T, Maemondo M, Nukiwa T and Tanaka M: Peritumoral injection of adenovirus vector expressing NK4 combined with gemcitabine treatment suppresses growth and metastasis of human pancreatic cancer cells implanted orthotopically in nude mice and prolongs survival.

Nakamura T, Sakai K, Nakamura T and Matsumoto K: Anti-cancer approach with NK4: Bivalent action and mechanisms. Anticancer Agents Med Chem. Matsumoto K and Nakamura T: Mechanisms and significance of bifunctional NK4 in cancer treatment. Matsumoto G, Omi Y, Lee U, Kubota E and Tabata Y: NK4 gene therapy combined with cisplatin inhibits tumour growth and metastasis of squamous cell carcinoma.

Anticancer Res. Taiyoh H, Kubota T, Fujiwara H, Matsumura A, Murayama Y, Okamoto K, Ichikawa D, Ochiai T, Nakamura T, Matsumoto K, et al: NK4 gene expression enhances 5-fluorouracil-induced apoptosis of murine colon cancer cells. Zhu Y, Cheng M, Yang Z, Zeng CY, Chen J, Xie Y, Luo SW, Zhang KH, Zhou SF and Lu NH: Mesenchymal stem cell-based NK4 gene therapy in nude mice bearing gastric cancer xenografts.

Drug Des Devel Ther. Ten Dijke P, Goumans MJ and Pardali E: Endoglin in angiogenesis and vascular diseases. Nassiri F, Cusimano MD, Scheithauer BW, Rotondo F, Fazio A, Yousef GM, Syro LV, Kovacs K and Lloyd RV: Endoglin CD : A review of its role in angiogenesis and tumor diagnosis, progression and therapy.

Tsujie M, Tsujie T, Toi H, Uneda S, Shiozaki K, Tsai H and Seon BK: Anti-tumor activity of an anti-endoglin monoclonal antibody is enhanced in immunocompetent mice.

Uneda S, Toi H, Tsujie T, Tsujie M, Harada N, Tsai H and Seon BK: Anti-endoglin monoclonal antibodies are effective for suppressing metastasis and the primary tumors by targeting tumor vasculature.

Muñoz R, Arias Y, Ferreras JM, Jiménez P, Langa C, Rojo MA, Gayoso MJ, Córdoba-Díaz D, Bernabéu C and Girbés T: In vitro and in vivo effects of an anti-mouse endoglin CD -immunotoxin on the early stages of mouse B16MEL4A5 melanoma tumours.

Cancer Immunol Immunother. Tabata M, Kondo M, Haruta Y and Seon BK: Antiangiogenic radioimmunotherapy of human solid tumors in SCID mice using I-labeled anti-endoglin monoclonal antibodies. Dolinsek T, Markelc B, Sersa G, Coer A, Stimac M, Lavrencak J, Brozic A, Kranjc S and Cemazar M: Multiple delivery of siRNA against endoglin into murine mammary adenocarcinoma prevents angiogenesis and delays tumor growth.

Dolinsek T, Markelc B, Bosnjak M, Blagus T, Prosen L, Kranjc S, Stimac M, Lampreht U, Sersa G and Cemazar M: Endoglin silencing has significant antitumor effect on murine mammary adenocarcinoma mediated by vascular targeted effect. Xu Y, Hou J, Liu Z, Yu H, Sun W, Xiong J, Liao Z, Zhou F, Xie C and Zhou Y: Gene therapy with tumor-specific promoter mediated suicide gene plus IL gene enhanced tumor inhibition and prolonged host survival in a murine model of Lewis lung carcinoma.

Dolinsek T, Sersa G and Cemazar M: Melanoma cell viability is reduced after endoglin silencing with gene electrotransfer. Biol Med Food Environ Technol. Pujade-Lauraine E, Hilpert F, Weber B, et al: Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: the AURELIA open-label randomized phase III trial.

Heinemann V, von Weikersthal LF, Decker T, Kiani A, Vehling-Kaiser U, Al-Batran SE, Heintges T, Lerchenmüller C, Kahl C, Seipelt G, et al: FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer FIRE-3 : A randomised, open-label, phase 3 trial.

Lancet Oncol. Rini BI, Bellmunt J, Clancy J, Wang K, Niethammer AG, Hariharan S and Escudier B: Randomized phase III trial of temsirolimus and bevacizumab versus interferon alfa and bevacizumab in metastatic renal cell carcinoma: INTORACT trial.

Bear HD, Tang G, Rastogi P, Geyer CE Jr, Liu Q, Robidoux A, Baez-Diaz L, Brufsky AM, Mehta RS, Fehrenbacher L, et al: Neoadjuvant plus adjuvant bevacizumab in early breast cancer NSABP B [NRG Oncology] : Secondary outcomes of a phase 3, randomised controlled trial.

Van Cutsem E, Tabernero J, Lakomy R, Prenen H, Prausová J, Macarulla T, Ruff P, van Hazel GA, Moiseyenko V, Ferry D, et al: Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen.

Tannock IF, Fizazi K, Ivanov S, Karlsson CT, Fléchon A, Skoneczna I, Orlandi F, Gravis G, Matveev V, Bavbek S, et al: Aflibercept versus placebo in combination with docetaxel and prednisone for treatment of men with metastatic castration-resistant prostate cancer VENICE : A phase 3, double-blind randomised trial.

Ramlau R, Gorbunova V, Ciuleanu TE, Novello S, Ozguroglu M, Goksel T, Baldotto C, Bennouna J, Shepherd FA, Le-Guennec S, et al: Aflibercept and docetaxel versus docetaxel alone after platinum failure in patients with advanced or metastatic non-small-cell lung cancer: A randomized, controlled phase III trial.

Allen JW, Moon J, Redman M, Gadgeel SM, Kelly K, Mack PC, Saba HM, Mohamed MK, Jahanzeb M and Gandara DR: Southwest oncology group S A randomized, phase II trial of weekly topotecan with and without ziv-aflibercept in patients with platinum-treated small-cell lung cancer.

Siu LL, Shapiro JD, Jonker DJ, Karapetis CS, Zalcberg JR, Simes J, Couture F, Moore MJ, Price TJ, Siddiqui J, et al: Phase III randomized, placebo-controlled study of cetuximab plus brivanib alaninate versus cetuximab plus placebo in patients with metastatic, chemotherapy-refractory, wild-type K-RAS colorectal carcinoma: The NCIC clinical trials group and AGITG CO.

Lordick F, Kang YK, Chung HC, Salman P, Oh SC, Bodoky G, Kurteva G, Volovat C, Moiseyenko VM, Gorbunova V, et al: Capecitabine and cisplatin with or without cetuximab for patients with previously untreated advanced gastric cancer EXPAND : A randomised, open-label phase 3 trial. Hitre E, Budai B, Takácsi-Nagy Z, Rubovszky G, Tóth E, Remenár É, Polgár C and Láng I: Cetuximab and platinum-based chemoradio- or chemotherapy of patients with epidermal growth factor receptor expressing adenoid cystic carcinoma: A phase II trial.

Br J Cancer. Massarelli E, Haddad RI, Lee JJ, Garden AS, Blumenschein GR, William WN, Tisshler RB, Glisson BS, Gold KA, Johnson FM, et al: Randomized phase II trial of weekly paclitaxel, carboplatin, cetuximab PCC versus cetuximab, docetaxel, cisplatin, and fluorouracil C-TPF in previously untreated patients with locally advanced head and neck squamous cell carcinoma.

Wang J, Sun Y and Qin S: Endostar Phase IV Study Group: Results of phase IV clinical trial of combining endostar with chemotherapy for treatment of advanced non-small cell lung cancer NSCLC.

Cui C, Mao L, Chi Z, Si L, Sheng X, Kong Y, Li S, Lian B, Gu K, Tao M, et al: A phase II, randomized, double-blind, placebo-controlled multicenter trial of Endostar in patients with metastatic melanoma. Jin T, Li B and Chen XZ: AA phase II trial of Endostar combined with gemcitabine and cisplatin chemotherapy in patients with metastatic nasopharyngeal carcinoma NCT Oncol Res.

Chen Z, Guo W, Cao J, Lv F, Zhang W, Qiu L, Li W, Ji D, Zhang S, Xia Z, et al: Endostar in combination with modified FOLFOX6 as an initial therapy in advanced colorectal cancer patients: A phase I clinical trial.

Cancer Chemother Pharmacol. Zhu AX, Rosmorduc O, Evans TJ, Ross PJ, Santoro A, Carrilho FJ, Bruix J, Qin S, Thuluvath PJ, Llovet JM, et al: SEARCH: A phase III, randomized, double-blind, placebo-controlled trial of sorafenib plus erlotinib in patients with advanced hepatocellular carcinoma.

Brose MS, Nutting CM, Jarzab B, Elisei R, Siena S, Bastholt L, de la Fouchardiere C, Pacini F, Paschke R, Shong YK, et al: Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: A randomised, double-blind, phase 3 trial trial.

Bruix J, Takayama T, Mazzaferro V, Chau GY, Yang J, Kudo M, Cai J, Poon RT, Han KH, Tak WY, et al: STORM: A phase III randomized, double-blind, placebo-controlled trial of adjuvant sorafenib after resection or ablation to prevent recurrence of hepatocellular carcinoma HCC.

Invest New Drugs. Motzer RJ, Hutson TE, Tomczak P, Tomczak P, Michaelson M, Bukowski RM, Rixe O, Oudard S, Kim ST, Baum CM and Figlin RA: Phase III randomized trial of sunitinib malate SU versus interferon-alfa IFN-{alpha} as first-line systemic therapy for patients with metastatic renal cell carcinoma mRCC.

Socinski MA, Novello S, Brahmer JR, Rosell R, Sanchez JM, Belani CP, Govindan R, Atkins JN, Gillenwater HH, Pallares C, et al: Multicenter, phase II trial of sunitinib in previously treated, advanced non-small-cell lung cance.

Cheng A, Kang Y, Lin D, Park J, Kudo M, Qin S, Omata M, Lowenthal SWP, Lanzalone S, Yang L, et al: Phase III trial of sunitinib Su versus sorafenib So in advanced hepatocellular carcinoma HCC. Michaelson MD, Oudard S, Ou YC, Sengeløv L, Saad F, Houede N, Ostler P, Stenzl A, Daugaard G, Jones R, et al: Randomized, placebo-controlled, phase III trial of sunitinib plus prednisone versus prednisone alone in progressive, metastatic, castration-resistant prostate cancer.

Kantarjian HM, Shah NP, Cortes JE, Baccarani M, Agarwal MB, Undurraga MS, Wang J, Ipiña JJ, Kim DW, Ogura M, et al: Dasatinib or imatinib in newly diagnosed chronic-phase chronic myeloid leukemia: 2-year follow-up from a randomized phase 3 trial DASISION.

Kluger HM, Dudek AZ, McCann C, Ritacco J, Southard N, Jilaveanu LB, Molinaro A and Sznol M: A phase 2 trial of dasatinib in advanced melanoma. Wong SJ, Karrison T, Hayes DN, Kies MS, Cullen KJ, Tanvetyanon T, Argiris A, Takebe N, Lim D, Saba NF, et al: Phase II trial of dasatinib for recurrent or metastatic c-KIT expressing adenoid cystic carcinoma and for nonadenoid cystic malignant salivary tumors.

Herbst RS, Giaccone G, Schiller JH, Natale RB, Miller V, Manegold C, Scagliotti G, Rosell R, Oliff I, Reeves JA, et al: Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: a phase III trial-INTACT 2. Argiris A, Ghebremichael M, Gilbert J, Lee JW, Sachidanandam K, Kolesar JM, Burtness B and Forastiere AA: Phase III randomized, placebo-controlled trial of docetaxel with or without gefitinib in recurrent or metastatic head and neck cancer: An eastern cooperative oncology group trial.

Dutton SJ, Ferry DR, Blazeby JM, Abbas H, Dahle-Smith A, Mansoor W, Thompson J, Harrison M, Chatterjee A, Falk S, et al: Gefitinib for oesophageal cancer progressing after chemotherapy COG : A phase 3, multicentre, double-blind, placebo-controlled randomised tria.

Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, et al: Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: A phase III trial of the national cancer institute of canada clinical trials group.

Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, Palmero R, Garcia-Gomez R, Pallares C, Sanchez JM, et al: Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer EURTAC : A multicentre, open-label, randomised phase 3 trial.

Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, Barrios CH, Salman P, Gladkov OA, Kavina A, et al: Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial.

van der Graaf WT, Blay JY, Chawla SP, Kim DW, Bui-Nguyen B, Casali PG, Schöffski P, Aglietta M, Staddon AP, Beppu Y, et al: Pazopanib for metastatic soft-tissue sarcoma PALETTE : A randomised, double-blind, placebo-controlled phase 3 trial.

July Volume 16 Issue 1.

Thank you for visiting nature. You Anri-angiogenesis using Anti-angiogenesis therapy for solid tumors browser version Anti-angiiogenesis limited support for CSS. To obtain the tjmors experience, Anti-angiogenesis therapy for solid tumors recommend you use a more Intense herbal beverage to date browser or thefapy off compatibility mode in Internet Explorer. In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript. Angiogenesis, the formation of new blood vessels, is a complex and dynamic process regulated by various pro- and anti-angiogenic molecules, which plays a crucial role in tumor growth, invasion, and metastasis. With the advances in molecular and cellular biology, various biomolecules such as growth factors, chemokines, and adhesion factors involved in tumor angiogenesis has gradually been elucidated.

Author: Malrajas

2 thoughts on “Anti-angiogenesis therapy for solid tumors

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com