Category: Moms

Anti-angiogenesis clinical trials

Anti-angiogenesis clinical trials

Chandra A, Anti-angiogenesis clinical trials Vlinical, Yagnik G, Aghi MK. Drugs Anti-zngiogenesis works in Anti-angiofenesis way Anti-angiogenesis clinical trials Bone fractures and prevention and lenalidomide Revlimid. Many physiological, cellular, and molecular clijical candidates clinixal to anti-angiogenic therapy-induced Anti-angiogenesis clinical trials effects have been proposed, but in clinical practice physiological responses are the most commonly used biomarkers. Proc Am Soc Clin Oncol 21 : A Google Scholar Reyes M, Dudek A, Jahagirdar B, Koodie L, Marker PH, Verfaillie CM Origin of endothelial progenitors in human postnatal bone marrow. Anti-angiogenic drugs in cancer therapeutics: a review of the latest preclinical and clinical studies of anti-angiogenic agents with anticancer potential Download PDF. Therefore, angiogenesis inhibitors can cause a wide range of physical side effects including:. Akbari M, Tamtaji OR, Lankarani KB, et al.

Anti-angiogenesis clinical trials -

Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cao Y, Xue L. Semin Thromb Hemost. Cao Y. Endogenous angiogenesis inhibitors and their therapeutic implications.

Int J Biochem Cell Biol. Folberg R, Hendrix MJ, Maniotis AJ. Vasculogenic mimicry and tumor angiogenesis. Am J Pathol. Article PubMed Central CAS PubMed Google Scholar. Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry.

Bissell MJ. Tumor plasticity allows vasculogenic mimicry, a novel form of angiogenic switch. A rose by any other name? Ribatti D, Vacca A, Dammacco F. New non-angiogenesis dependent pathways for tumour growth. Eur J Cancer. Leenders WP, Kusters B, de Waal RM. Vessel co-option: how tumors obtain blood supply in the absence of sprouting angiogenesis.

Dome B, Paku S, Somlai B, Timar J. Vascularization of cutaneous melanoma involves vessel co-option and has clinical significance. J Pathol. Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, et al. A phase 3 trial of bevacizumab in ovarian cancer.

Cataldo VD, Gibbons DL, Perez-Soler R, Quintas-Cardama A. Treatment of non-small-cell lung cancer with erlotinib or gefitinib. Haines IE, Miklos GL. Paclitaxel plus bevacizumab for metastatic breast cancer. Miller K, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, et al.

Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al.

Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. VEGF-targeted cancer therapeutics-paradoxical effects in endocrine organs. Nat Rev Endocrinol. Off-tumor target—beneficial site for antiangiogenic cancer therapy? Nat Rev Clin Oncol. Fidler IJ.

Cancer metastasis. Br Med Bull. CAS PubMed Google Scholar. Lee SL, Rouhi P, Dahl Jensen L, Zhang D, Ji H, Hauptmann G, et al. Hypoxia-induced pathological angiogenesis mediates tumor cell dissemination, invasion, and metastasis in a zebrafish tumor model.

Proc Natl Acad Sci USA. Rouhi P, Jensen LD, Cao Z, Hosaka K, Lanne T, Wahlberg E, et al. Hypoxia-induced metastasis model in embryonic zebrafish. Nat Protoc. Opinion: emerging mechanisms of tumour lymphangiogenesis and lymphatic metastasis.

Nat Rev Cancer. Argiles JM, Busquets S, Stemmler B, Lopez-Soriano FJ. Cancer cachexia: understanding the molecular basis. Whelan AJ, Bartsch D, Goodfellow PJ. Brief report: a familial syndrome of pancreatic cancer and melanoma with a mutation in the CDKN2 tumor-suppressor gene.

Argiles JM, Busquets S, Lopez-Soriano FJ. Anti-inflammatory therapies in cancer cachexia. Eur J Pharmacol. Oliff A, Defeo-Jones D, Boyer M, Martinez D, Kiefer D, Vuocolo G, et al.

Zhou X, Wang JL, Lu J, Song Y, Kwak KS, Jiao Q, et al. Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival.

Todorov P, Cariuk P, McDevitt T, Coles B, Fearon K, Tisdale M. Characterization of a cancer cachectic factor. Tannock IF, Fizazi K, Ivanov S, Karlsson CT, Flechon A, Skoneczna I, 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. Garon EB, Ciuleanu TE, Arrieta O, Prabhash K, Syrigos KN, Goksel T, et al. Ramucirumab plus docetaxel versus placebo plus docetaxel for second-line treatment of stage IV non-small-cell lung cancer after disease progression on platinum-based therapy REVEL : a multicentre, double-blind, randomised phase 3 trial.

Positive and negative modulation of angiogenesis by VEGFR1 ligands. Sci Signal. Cao Y, Zhong W, Sun Y. Improvement of antiangiogenic cancer therapy by understanding the mechanisms of angiogenic factor interplay and drug resistance.

Semin Cancer Biol. Tumor angiogenesis and molecular targets for therapy. Front Biosci Landmark Ed. Article CAS Google Scholar. Motzer RJ, Hutson TE, McCann L, Deen K, Choueiri TK.

Overall survival in renal-cell carcinoma with pazopanib versus sunitinib. Sitohy B, Nagy JA, Jaminet SC, Dvorak HF. Tumor-surrogate blood vessel subtypes exhibit differential susceptibility to anti-VEGF therapy.

Cancer Res. Kamba T, Tam BY, Hashizume H, Haskell A, Sennino B, Mancuso MR, et al. VEGF-dependent plasticity of fenestrated capillaries in the normal adult microvasculature. Am J Physiol Heart Circ Physiol. Yang Y, Zhang Y, Cao Z, Ji H, Yang X, Iwamoto H, et al. Anti-VEGF- and anti-VEGF receptor-induced vascular alteration in mouse healthy tissues.

Maynard MA, Marino-Enriquez A, Fletcher JA, Dorfman DM, Raut CP, Yassa L, et al. Thyroid hormone inactivation in gastrointestinal stromal tumors. Jubb AM, Pham TQ, Hanby AM, Frantz GD, Peale FV, Wu TD, et al. Expression of vascular endothelial growth factor, hypoxia inducible factor 1alpha, and carbonic anhydrase ix in human tumours.

J Clin Pathol. Xue Y, Religa P, Cao R, Hansen AJ, Lucchini F, Jones B, et al. Anti-VEGF agents confer survival advantages to tumor-bearing mice by improving cancer-associated systemic syndrome.

Rapid vascular regrowth in tumors after reversal of VEGF inhibition. J Clin Invest. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance. Ghosh K, Thodeti CK, Dudley AC, Mammoto A, Klagsbrun M, Ingber DE.

Tumor-derived endothelial cells exhibit aberrant Rho-mediated mechanosensing and abnormal angiogenesis in vitro. Casanovas O, Hicklin DJ, Bergers G, Hanahan D. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell.

Crawford Y, Kasman I, Yu L, Zhong C, Wu X, Modrusan Z, et al. PDGF-C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti-VEGF treatment. Blouw B, Song H, Tihan T, Bosze J, Ferrara N, Gerber HP, et al.

The hypoxic response of tumors is dependent on their microenvironment. Stacker SA, Achen MG. The vegf signaling pathway in cancer: the road ahead. Chin J Cancer. PubMed Central CAS PubMed Google Scholar. Chen YS, Chen ZP. Vasculogenic mimicry: a novel target for glioma therapy.

Donnem T, Hu J, Ferguson M, Adighibe O, Snell C, Harris AL, et al. Vessel co-option in primary human tumors and metastases: an obstacle to effective anti-angiogenic treatment?

Cancer Med. Nissen LJ, Cao R, Hedlund EM, Wang Z, Zhao X, Wetterskog D, et al. Angiogenic factors FGF2 and PDGF-BB synergistically promote murine tumor neovascularization and metastasis. Cao R, Brakenhielm E, Pawliuk R, Wariaro D, Post MJ, Wahlberg E, et al.

Angiogenic synergism, vascular stability and improvement of hind-limb ischemia by a combination of PDGF-BB and FGF Kerbel RS. Antiangiogenic therapy: a universal chemosensitization strategy for cancer? Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy.

Winkler F, Kozin SV, Tong RT, Chae SS, Booth MF, Garkavtsev I, et al. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, angiopoietin-1, and matrix metalloproteinases.

Ebos JM, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, Kerbel RS. Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Vinals F, et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis.

Zhang D, Hedlund EM, Lim S, Chen F, Zhang Y, Sun B, et al. Antiangiogenic agents significantly improve survival in tumor-bearing mice by increasing tolerance to chemotherapy-induced toxicity.

Ribatti D. Erythropoietin and tumor angiogenesis. Stem Cells Dev. Garcia-Donas J, Rodriguez-Antona C, Jonasch E.

Molecular markers to predict response to therapy. Semin Oncol. Maru D, Venook AP, Ellis LM. Predictive biomarkers for bevacizumab: are we there yet? Clin Cancer Res. Pohl M, Werner N, Munding J, Tannapfel A, Graeven U, Nickenig G, et al.

Biomarkers of anti-angiogenic therapy in metastatic colorectal cancer mCRC : original data and review of the literature. Z Gastroenterol. Niers TM, Richel DJ, Meijers JC, Schlingemann RO. Vascular endothelial growth factor in the circulation in cancer patients may not be a relevant biomarker.

PLoS ONE. Denduluri N, Yang SX, Berman AW, Nguyen D, Liewehr DJ, Steinberg SM, et al. Circulating biomarkers of bevacizumab activity in patients with breast cancer. Cancer Biol Ther. Hegde PS, Jubb AM, Chen D, Li NF, Meng YG, Bernaards C, et al.

Predictive impact of circulating vascular endothelial growth factor in four phase III trials evaluating bevacizumab. Bunni J, Shelley-Fraser G, Stevenson K, Oltean S, Salmon A, Harper SJ, et al. Circulating levels of anti-angiogenic VEGF-A isoform VEGF-AXXXB in colorectal cancer patients predicts tumour VEGF-A ratios.

Am J Cancer Res. PubMed Central PubMed Google Scholar. Lambrechts D, Lenz HJ, de Haas S, Carmeliet P, Scherer SJ. Markers of response for the antiangiogenic agent bevacizumab. J Clin Oncol. Chen C, Sun P, Ye S, Weng HW, Dai QS. Hypertension as a predictive biomarker for efficacy of bevacizumab treatment in metastatic colorectal cancer: a meta-analysis.

J BUON. PubMed Google Scholar. Penzvalto Z, Surowiak P, Gyorffy B. Biomarkers for systemic therapy in ovarian cancer. Angiogenesis inhibitors for cancer can be prescribed by a doctor to take orally by mouth or intravenously by vein; IV.

If you are prescribed an oral angiogenesis inhibitor to take at home, ask if you need to fill the prescription at a pharmacy that handles complex medications, such as a specialty pharmacy. Check with the pharmacy and your insurance company about your insurance coverage and co-pay of the oral medication.

Also, be sure to ask about how to safely store and handle your prescription at home. If you are prescribed an IV treatment, that will be given at the hospital or other cancer treatment facility. Talk with your treatment center and insurance company about how your specific prescription is covered and how any co-pays will be billed.

If you need financial assistance, talk with your health care team, including the pharmacist or a social worker , about co-pay assistance options. National Cancer Institute: Angiogenesis Inhibitors. The Angiogenesis Foundation: Treatments.

Comprehensive information for people with cancer, families, and caregivers, from the American Society of Clinical Oncology ASCO , the voice of the world's oncology professionals.

org Conquer Cancer ASCO Journals Donate. What is Targeted Therapy? Angiogenesis and Angiogenesis Inhibitors to Treat Cancer Understanding Pharmacogenomics Radiation Therapy Surgery When to Call the Doctor During Cancer Treatment What is Maintenance Therapy?

Veterans Prevention and Healthy Living Cancer. Net Videos Coping With Cancer Research and Advocacy Survivorship Blog About Us. Angiogenesis and Angiogenesis Inhibitors to Treat Cancer Approved by the Cancer. What is angiogenesis? H ow do angiogenesis inhibitors treat cancer?

What angiogenesis inhibitors are approved to treat cancer? Thalidomide is not recommended during pregnancy because it causes severe birth defects.

Vandetanib Caprelsa is approved to treat: Medullary thyroid cancer Ziv-aflibercept Zaltrap is approved to treat: Colorectal cancer Researchers are studying whether some of these drugs may treat other types of cancer. Therefore, it is important to summarise the results of these recent studies and to compare the drugs to each other.

This knowledge will help anticancer research move forward, as emerging therapies and promising clinical agents are highlighted in this review to point out future directions of this field. Ogasawara et al. The results show that lenvatinib induced dose- and time-dependent growth suppression of HCC cell lines, with no signs of apoptosis.

Cell lines expressing FGFR1, -2, -3 and -4, FGF19, FRS2α fibroblast growth factor receptor substrate 2 alpha and RET showed the greatest response to treatment. In the in vivo study, lenvatinib-treated mice showed dose-dependent inhibition of tumour growth. In addition, a decrease in blood vessel density and an increase in necrosis were observed in mice, but again no signs of apoptosis were observed.

Therefore, this study demonstrates the antiproliferative and anti-angiogenic effects of lenvatinib on liver cancer cells both in vitro and in vivo , indicating that it could be a promising treatment for patients with HCC.

Indeed, lenvatinib is currently used in patients with advanced HCC, and, although the clinical benefits remain limited [ ] , lenvatinib also seems to delay functional deterioration in these patients, helping with the preservation of health-related quality of life during treatment [ ].

In another more recent study on lenvatinib, Jin et al. This drug combination resulted in synthetic lethality against HCC both in vitro and in vivo in patient-derived HCC tumours in mice. The mechanistic studies showed that gefitinib was able to abrogate the feedback activation of the EGFR-PAK2 [P21 RAC1 activated kinase 2]-ERK5 extracellular signal-regulated kinase 5 signalling pathway that was induced by the inhibition of FGFR by lenvatinib.

The combination treatment was subsequently administered to 12 patients unresponsive to lenvatinib with advanced HCC, where a better clinical response was achieved, proposing this regimen as a novel strategy for advanced HCC patients with overexpression of EGFR.

Anlotinib is a multiple kinase inhibitor that has shown efficacy against various types of cancer [ , ]. To date, no effective treatment has been found for patients with poorly differentiated papillary thyroid cancer PTC or anaplastic thyroid cancer ATC [ ] , a recent study aimed to investigate the effect of anlotinib against thyroid cancer in vitro and in vivo [ ].

The results show growth inhibition of ATC and PTC cells in vitro with IC 50 values of 3. In addition, anlotinib inhibited cell migration, as well as the in vivo growth of thyroid tumours transplanted to mice. Thus, this study shows that anlotinib has anticancer activity and could be considered as an effective therapeutic approach for patients with advanced thyroid carcinoma.

For more effective delivery of anlotinib, Gao et al. The hydrogel was prepared by encapsulating anlotinib with hyaluronic acid-tyramine HA-Tyr conjugates AL-HA-Tyr.

The in vitro investigation showed that AL-HA-Tyr successfully suppressed the angiogenic capacity and proliferation of HUVECs and LLC cells, respectively.

In addition, inhibition of invasion and migration of HUVECs and LLC cells was observed. In the in vivo studies, LLC mouse models were treated with oral saline solution, oral anlotinib or intratumoral injection of HA-Tyr or AL-HA-Tyr. AL-HA-Tyr reduced visceral toxicity and downregulated Ki67 and VEGF-A in cancer cells, increasing mouse survival.

Thus, this study proposes a more effective anlotinib delivery strategy in order to reduce the systemic toxicity of anlotinib and potentiate its efficacy. Goff et al. At the same time, this molecule is involved in pathways that contribute to tumour growth, angiogenesis and metastasis [ , ].

One of the double inhibitors developed, R, showed comparable activity to sunitinib against breast cancer and renal cell carcinoma xenograft models.

Thus, it appears that dual inhibition of Axl receptor and VEGFR may be an effective anticancer treatment comparable to the known anti-angiogenic agent sunitinib. Previously reported data show that FGF promotes angiogenic signalling in HCC through the activation of the VEGF pathway [ - ].

Therefore, infigratinib-a pan-FGFR kinase inhibitor-was explored for its potential to overcome HCC resistance to VEGF-targeting drugs, such as bevacizumab [ ]. The combination regimen inhibited tumour growth, as well as invasion and metastasis to the lungs in vivo. Mice bearing high-FGFR HCC tumours showed an additional prolongation in overall survival OS.

Indeed, this combination was found effective in providing long-lasting tumour growth inhibition, reduced cell differentiation and reduced drug resistance [ ].

In a recent study, Lu et al. Indeed, the results show that anti-VEGFRAF binds effectively to VEGFR2 Ig immunoglobulin -like domain 3, thus inhibiting its interaction with VEGF-A. This led to in vitro inhibition of angiogenesis and Ab-dependent cell cytotoxicity and complement activation.

By testing the Ab activity in a mouse prostate cancer model, the treated mice showed reduced tumour growth and angiogenesis comparable with the FDA Food and Drug Administration -approved ramucirumab. In addition, the combination therapy of anti-VEGFRAF with docetaxel was even more effective in vivo against prostate cancer.

Anti-VEGFRAF treatment was also applied to mice with HL leukaemia. These mice showed increased survival and inhibition of leukemic cell metastasis to the ovaries and lymph nodes compared to ramucirumab.

Therefore, this study shows that the novel Ab anti-VEGFRAF is a potentially effective treatment for prostate cancer, leukaemia and possibly other types of cancer that overexpress VEGFR2. A novel VEGFR2 inhibitor, YLL, was recently developed against triple-negative breast cancer TNBC [ ].

Other in vitro results report inhibition of cancer cell proliferation, migration and invasion. The effects of YLL were higher than or equal to those of sorafenib, a known inhibitor of angiogenic factors such as VEGFR1, VEGFR2, VEGFR3, PDGFR-β, KIT and RET [ ].

The results of this study suggest that the VEGFR2 inhibitor YLL is a potentially useful anticancer drug. The observed resistance against clinically used anti-VEGF therapies limits the efficacy of such therapies.

This resistance could be attributed to an independent pathway of angiogenesis that can compensate for the inhibition of the VEGF-related cascade, which is mediated by 2- ω-carboxyethyl pyrrole CEP in a TLR2 Toll-like receptor 2 -dependent manner [ ]. An antibody against CEP was, thus, constructed and tested for its anti-angiogenic properties both as a monotherapy and in combination with bevacizumab [ ].

Comparing the efficacy of the combination therapy with bevacizumab monotherapy at 2. Thus, multiple targeting of angiogenic pathways may increase the efficacy of anticancer therapies. Another recent study [ ] tested the anticancer efficacy of a combination of four active ingredients [astragaloside IV, α-solanine, neferine and 2,3,5,6-tetramethylpyrazine SANT ] isolated from traditional Chinese plants against TNBC.

Overexpression of heparanase HPSE is often found in breast cancer tissues with the potential to enhance carcinogenesis and affect the process of autophagy in cancer cells [ - ]. HPSE upregulation was associated with poor outcomes in these patients.

Administration of the SANT combination resulted in in vitro inhibition of cancer cell proliferation and migration and increased the rate of autophagy in TNBC cells. Several genes were affected, and the proteomics studies found that SANT downregulated HB-EGF heparin-binding EGF-like growth factor , thrombospondin-2, amphiregulin, leptin, IGFBP-9 insulin-like growth factor-binding protein 9 , EGF, coagulation factor III and MMP-9 matrix metalloproteinase 9 , while increasing the levels of serpin E1 and platelet factor 4, proteins that play an important role in angiogenesis.

Finally, a novel Hsp90 inhibitor, AT, was recently developed against breast cancer [ ]. Hsp90 has been shown to play an important role in oncogenesis, as it regulates the stabilisation and activation of many proteins that are essential for cell survival and tumour growth [ , ].

The results of the study show that AT suppressed capillary formation, as well as the migration and invasion of HUVECs more efficiently than another Hsp90 inhibitor, AAG.

Thus, these results suggest that AT may be a new drug for the treatment of breast cancer. In , a phase II clinical trial [ ] involving patients with refractory metastatic soft tissue sarcoma after receiving anthracycline-based chemotherapy, naïve from angiogenesis inhibitors and with at least one measurable lesion according to RECIST 1.

More specifically, the median progression-free survival PFS was 5. The most common grade 3 or higher side effects associated with anlotinib treatment were hypertension, increased blood triglyceride levels and pneumothorax. No deaths were reported due to anlotinib treatment. Thus, anlotinib proved effective in increasing patient survival, while no significant adverse effects were reported.

Patients progressing after second-line or further treatment and receiving anlotinib had significantly longer OS than the placebo group, 9. In addition, a significant increase was observed in the PFS 5.

Common grade 3 or higher side effects included hypertension and hyponatremia. Therefore, the results of this study show that anlotinib is a well-tolerated further treatment, which results in a significant improvement in patients with advanced NSCLC.

Surufatinib is a small molecular inhibitor of VEGFR1, VEGFR2, VEGFR3 and FGFR1 [ ]. A randomised, double-blind, placebo-controlled, phase III study was conducted to investigate the efficacy and safety of this drug in patients with extrapancreatic neuroendocrine tumours [ ].

The study included adult patients [after receiving up to two kinds of previous systemic regimens and with unresectable or metastatic, well-differentiated, extrapancreatic neuroendocrine tumours of Eastern Cooperative Oncology Group ECOG performance status of 0 or 1] who were randomised to the surufatinib group mg or the placebo group.

The median PFS in the surufatinib group was 9. The most common grade 3 or higher side effects associated with surufatinib treatment were hypertension and proteinuria. Hence, surufatinib significantly increases the PFS of patients with progressive, advanced, well-differentiated extrapancreatic neuroendocrine tumours, while having an acceptable toxicity profile.

In a similar multicentre, randomised, double-blind, placebo-controlled, phase III trial, surufatinib was evaluated for its efficacy and safety in patients with advanced pancreatic neuroendocrine tumours [ ]. The study included adult patients after receiving up to two kinds of previous systemic regimens for advanced disease and with progressive, advanced, well-differentiated pancreatic neuroendocrine tumours of ECOG performance status of 0 or 1 who were randomised to the surufatinib mg or placebo group, both administered orally once daily in successive four-week treatment cycles.

The median PFS in the surufatinib group was The most common grade 3 or higher side effects associated with surufatinib treatment were hypertension, proteinuria and hypertriglyceridemia.

Therefore, surufatinib significantly increases the PFS of patients with pancreatic neuroendocrine tumours, as similarly reported for extrapancreatic neuroendocrine tumours. The drug has significant toxicity, which, however, is acceptable when compared to its overall benefit.

Based on the above trials, surufatinib received its first approval in China for the treatment of late-stage, well-differentiated, extrapancreatic neuroendocrine tumours in December [ ]. Surufatinib is currently in preregistration in China for pancreatic neuroendocrine tumours and in the USA for pancreatic and extrapancreatic neuroendocrine tumours, while other ongoing clinical trials are investigating its benefit for other solid tumours, including thyroid cancer, biliary tract carcinoma and soft tissue sarcoma.

The highest dose of lenvatinib 24 mg daily plus pembrolizumab was associated with two grade 3 toxicities arthralgia and fatigue , so the recommended phase II dose of lenvatinib was established at 20 mg daily plus pembrolizumab.

The most common side effects reported were fatigue, diarrhoea, hypertension and hypothyroidism. Thus, the combination of lenvatinib with pembrolizumab in patients with advanced solid tumours shows good efficacy, while the side effects observed are manageable.

Previous studies have shown that the combination of PD-1 programmed cell death protein 1 checkpoint inhibitors with VEGF-pathway TKIs leads to increased cytotoxicity and decreased cancer cell growth, but it is also related to severe side effects [ ].

Based on this, Atkins et al. Thus, the combination therapy of axitinib and pembrolizumab appears to be a relatively well-tolerated treatment, with significant efficacy against treatment-naïve advanced renal cell carcinoma.

A subsequent phase III clinical trial trying to assess the long-term efficacy and safety of the combination of pembrolizumab mg intravenously, every three weeks for up to 35 cycles with axitinib 5 mg orally, twice daily , showed increased efficacy of the combination treatment over sunitinib monotherapy 50 mg orally, once daily for four weeks per six-week cycle in treatment-naive, advanced renal cell carcinoma [ ].

Of the registered patients with a median follow-up of Therefore, the combination treatment has a superior clinical benefit over sunitinib monotherapy and can be considered as the first-line treatment for advanced renal cell carcinoma patients.

Regorafenib is an oral diphenylurea multikinase inhibitor against angiogenic VEGFR1, VEGFR2, VEGFR3 and TIE2 , stromal PDGFR-β and FGFR and oncogenic receptor tyrosine kinases KIT, RET and RAF , which has been established for metastatic colorectal cancer [ , ].

The median PFS achieved was 3. The most common grade 3 or higher side effects were hypertension, hand-foot skin reaction and hypophosphataemia, with no treatment-related deaths.

Hence, this study shows that regorafenib is an effective treatment for chemotherapy-refractory metastatic colorectal cancer with an acceptable safety profile.

The overexpression of α-fetoprotein in patients with advanced HCC has been correlated with poor prognosis [ - ]. The median follow-up was 7. However, the analysis of the results did not show a significant difference in the objective response and the median deterioration time between the two groups.

Common grade 3 or higher treatment-related adverse effects were hypertension, hyponatraemia and elevated aspartate aminotransferase. Ramucirumab-related side effects were also responsible for the death of three patients.

Hence, this study shows that ramucirumab is a potentially useful treatment for patients with advanced HCC and α-fetoprotein overexpression previously on sorafenib, although the treatment causes significant side effects, and further studies are needed to gain statistically significant results.

TAS, an oral nucleoside anticancer agent that combines trifluridine and tipiracil, has shown a significant OS prolongation in patients with refractory metastatic colorectal cancer [ ]. The co-treatment of TAS with bevacizumab has also shown promising results in colorectal cancer xenograft models compared to the corresponding monotherapies [ ].

Thus, Kuboki et al. After 16 weeks of treatment, PFS was achieved in The most common grade 3 or higher side effects observed in patients were neutropenia, leukopenia, anaemia, febrile neutropenia and thrombocytopenia.

Serious adverse reactions were reported in only three patients, but no deaths were reported due to the regimen. This study shows that the combination of TAS with bevacizumab has significant efficacy in patients with refractory metastatic colorectal cancer, while it is a treatment that does not present significant overall toxicity.

The aim of the phase II study by Palazzo et al. An objective response was observed in 30 patients and a pathologic complete remission pCR in 10 of the 34 patients.

This study showed that this neoadjuvant therapy, including chemotherapy and anti-angiogenic treatment, significantly increases disease-free survival and OS for patients with inflammatory breast cancer.

Another recent phase II study evaluated the efficacy and safety of a treatment that included apatinib mg orally once daily and etoposide 50 mg orally once daily on Days of a day cycle for up to six cycles in patients with platinum-resistant or platinum-refractory ovarian cancer [ ].

Of the 35 patients in the study, 19 showed an objective response to treatment, while the most common grade 3 or higher side effects observed were neutropenia, fatigue, anaemia and mucositis, and two patients developed severe side effects.

However, no treatment-related deaths were reported. Therefore, the combination of apatinib with etoposide seemed to be significantly effective in more than half of the platinum-resistant ovarian cancer patients registered in the present study, with manageable adverse reactions.

The aim of this review article is to present the most up-to-date data on novel anti-angiogenic factors that are under development or clinical evaluation.

In total, 15 different anti-angiogenic agents are included based on recent publications of the last five years, which confirms that the discovery of anti-angiogenic factors has been an active and ongoing field of research.

The obvious disadvantage of preclinical studies is that the pharmacokinetics and possible side effects of the proposed drugs are usually not studied. Indeed, a regimen may be effective in vitro and in vivo but toxic for humans and, therefore, not pursued further.

In addition, we must always consider the differences between the in vivo models and humans, which may be responsible for the clinical failure of preclinically identified promising agents. In this scope, both preclinical and clinical studies of lenvatinib and anlotinib are included to follow up on the overall efficacy of these two agents.

Lenvatinib showed promising in vitro and in vivo antiproliferative and anti-angiogenic activity against liver cancer cells [ ]. As proof of concept, lenvatinib showed a significant response rate in patients with metastatic renal cell carcinoma, endometrial cancer, squamous cell carcinoma of the head and neck, melanoma, non-small cell lung cancer and urothelial carcinoma, while it was not correlated with serious side effects [ ].

Anlotinib was found cytotoxic against thyroid cancer cells, suppressed in vitro migration and decreased tumour growth in vivo [ ]. In clinical trials, anlotinib was effective against refractory soft tissue sarcoma [ ] and non-small cell lung cancer [ ].

Although no treatment-related deaths were reported, several adverse reactions were observed in patients in both clinical trials. Of particular interest are the results of a recent study [ ] where, to avoid anlotinib-related systemic side effects, a hydrogel containing anlotinib was manufactured, followed by an intratumoral injection to a mouse model of Lewis lung cancer.

The results are particularly encouraging, giving new hope for providing targeted therapies with increased efficacy and reduced side effects via novel delivery systems. Several other anti-angiogenic agents under preclinical development are also discussed, including YLL for breast cancer [ ] , R for breast and renal cancer [ ] and AT for breast cancer [ ].

Recently completed clinical trials with positive results are also presented, namely for monotherapies such as oral surufatinib against pancreatic and extrapancreatic neuroendocrine cancers, even though severe side effects were reported [ , ] , and regorafenib against advanced colorectal cancer [ ].

In addition, ramucirumab, a mAb under development, was clinically tested against advanced hepatocellular carcinoma in patients with increased α-fetoprotein levels, where it showed high efficacy [ ]. Finally, the combination of chemotherapeutics with different mechanisms of action is a common practice in cancer patient care; therefore, some studies with combination regimens are also discussed here.

Indicatively, bevacizumab was combined with carboplatin, paclitaxel and metronomic cyclophosphamide, where it was found useful against inflammatory breast cancer [ ] , and in another clinical trial, it was combined with TAS against metastatic colorectal cancer, where again positive results were obtained [ ].

Moreover, apatinib combined with etoposide was effective against ovarian cancer unresponsive to platinum [ ] , whereas an anti-PD-1 mAb combined with axitinib also showed an increased patient response, although several side effects were induced [ , ]. Summaries of the presented anti-angiogenic agents categorised into preclinical and clinical studies, along with the main findings of each study, are shown in Tables 1 and 2.

Summary of the anti-angiogenic agents currently being tested in pre-clinical models discussed in this article.

Summary of the anti-angiogenic agents currently in clinical trials discussed in this article. It should be noted that most clinical studies found treatment-related side effects, which were mostly mild, while few deaths were reported. However, since most of the studies include patients with highly resistant or metastatic cancers that do not respond to other treatments, the overall efficacy of these therapies in prolonging patient survival outweighs the frequent side effects.

Cancer is a global epidemic and the second most common cause of death. Despite the active research, it remains a condition that, in most cases, is not cured and leads to death. Observing the distinctive physiology of cancer cells, the angiogenesis pathway emerged as a promising anticancer target.

Thus, in recent years, more and more researchers are aiming to develop various anti-angiogenic agents and regulate their efficacy and safety. Regarding this, the present review summarises the results of very recent preclinical and clinical studies involving monotherapies or combination therapies of anti-angiogenic agents, in order to highlight the updates in the field and encourage more related research.

Apart from these therapies, however, several other anti-angiogenic approaches are currently being explored, such as the use of natural phytochemicals as anti-angiogenic agents in cancer [ ]. However, the clinical translation of dietary phytochemicals still faces challenges, as the results thus far remain limited, contradictory, inconclusive or even inexistent for some of these compounds.

In this context, a significant effort is needed to improve clinical trial design, taking serious consideration of the target population, as well as the pharmacokinetic and pharmacodynamic properties of these compounds. Other novel anti-angiogenic strategies include the exploitation of miRNAs-elucidating their role in angiogenesis and evaluating novel miRNA-mediated immunotherapies [ ].

Advances in miRNA targeting and delivery strategies, such as the use of nanoparticles or cell-derived membrane vesicles for therapeutic miRNA delivery, may provide miRNA-based therapeutics as anti-angiogenic treatments.

Additionally, the use of anti-angiogenic peptides has emerged as a promising anticancer strategy, as they present several advantages, with rapid production through automated chemical synthesis and the ability to undergo structural modifications. Therefore, several anticancer peptides that have been discovered derive from nanobodies and mimotopes resources, as well as natural resources such as venoms, bacteria, fungi, sponges, animals and plants [ ].

The new methods available in phage display and peptidomimetics have contributed to the development of new anti-angiogenic peptides, while the novel technologies such as pegylation, polymer-supported formulation and macromolecule conjugation can now provide these peptides with better pharmacokinetic properties and contribute to more anti-angiogenic peptide-based therapeutics.

Although all of the above treatment strategies appear to be particularly promising for the future, the issue of developing angiogenic resistance should first be addressed. The combinatorial treatments mentioned have shown excellent results and will be a key concept of future therapeutic regimens.

Emerging ideas about novel delivery systems of such therapies might be the way forward. In addition, the simplification of regimens should be explored, especially when combination therapies are indicated, such as oral administration or the preparation of a combination formulation to increase patient adherence.

Mattiuzzi C, Lippi G. Current cancer epidemiology. J Epidemiol Glob Health ; Guerra A, Belinha J, Mangir N, MacNeil S, Natal Jorge R. Simulation of the process of angiogenesis: quantification and assessment of vascular patterning in the chicken chorioallantoic membrane.

Comput Biol Med ; Naito H, Iba T, Takakura N. Mechanisms of new blood-vessel formation and proliferative heterogeneity of endothelial cells. Int Immunol ; Carmeliet P. Mechanisms of angiogenesis and arteriogenesis. Nat Med ; Folkman J, Long DM Jr, Becker FF. Growth and metastasis of tumor in organ culture.

Cancer ; de Heer EC, Jalving M, Harris AL. HIFs, angiogenesis, and metabolism: elusive enemies in breast cancer. J Clin Invest ; Lugano R, Ramachandran M, Dimberg A.

Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci ; Rajabi M, Mousa SA.

The role of angiogenesis in cancer treatment. Biomedicines ; Melincovici CS, Boşca AB, Şuşman S, et al. Vascular endothelial growth factor VEGF -key factor in normal and pathological angiogenesis. Rom J Morphol Embryol ; Bai Y, Bai L, Zhou J, Chen H, Zhang L.

Sequential delivery of VEGF, FGF-2 and PDGF from the polymeric system enhance HUVECs angiogenesis in vitro and CAM angiogenesis. Cell Immunol ; Armani G, Pozzi E, Pagani A, et al. The heterogeneity of cancer endothelium: the relevance of angiogenesis and endothelial progenitor cells in cancer microenvironment.

Microvasc Res ; Huang XL, Khan MI, Wang J, et al. Role of receptor tyrosine kinases mediated signal transduction pathways in tumor growth and angiogenesis-New insight and futuristic vision. Int J Biol Macromol ; Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen.

Science ; Montesano R, Vassalli JD, Baird A, Guillemin R, Orci L. Basic fibroblast growth factor induces angiogenesis in vitro. Proc Natl Acad Sci U S A ; Fett JW, Strydom DJ, Lobb RR, et al.

Isolation and characterization of angiogenin, an angiogenic protein from human carcinoma cells. Biochemistry ; Schreiber AB, Winkler ME, Derynck R. Transforming growth factor-alpha: a more potent angiogenic mediator than epidermal growth factor.

Roberts AB, Sporn MB, Assoian RK, et al. Transforming growth factor type beta: rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro.

Fràter-Schröder M, Risau W, Hallmann R, Gautschi P, Böhlen P. Tumor necrosis factor type alpha, a potent inhibitor of endothelial cell growth in vitro, is angiogenic in vivo. Ishikawa F, Miyazono K, Hellman U, et al. Identification of angiogenic activity and the cloning and expression of platelet-derived endothelial cell growth factor.

Nature ; Bussolino F, Ziche M, Wang JM, et al. In vitro and in vivo activation of endothelial cells by colony-stimulating factors. Zhao J, Chen L, Shu B, et al. PLoS One ;9:e Zheng Q, Li X, Cheng X, et al. Granulocyte-macrophage colony-stimulating factor increases tumor growth and angiogenesis directly by promoting endothelial cell function and indirectly by enhancing the mobilization and recruitment of proangiogenic granulocytes.

Tumour Biol ; Zhou Y, Tu C, Zhao Y, Liu H, Zhang S. Biochem Biophys Res Commun ; Kitadai Y, Takahashi Y, Haruma K, et al. Transfection of interleukin-8 increases angiogenesis and tumorigenesis of human gastric carcinoma cells in nude mice.

Br J Cancer ; Kaga T, Kawano H, Sakaguchi M, Nakazawa T, Taniyama Y, Morishita R. Hepatocyte growth factor stimulated angiogenesis without inflammation: differential actions between hepatocyte growth factor, vascular endothelial growth factor and basic fibroblast growth factor.

Vascul Pharmacol ; Gospodarowicz D, Bialecki H, Thakral T. The angiogenic activity of the fibroblast and epidermal growth factor.

Experimental Eye Research ; Yokoi A, Mccrudden KW, Huang J, et al. Journal of Pediatric Surgery ; Palinski W, Monti M, Camerlingo R, et al. Lysosome purinergic receptor P2X4 regulates neoangiogenesis induced by microvesicles from sarcoma patients.

Cell Death Dis ; Pavlakovic H, Havers W, Schweigerer L. Multiple angiogenesis stimulators in a single malignancy: implications for anti-angiogenic tumour therapy. Angiogenesis ; Shibuya M. Vascular endothelial growth factor VEGF and its receptor VEGFR signaling in angiogenesis: a crucial target for anti- and pro-angiogenic therapies.

Genes Cancer ; Muller YA, Li B, Christinger HW, Wells JA, Cunningham BC, de Vos AM. Vascular endothelial growth factor: crystal structure and functional mapping of the kinase domain receptor binding site. Role of VEGF-flt receptor system in normal and tumor angiogenesis.

Adv Cancer Res ; Li L, Liu H, Xu C, et al. VEGF promotes endothelial progenitor cell differentiation and vascular repair through connexin Stem Cell Res Ther ; Zhu D, Li Y, Zhang Z, et al. Recent advances of nanotechnology-based tumor vessel-targeting strategies.

J Nanobiotechnology ; El-Kenawi AE, El-Remessy AB. Angiogenesis inhibitors in cancer therapy: mechanistic perspective on classification and treatment rationales. Br J Pharmacol ; Mundel TM, Kalluri R.

Type IV collagen-derived angiogenesis inhibitors. Abdollahi A, Hahnfeldt P, Maercker C, et al. Molecular Cell ; Ranjit PM, Anuradha C, Vishnupriya S, Girijasankar G, Girish K, Chowdary YA.

Metrics details. Angiogenesis has Ant-iangiogenesis Anti-angiogenesis clinical trials the topic of major scientific interest Weight management program the field of malignant tumors. Nowadays, targeting Anti-angiogenesis clinical trials Anti-angiogdnesis achieved success Anti-angiogenesia various Anti-angiogenesis clinical trials by several mechanisms, including the use of anti-angiogenic small molecule receptor tyrosine kinase inhibitors TKIs. The development of TKIs targeting pro-angiogenic receptors, mainly vascular endothelial growth factor receptor VEGFR family, have significantly improved the outcome of certain types of cancers, like renal cell carcinoma, hepatocellular carcinoma, and colorectal carcinoma. However, the general response rate is not very satisfactory. Journal Anti-wngiogenesis the Egyptian National Cancer Anti-antiogenesis volume 33Article number: 15 Cite this article. Breakfast skipping and cultural influences Anti-angiogenesis clinical trials. Anti-angigenesis is trialss formation of new vascular networks from preexisting Anti-angiogenesis clinical trials through the migration and proliferation of differentiated endothelial cells. Available evidence suggests that while antiangiogenic therapy could inhibit tumour growth, the response to these agents is not sustained. The aim of this paper was to review the evidence for anti-angiogenic therapy in cancer therapeutics and the mechanisms and management of tumour resistance to antiangiogenic agents.

Skip to Content. Angiogenesis Endurance nutrition for team sports are grials type of cancer treatment. They stop Anti-angiogenesiss Anti-angiogenesis clinical trials in the body called angiogenesis, or ttrials vessel formation.

Angiogenesis is how the trixls forms new Anti-angiofenesis vessels. This is a normal part of growth Anti-agniogenesis healing. But Anti-angioogenesis angiogenesis can play a role in diseases such as cancer. Ati-angiogenesis grow, a tumor needs nutrients and oxygen from your blood.

The Anti-angiogfnesis sends trails that stimulate more trjals vessels to Anti-angigoenesis and carry Anti-angiogenesis clinical trials blood. Angiogenesis inhibitors, also clnical anti-angiogenics, cliniczl blood vessel growth.

By blocking nutrients and oxygen Boosting immune function through nutrition a tumor, the angiogenesis inhibitors "starve" the tumor. The Anti-ajgiogenesis.

Food and Troals Administration has approved Anti-angiogenesix angiogenesis inhibitors. They may affect angiogenesis in frials than one way, and some of them can also affect other triials that a Anto-angiogenesis grows. Angiogenesis inhibitors can be given coinical or in combination with other types of clinicla.

Researchers are studying whether tdials of these drugs may Hyperglycemia and cellular damage other clinicla Anti-angiogenesis clinical trials cancer.

Talk trias your health care team about tirals trials for clinlcal inhibitors. Clinival of clunical body's Anti-angiogeneeis functions Anti-angiogenesis clinical trials on angiogenesis. Therefore, angiogenesis inhibitors Anti-sngiogenesis cause a wide range of physical coinical effects including:.

Anti-angiogenesis clinical trials syndromewhich Anti-angiogenesis clinical trials tender, thickened areas on Anti-angiogenesiss palms and soles. Caffeine and muscular endurance, it causes blisters.

Although Anti-angiogenesis clinical trials, these side effects do cliniccal happen with every drug or Herbal tea for digestion person. Ajti-angiogenesis, there are Anti-ngiogenesis can help manage these side effects when they Ant-angiogenesis occur.

Be Trjals to let your health care team Anti-angiogenesia Anti-angiogenesis clinical trials side effects you experience. If an angiogenesis inhibitor is recommended Anti-angiogenesid you, talk with triasl doctor about the specific triasl benefits and risks of that medication.

Also, ask about Anti-angiognesis side effects can be managed and what side effects to watch for. Angiogenesis inhibitors for cancer can be prescribed by a doctor to take orally by mouth or intravenously by vein; IV.

If you are prescribed an oral angiogenesis inhibitor to take at home, ask if you need to fill the prescription at a pharmacy that handles complex medications, such as a specialty pharmacy.

Check with the pharmacy and your insurance company about your insurance coverage and co-pay of the oral medication. Also, be sure to ask about how to safely store and handle your prescription at home. If you are prescribed an IV treatment, that will be given at the hospital or other cancer treatment facility.

Talk with your treatment center and insurance company about how your specific prescription is covered and how any co-pays will be billed. If you need financial assistance, talk with your health care team, including the pharmacist or a social workerabout co-pay assistance options.

National Cancer Institute: Angiogenesis Inhibitors. The Angiogenesis Foundation: Treatments. Comprehensive information for people with cancer, families, and caregivers, from the American Society of Clinical Oncology ASCOthe voice of the world's oncology professionals.

org Conquer Cancer ASCO Journals Donate. What is Targeted Therapy? Angiogenesis and Angiogenesis Inhibitors to Treat Cancer Understanding Pharmacogenomics Radiation Therapy Surgery When to Call the Doctor During Cancer Treatment What is Maintenance Therapy? Veterans Prevention and Healthy Living Cancer.

Net Videos Coping With Cancer Research and Advocacy Survivorship Blog About Us. Angiogenesis and Angiogenesis Inhibitors to Treat Cancer Approved by the Cancer. What is angiogenesis? H ow do angiogenesis inhibitors treat cancer? What angiogenesis inhibitors are approved to treat cancer?

Thalidomide is not recommended during pregnancy because it causes severe birth defects. Vandetanib Caprelsa is approved to treat: Medullary thyroid cancer Ziv-aflibercept Zaltrap is approved to treat: Colorectal cancer Researchers are studying whether some of these drugs may treat other types of cancer.

What are the side effects of angiogenesis inhibitors? Therefore, angiogenesis inhibitors can cause a wide range of physical side effects including: High blood pressure A rash or dry, itchy skin Hand-foot syndromewhich causes tender, thickened areas on your palms and soles.

Diarrhea Fatigue Low blood counts Problems with wound healing or cuts reopening Although common, these side effects do not happen with every drug or every person.

Rare side effects include: Serious bleeding Heart attacks Heart failure Blood clots Holes in the intestines, called bowel perforations If an angiogenesis inhibitor is recommended for you, talk with your doctor about the specific potential benefits and risks of that medication.

How are angiogenesis inhibitors given? Questions to ask your health care team Consider asking these questions about angiogenesis inhibitors: Do you recommend an angiogenesis inhibitor as part of my treatment plan? Which one? What are the possible risks and benefits of the drug?

What are the potential short- and long-term side effects of this medication? How long will this treatment last? How is this drug different from chemotherapy or other treatments? Will I take this drug at home or at the hospital? Will I need other cancer treatments in addition to this angiogenesis inhibitor?

Which clinical trials are options for me? Who can help me manage the costs of my prescriptions? Related Resources Understanding Targeted Therapy Skin Reactions to Targeted Therapy and Immunotherapy More Information National Cancer Institute: Angiogenesis Inhibitors The Angiogenesis Foundation: Treatments.

Navigating Cancer Care. Net Videos. Find a Cancer Doctor.

: Anti-angiogenesis clinical trials

What is anti angiogenesis treatment? Lancet Oncol ; Leuk Res ; J Neuropathol Exp Neurol ; A non—RGD-based integrin binding peptide ATN blocks breast cancer growth and metastasis in vivo. Vafopoulou, Polyxeni, Malamati Kourti.
Anti-angiogenesis in cancer therapeutics: the magic bullet

Anti-angiogenic drug targets. Monospecific bevacizumab, 2—3-targeted aflibercept and ramucirumab, and multi-targeted tyrosine kinase inhibitor anti-angiogenic drugs are currently used to treat cancer in human patients. VEGF signaling and anti-VEGF drug targets. VEGF stimulates tumor angiogenesis by activating endothelial VEGFR2 and its downstream signaling.

Drugs targeting various signaling components have been developed for clinical use. VEGF vascular endothelial growth factor, VEGFR2 vascular endothelial growth factor receptor 2.

In contrast to antibody-based and soluble receptor-based biologics, small chemical compound-based drugs are far less specific. The most commonly used tyrosine kinase inhibitors TKIs that block VEGFR-mediated signaling pathways are small chemical molecules targeting a broad spectrum of kinases [ 36 , 37 ].

Most VEGFR-TKIs, including sunitinib, sorafenib, and pazopanib, indistinguishably target VEGFR1, VEGFR2, and VEGFR3 signaling pathways. Additionally, these receptor inhibitors also block many other receptor kinases that are not parts of the VEGFR family but are often related to angiogenic signaling pathways, including members of the platelet-derived growth factor PDGF receptor and fibroblast growth factor FGF receptor families [ 38 ].

Theoretically, anti-angiogenic drugs that target abroad spectrum of signaling pathways would be more desirable and effective for treating cancer since malignant tissues are heterogeneous with different populations of tumor and host cells that produce various angiogenic factors.

In this regard, anti-angiogenic TKIs would be more effective than antibody-based and soluble receptor-based drugs that solely target the VEGF pathway. However, clinical experience with anti-angiogenic therapy shows that TKIs may not necessarily be more effective than bevacizumab.

Additionally, anti-angiogenic TKIs and bevacizumab show different profiles of toxicity, although both classes of drugs commonly cause some adverse effects.

An important difference between biologics and TKIs is that antibody-based drugs have a longer half-life than small chemical molecules. They are inactivated using different metabolic pathways.

Anti-angiogenic drugs target tumor blood vessels that exhibit heterogeneity [ 39 ]. However, none of available drugs are specifically delivered to the tumor tissue. They are delivered systemically to cancer patients, exposing all the tissues and organs to the drugs [ 22 ].

Would systemic delivery of anti-angiogenic drugs affect non-tumoral healthy vasculatures? In tumor-free healthy mice, systemic treatment with anti-angiogenic drugs, including an anti-VEGF neutralizing antibody and TKI-targeting VEGFRs, resulted in robust vascular regression in many tissues and organs.

In all tissues, vasculatures in endocrine organs, including the thyroid, adrenal gland, ovary, and pancreatic β-islets, underwent robust regression in response to systemic anti-angiogenic therapy [ 40 ].

In addition to changes in vascular density, the endothelia underwent structural changes by replacing fenestrae with the intracellular vesiculo-vacuolar organelles. In normal physiological conditions, VEGF is a crucial hemostatic factor for endothelial cell survival and endothelium fenestrations in endocrine vasculatures.

Thus, systemic inhibition of VEGF function would inevitably cause structural changes and decreases in vascular density. The anti-angiogenic drug-induced vascular changes also produce functional alterations in their respective organs.

For example, thyroid hormones are significantly decreased after prolonged treatment with anti-VEGF drugs, resulting in hypothyroidism [ 41 ]. In addition to causing changes to the endocrine organs, anti-VEGF drugs also induce rigorous vascular regression in the liver, gastrointestinal wall, and kidney cortex [ 41 ].

Vascular regression inevitably creates a hypoxic environment in the targeted tissues and organs that eventually affects organ functions. These functional changes manifest as clinically adverse effects, such as hypertension, gastrointestinal perforation, hemorrhages, and protein in urine, which are commonly seen in cancer patients who are treated with anti-angiogenic drugs [ 15 , 38 , 42 ].

Paradoxically, off-tumor targets of anti-VEGF drugs can sometimes be beneficial for cancer patients [ 22 ]. This is particularly the case if circulating VEGF expression levels are extremely high in the patients whose tumors produce high amounts of VEGF.

For example, in patients with von Hippel—Lindau Vhl gene-mutated renal cell carcinoma, VEGF expression levels can be very high [ 43 ].

Circulating VEGF also causes destructive effects in remote healthy tissues and organs, such as the bone marrow, liver, and spleen [ 44 ].

In this case, inhibition of VEGF-induced vascular impairment would potentially improve patient survival, as shown in preclinical models.

An important and clinically practical issue related to anti-angiogenic therapy is length of treatment. How long should a cancer patient be treated with anti-angiogenic drugs? What would happen if anti-angiogenic treatment was discontinued? Currently, no consensus exists regarding treatment timeline with anti-angiogenic drugs.

As an anti-angiogenic component is added to the standard chemotherapy regimen, anti-angiogenic therapy follows the timeline of chemotherapy. In clinical practice, anti-angiogenic treatment will inevitably be discontinued.

Additionally, anti-angiogenic treatment will likely result in adverse effects that make therapy withdrawal difficult. Similarly, if patients acquire drug resistance during treatment, this can also result in discontinuation of therapy.

In animal cancer models, discontinuation of anti-angiogenic therapy resulted in rapid regrowth of tumor blood vessels [ 45 ]. For small chemical compound-based drugs, revascularization occurs 2—3 days after drug withdrawal and reaches a maximal level around day 7.

Around this time, revascularization generates a rebound time window that drives angiogenesis to a level higher than it was prior to treatment [ 41 ]. It is possible that rebound angiogenesis reflects the time course of angiogenic vessel growth before vascular remodeling and maturation.

Effects of ON and OFF treatment with anti-angiogenic drugs on tumor vasculatures. Rapid revascularization and rebound angiogenesis can occur after treatment is discontinued.

It is unclear if, after discontinuation of anti-angiogenic therapy, rebound angiogenesis also occurs in human patients.

However, reasonable speculation suggests that human tumors and mouse tumors would respond similarly. If rebound angiogenesis does occur in human cancer patients, discontinuation of anti-angiogenic therapy could potentially result in accelerated tumor growth.

Thus, non-stop, lifetime anti-angiogenic treatment should be recommended. In support of this view, prolonged anti-angiogenic therapy has resulted in prolonged survival for human cancer patients. Originally, researchers believed that angiogenesis inhibitors, especially the endogenous inhibitors such as angiostatin, endostatin, and other generic inhibitors, would not develop drug resistance of tumor cells because they target endothelial cells rather than tumor cells [ 46 , 47 ].

Unlike malignant cells, endothelial cells, even those located in tumor tissues, have stable genomes and do not seem to use the canonical drug-resistant mechanisms.

However, both experimental and clinical findings have challenged this view. Some studies showed that endothelial cells in angiogenic tumor vessels contain aberrant genomes that would not be present in healthy vasculatures [ 48 ]. It is unclear if the tumor-like aberrant genetic information in endothelial cells is transferred from tumor cells or if the intrinsic development genomic instability develops in endothelial cells.

Inhibition of tumor angiogenesis could alter the cellular and molecular components in the tumor microenvironment, leading to development of drug resistance. For example, anti-angiogenic drug—induced vascular regression in the tumors creates tissue hypoxia in a local microenvironment, which augments expression levels of multiple angiogenic factors unrelated to the drug targets [ 36 , 49 ].

Investigators have shown that anti-VEGF drugs develop resistance of tumor cells by this compensatory mechanism. Moreover, anti-VEGF drugs also tip the balance between various cellular compositions, including inflammatory cells and stromal fibroblasts, which are important sources of cytokines and non-VEGF angiogenic factors that contribute to drug resistance [ 50 , 51 ].

Alternative mechanisms of tumor neovascularization that are not affected by drug targets also contribute to anti-angiogenic drug resistance. For example, vessel co-option, vascular mimicry, intussusception, and vasculogenesis support tumor growth and potentially inhibit anti-angiogenic treatment [ 10 , 37 , 52 — 54 ].

In patients who demonstrate intrinsic resistance to anti-VEGF therapy, non-VEGF angiogenic factors probably stimulate angiogenesis in their tumors. Thus, combination therapeutic approaches that target different angiogenesis signaling pathways would likely be more effective.

Also, patients who take multi-targeted drugs such as TKIs would be less likely to develop drug resistance.

Importantly, cross-communication between different angiogenic signaling pathways can generate synergistic effects, even though expression levels of each individual factor are low. For example, the synergistic effects between FGF receptor 2 and PDGF-BB on angiogenesis promote tumor growth and metastasis [ 55 , 56 ].

In clinical practice, combination therapy represents a major mechanistic challenge [ 57 ]. Why would clinical benefits be achieved by combining anti-angiogenic drugs with chemotherapy? Why would anti-angiogenic treatment alone be sufficiently effective? A few possible hypotheses may explain the mechanism underlying combination therapy.

One hypothesis suggests that treatment with anti-angiogenic drugs produces a normalized vascular phenotype, which increases vascular perfusion rather than decreases it [ 58 ]. In the presence of chemotherapeutic agents, increased vascular perfusion enables more cytotoxic drugs to reach the tumors, leading to increased tumor cell death.

In other words, when administered with chemotherapeutics, anti-angiogenic treatment inhibits tumor growth. Also, the results of animal tumor models have demonstrated that anti-angiogenic drug-induced vascular normalization occurs within a limited time during treatment i.

The mechanism underlying how combination therapy relates to vascular normalization is a paradox. If anti-angiogenic drugs induce vascular normalization and possibly blood perfusion in tumors, tumor growth would be accelerated. However, in both preclinical cancer models and clinical cancer patients, anti-angiogenic treatment does not promote tumor growth, although some researchers have suggested that the treatment facilitates cancer invasion [ 60 , 61 ].

Another experimental study suggested that the mechanism underlying combination therapy can be explained by a decrease of chemotherapeutic toxicity [ 62 ]. Chemotherapeutics produce a broad spectrum of toxicity, including suppression of bone marrow hematopoiesis and high levels of circulating VEGF.

Many cancer patients have high levels of circulating VEGF and manifest anemia [ 63 ]. A causal relationship between VEGF and anemia in human cancer patients has yet to be established, but studies of animal cancer models have shown that tumor-derived high-circulating VEGF causes severe anemia [ 44 ].

In high VEGF-producing tumor-bearing mice, chemotherapy and VEGF synergistically suppressed bone marrow hematopoiesis, resulting in early death [ 62 ]. Anti-VEGF treatment ablates VEGF-induced anemia and thus increases tolerance of chemotoxicity.

Anti-angiogenic drugs recover bone marrow hematopoiesis prior to chemotherapy and increase tolerance of chemotoxicity [ 62 ]. If this regimen were approved for at least a subset of human cancer patients, it would probably result in substantially increased survival benefits for these patients.

Mono-specific anti-VEGF drugs such as bevacizumab target only VEGF without binding to other proteins. VEGF expression levels would serve as a reliable predictive marker for selecting cancer patients who are likely to benefit from anti-VEGF therapy.

Based on more than 10 years of clinical experience with various cancer types, simply measuring VEGF expression levels, in either the circulation or tumor biopsies, has not fulfilled the criterion for predicting responders [ 64 — 68 ].

Why would VEGF, as the sole target for bevacizumab, not serve as a reliable predictive marker for patient selection? There is no satisfactory answer to this puzzling question.

However, some researchers have suggested that measuring different isoforms of VEGF might more reliably predict responders of anti-VEGF therapy [ 69 — 71 ]. Smaller VEGF isoforms, including VEGF, lack heparin-binding affinity and diffuse distally from their productive sites.

Additionally, proteolytically processed smaller versions of VEGF can also lack high heparin-binding affinity and can be transported to distal tissues and organs. Interestingly, these small versions of VEGF proteins have some predictive values, although their targets may not be limited to tumor tissues.

It is possible that off-tumor targets of these small VEGF proteins predict their therapeutic values [ 22 ]. Indeed, based on preclinical and clinical findings, the potentially beneficial effects of anti-VEGF drug off-tumor targets have been proposed [ 22 ].

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.

For example, anti-angiogenic drug-induced hypertension has been associated with clinical benefits; however, the molecular mechanism underlying the benefit is unknown [ 72 — 78 ]. Given that adding anti-angiogenic components to conventional chemotherapy is widely used for the treatment of cancer, significant clinical benefits without selection biomarkers are truly valuable.

Assuming a reliable predictive biomarker exists, treating a selected population of responders with anti-angiogenic drugs would likely markedly increase clinical benefits. Future efforts should focus on identifying such a reliable biomarker for clinical use.

Systemic delivery of anti-angiogenic drugs to cancer patients would inevitably expose non-cancerous healthy tissues to these drugs [ 40 , 41 ]. In preclinical studies, investigators have shown that systemic treatment induces vascular changes in multiple tissues and organs.

Additionally, anti-VEGF therapy caused a marked reduction in micro vasculatures in the liver, kidney, and gastrointestinal wall [ 40 , 41 ]. Vascular changes in non-tumor tissues are associated with clinical adverse effects, including hypertension, hypothyroidism, gastrointestinal perforation, and cardiovascular disease [ 15 , 79 ].

Since VEGF is an important hemostatic factor for maintaining the number and structure of microvessels in various tissues and organs, it is perhaps not surprising that anti-VEGF-based anti-angiogenic drugs would cause broad adverse effects.

How would anti-angiogenic drugs be directly delivered to tumorous tissues without affecting the healthy vasculature? Designing a new generation of targeted drugs would be a very challenging task.

Even though anti-angiogenic drugs are locally injected into tumorous tissues, they still enter the circulation. Additionally, this approach would prevent anti-angiogenic agents from reaching metastatic tumors.

In fact, clinical indications of using anti-angiogenic therapies approved by the U. FDA often include metastatic disease. Inhibition of angiogenesis for the treatment of cancer has been successfully translated into clinical use.

The key issue is that patients who receive anti-angiogenic drugs experience relatively few clinical benefits. For patients with some cancer types, including pancreatic cancer and breast cancer, the addition of an anti-angiogenic component to chemotherapy has not produced meaningful improvement in overall survival.

If all solid tumor growth depends on angiogenesis, why would anti-angiogenic treatments not be beneficial? Why would anti-angiogenic monotherapies fail to demonstrate clinical benefits? What is the mechanistic rationale of combination therapy with chemotherapeutics? Could a predictive marker be identified?

How long should cancer patients be treated with anti-angiogenic drugs? What could happen if the anti-angiogenic therapy is discontinued? Would combinations of drugs that target different angiogenic pathways improve therapeutic outcomes?

There are no unified opinions on these clinical issues. Possibly, an important means to address these issues is to establish clinically relevant cancer models in animals. Given sophisticated cancer biology, metastatic disease, and systemic disorders in cancer patients, the complex mechanisms underlying malignant disease cannot likely be simply explained.

The same type of cancer in different patients may represent a different disease. Likewise, the same cancer in the same patient may represent a different disease at different stages of progression. This means that personalized medicine may not be sufficiently effective and that dynamic approaches should be developed for treating cancer at different stages during disease development.

In clinical practice, developing both personalized therapy and dynamic therapy is an extremely challenging task. Forty-year journey of angiogenesis translational research. Sci Transl Med. Article PubMed Google Scholar. Folkman J. Tumor angiogenesis: therapeutic implications.

N Engl J Med. Article CAS PubMed Google Scholar. Angiogenesis: an organizing principle for drug discovery? Nat Rev Drug Discov. Cao Y, Langer R. Optimizing the delivery of cancer drugs that block angiogenesis.

Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma.

Cao Y, Xue L. Semin Thromb Hemost. Cao Y. Endogenous angiogenesis inhibitors and their therapeutic implications. Int J Biochem Cell Biol. Folberg R, Hendrix MJ, Maniotis AJ.

Vasculogenic mimicry and tumor angiogenesis. Am J Pathol. Article PubMed Central CAS PubMed Google Scholar. Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Bissell MJ. Tumor plasticity allows vasculogenic mimicry, a novel form of angiogenic switch.

A rose by any other name? Ribatti D, Vacca A, Dammacco F. New non-angiogenesis dependent pathways for tumour growth. Eur J Cancer. Leenders WP, Kusters B, de Waal RM. Vessel co-option: how tumors obtain blood supply in the absence of sprouting angiogenesis.

Dome B, Paku S, Somlai B, Timar J. 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. Personalized prescription of tyrosine kinase inhibitors in unresectable metastatic cholangiocarcinoma. Exp Hematol Oncol. Motzer RJ, Escudier B, Gannon A, Figlin RA. Sunitinib: ten years of successful clinical use and study in advanced renal cell carcinoma.

Demetri GD, van Oosterom AT, Garrett CR, Blackstein ME, Shah MH, Verweij J, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial.

Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Oudard S, et al.

Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, et al.

Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. Frampton JE. Pazopanib: a review in advanced renal cell carcinoma.

Target Oncol. Article PubMed Google Scholar. Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial.

Motzer RJ, Hutson TE, Cella D, Reeves J, Hawkins R, Guo J, et al. Pazopanib versus sunitinib in metastatic renal-cell carcinoma. Guo J, Jin J, Oya M, Uemura H, Takahashi S, Tatsugami K, et al. Safety of pazopanib and sunitinib in treatment-naive patients with metastatic renal cell carcinoma: Asian versus non-Asian subgroup analysis of the COMPARZ trial.

J Hematol Oncol. Escudier B, Porta C, Bono P, Powles T, Eisen T, Sternberg CN, et al. Randomized, controlled, double-blind, cross-over trial assessing treatment preference for pazopanib versus sunitinib in patients with metastatic renal cell carcinoma: PISCES study.

Zarrabi K, Fang C, Wu S. New treatment options for metastatic renal cell carcinoma with prior anti-angiogenesis therapy. van der Graaf WT, Blay JY, Chawla SP, Kim DW, Bui-Nguyen B, Casali PG, et al.

Pazopanib for metastatic soft-tissue sarcoma PALETTE : a randomised, double-blind, placebo-controlled phase 3 trial. Article PubMed CAS Google Scholar. Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma AXIS : a randomised phase 3 trial.

Motzer RJ, Escudier B, Tomczak P, Hutson TE, Michaelson MD, Negrier S, et al. Axitinib versus sorafenib as second-line treatment for advanced renal cell carcinoma: overall survival analysis and updated results from a randomised phase 3 trial. Lancet Oncol. Keating GM. Axitinib: a review in advanced renal cell carcinoma.

de la Fouchardiere C. Regorafenib in the treatment of metastatic colorectal cancer. Future Oncol. Grothey A, Van Cutsem E, Sobrero A, Siena S, Falcone A, Ychou M, et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer CORRECT : an international, multicentre, randomised, placebo-controlled, phase 3 trial.

Li J, Qin S, Xu R, Yau TC, Ma B, Pan H, et al. Regorafenib plus best supportive care versus placebo plus best supportive care in Asian patients with previously treated metastatic colorectal cancer CONCUR : a randomised, double-blind, placebo-controlled, phase 3 trial.

Demetri GD, Reichardt P, Kang YK, Blay JY, Rutkowski P, Gelderblom H, et al. Efficacy and safety of regorafenib for advanced gastrointestinal stromal tumours after failure of imatinib and sunitinib GRID : an international, multicentre, randomised, placebo-controlled, phase 3 trial.

Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment RESORCE : a randomised, double-blind, placebo-controlled, phase 3 trial.

Medavaram S, Zhang Y. Emerging therapies in advanced hepatocellular carcinoma. Exp Hematol Onco. Article Google Scholar. Ranieri G, Marech I, Asabella AN, Di Palo A, Porcelli M, Lavelli V, et al. Int J Mol Sci. Elisei R, Schlumberger MJ, Muller SP, Schoffski P, Brose MS, Shah MH, et al.

Cabozantinib in progressive medullary thyroid cancer. Priya SR, Dravid CS, Digumarti R, Dandekar M. Targeted therapy for medullary thyroid cancer: A review. Front Oncol. Choueiri TK, Escudier B, Powles T, Mainwaring PN, Rini BI, Donskov F, et al.

Cabozantinib versus everolimus in advanced renal-cell carcinoma. Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring PN, Rini BI, et al. Cabozantinib versus everolimus in advanced renal cell carcinoma METEOR : final results from a randomised, open-label, phase 3 trial.

Abou-Alfa GK, Meyer T, Cheng AL, El-Khoueiry AB, Rimassa L, Ryoo BY, et al. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. Dhillon S. Nintedanib: a review of its use as second-line treatment in adults with advanced non-small cell lung cancer of adenocarcinoma histology.

Nintedanib: A review of its use in patients with idiopathic pulmonary fibrosis. Richeldi L, Costabel U, Selman M, Kim DS, Hansell DM, Nicholson AG, et al.

Efficacy of a tyrosine kinase inhibitor in idiopathic pulmonary fibrosis. Richeldi L, du Bois RM, Raghu G, Azuma A, Brown KK, Costabel U, et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. Alshangiti A, Chandhoke G, Ellis PM.

Antiangiogenic therapies in non-small-cell lung cancer. Curr Oncol. Reck M, Kaiser R, Mellemgaard A, Douillard JY, Orlov S, Krzakowski M, et al. Docetaxel plus nintedanib versus docetaxel plus placebo in patients with previously treated non-small-cell lung cancer LUME-Lung 1 : a phase 3, double-blind, randomised controlled trial.

Hanna NH, Kaiser R, Sullivan RN, Aren OR, Ahn MJ, Tiangco B, et al. Nintedanib plus pemetrexed versus placebo plus pemetrexed in patients with relapsed or refractory, advanced non-small cell lung cancer LUME-Lung 2 : a randomized, double-blind, phase III trial.

Lung Cancer. Yeung KT, Cohen EE. Lenvatinib in advanced, radioactive iodine-refractory, differentiated thyroid carcinoma. Clin Cancer Res. Scott LJ. Lenvatinib: first global approval. Schlumberger M, Tahara M, Wirth LJ, Robinson B, Brose MS, Elisei R, et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer.

Motzer RJ, Hutson TE, Glen H, Michaelson MD, Molina A, Eisen T, et al. Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, open-label, multicentre trial.

Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Forner A, Reig M, Bruix J. Hepatocellular carcinoma. Aoyama T, Yoshikawa T.

Targeted therapy: apatinib - new third-line option for refractory gastric or GEJ cancer. Nat Rev Clin Oncol. Tian S, Quan H, Xie C, Guo H, Lu F, Xu Y, et al. YND1 is a novel and selective inhibitor of vascular endothelial growth factor receptor-2 tyrosine kinase with potent activity in vitro and in vivo.

Cancer Sci. Li J, Qin S, Xu J, Xiong J, Wu C, Bai Y, et al. Randomized, double-blind, placebo-controlled phase III trial of apatinib in patients with chemotherapy-refractory advanced or metastatic adenocarcinoma of the stomach or gastroesophageal junction.

Syed YY. Anlotinib: first global approval. Han BH, Li K, Zhao YZ, Li BL, Cheng Y, Zhou JY, et al. Anlotinib as a third-line therapy in patients with refractory advanced non-small-cell lung cancer: a multicentre, randomised phase II trial ALTER Br J Cancer.

Han B, Li K, Wang Q, Zhang L, Shi J, Wang Z, et al. Effect of anlotinib as a third-line or further treatment on overall survival of patients with advanced non-small cell lung cancer: the ALTER phase 3 randomized clinical trial.

JAMA Oncol. Shen G, Zheng F, Ren D, Du F, Dong Q, Wang Z, et al. Anlotinib: a novel multi-targeting tyrosine kinase inhibitor in clinical development. Su S, Wu YL. Clinical trials of tyrosine kinase inhibitors for lung cancer in China: a review.

Xu RH, Li J, Bai Y, Xu J, Liu T, Shen L, et al. Safety and efficacy of fruquintinib in patients with previously treated metastatic colorectal cancer: a phase Ib study and a randomized double-blind phase II study. Li J, Qin S, Xu RH, Shen L, Xu J, Bai Y, et al. Effect of fruquintinib vs placebo on overall survival in patients with previously treated metastatic colorectal cancer: the FRESCO randomized clinical trial.

Shirley M. Fruquintinib: first global approval. Polverino A, Coxon A, Starnes C, Diaz Z, DeMelfi T, Wang L, et al. AMG , an oral, multikinase inhibitor that selectively targets vascular endothelial growth factor, platelet-derived growth factor, and kit receptors, potently inhibits angiogenesis and induces regression in tumor xenografts.

Cancer Res. Kubota K, Ichinose Y, Scagliotti G, Spigel D, Kim JH, Shinkai T, et al. Ann Oncol. Kubota K, Yoshioka H, Oshita F, Hida T, Yoh K, Hayashi H, et al. Phase III, randomized, placebo-controlled, double-blind trial of motesanib AMG in combination with paclitaxel and carboplatin in east Asian patients with advanced nonsquamous non-small-cell lung cancer.

Schlumberger MJ, Elisei R, Bastholt L, Wirth LJ, Martins RG, Locati LD, et al. Phase II study of safety and efficacy of motesanib in patients with progressive or symptomatic, advanced or metastatic medullary thyroid cancer. Sherman SI, Wirth LJ, Droz JP, Hofmann M, Bastholt L, Martins RG, et al.

Motesanib diphosphate in progressive differentiated thyroid cancer. Lubner S, Feng Y, Mulcahy M, O'Dwyer P, Giang GY, Hinshaw JL, et al.

E AMG and octreotide in patients with low-grade neuroendocrine tumors. Mahner S, Woelber L, Mueller V, Witzel I, Prieske K, Grimm D, et al.

Beyond bevacizumab: an outlook to new anti-angiogenics for the treatment of ovarian cancer. Goss GD, Arnold A, Shepherd FA, Dediu M, Ciuleanu TE, Fenton D, et al.

Randomized, double-blind trial of carboplatin and paclitaxel with either daily oral cediranib or placebo in advanced non-small-cell lung cancer: NCIC clinical trials group BR24 study.

Hoff PM, Hochhaus A, Pestalozzi BC, Tebbutt NC, Li J, Kim TW, et al. Schmoll HJ, Cunningham D, Sobrero A, Karapetis CS, Rougier P, Koski SL, et al. Cediranib with mFOLFOX6 versus bevacizumab with mFOLFOX6 as first-line treatment for patients with advanced colorectal cancer: a double-blind, randomized phase III study HORIZON III.

Batchelor TT, Mulholland P, Neyns B, Nabors LB, Campone M, Wick A, et al. Phase III randomized trial comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, versus lomustine alone in patients with recurrent glioblastoma. Ledermann JA, Embleton AC, Raja F, Perren TJ, Jayson GC, Rustin GJS, et al.

Cediranib in patients with relapsed platinum-sensitive ovarian cancer ICON6 : a randomised, double-blind, placebo-controlled phase 3 trial. Mullen MM, Kuroki LM, Thaker PH. Novel treatment options in platinum-sensitive recurrent ovarian cancer: a review.

Gynecol Oncol. Xu JM, Wang Y, Chen YL, Jia R, Li J, Gong JF, et al. Sulfatinib, a novel kinase inhibitor, in patients with advanced solid tumors: results from a phase I study. PubMed PubMed Central Google Scholar. Grillo F, Florio T, Ferrau F, Kara E, Fanciulli G, Faggiano A, et al.

Emerging multitarget tyrosine kinase inhibitors in the treatment of neuroendocrine neoplasms. Endocr Relat Cancer. Yu S, Li A, Liu Q, Li T, Yuan X, Han X, et al. Chimeric antigen receptor T cells: a novel therapy for solid tumors. Li J, Li W, Huang K, Zhang Y, Kupfer G, Zhao Q.

Chimeric antigen receptor T cell CAR-T immunotherapy for solid tumors: lessons learned and strategies for moving forward. Li Z, Song W, Rubinstein M, Liu D.

Recent updates in cancer immunotherapy: a comprehensive review and perspective of the China cancer immunotherapy workshop in Beijing. Yi M, Jiao D, Xu H, Liu Q, Zhao W, Han X, et al.

Choueiri TK, Larkin J, Oya M, Thistlethwaite F, Martignoni M, Nathan P, et al. Preliminary results for avelumab plus axitinib as first-line therapy in patients with advanced clear-cell renal-cell carcinoma JAVELIN renal : an open-label, dose-finding and dose-expansion, phase 1b trial.

Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. Kudo M. Systemic therapy for hepatocellular carcinoma: latest advances. Cancers Basel. Giuliano S, Pages G. Mechanisms of resistance to anti-angiogenesis therapies.

Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, et al. Renal cell carcinoma. Nat Rev Dis Primers. Chen Y, Chi P. Basket trial of TRK inhibitors demonstrates efficacy in TRK fusion-positive cancers.

Liu X, Qin S, Wang Z, Xu J, Xiong J, Bai Y, et al. Early presence of anti-angiogenesis-related adverse events as a potential biomarker of antitumor efficacy in metastatic gastric cancer patients treated with apatinib: a cohort study.

Rini BI, Schiller JH, Fruehauf JP, Cohen EE, Tarazi JC, Rosbrook B, et al. Diastolic blood pressure as a biomarker of axitinib efficacy in solid tumors. Haas NB, Manola J, Dutcher JP, Flaherty KT, Uzzo RG, Atkins MB, et al. Adjuvant treatment for high-risk clear cell renal cancer: updated results of a high-risk subset of the ASSURE randomized trial.

Motzer RJ, Haas NB, Donskov F, Gross-Goupil M, Varlamov S, Kopyltsov E, et al. Randomized phase III trial of adjuvant pazopanib versus placebo after nephrectomy in patients with localized or locally advanced renal cell carcinoma.

Ravaud A, Motzer RJ, Pandha HS, George DJ, Pantuck AJ, Patel A, et al. Adjuvant sunitinib in high-risk renal-cell carcinoma after nephrectomy. Bruix J, Takayama T, Mazzaferro V, Chau GY, Yang J, Kudo M, et al.

Adjuvant sorafenib for hepatocellular carcinoma after resection or ablation STORM : a phase 3, randomised, double-blind, placebo-controlled trial. Powles T, Sarwar N, Stockdale A, Sarker SJ, Boleti E, Protheroe A, et al. Safety and efficacy of pazopanib therapy prior to planned nephrectomy in metastatic clear cell renal cancer.

Ramjiawan RR, Griffioen AW, Duda DG. Anti-angiogenesis for cancer revisited: is there a role for combinations with immunotherapy? Download references. This work was funded by the National Natural Science Foundation of China No.

Wuhan Science and Technology Bureau No. Data sharing not applicable to this article as no datasets were generated or analyzed during the current study. Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Wuhan, , Hubei, China.

Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, , Henan, China. You can also search for this author in PubMed Google Scholar. SQ and AL performed the selection of literature, drafted the manuscript, and prepared the figures. MY and SY collected the related references.

MZ participated in the design and discussion. KW carried out the design of this review and revised the manuscript. All authors contributed to this manuscript. All authors read and approved the final manuscript.

Correspondence to Kongming Wu. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Open Access This article is distributed under the terms of the Creative Commons Attribution 4. Reprints and permissions. Qin, S. et al. Recent advances on anti-angiogenesis receptor tyrosine kinase inhibitors in cancer therapy.

J Hematol Oncol 12 , 27 Download citation. Received : 21 January Accepted : 05 March

Angiogenesis Inhibitors - NCI Folkman, J. Google Scholar Meert A-P, Paesmans M, Martin B, Delmotte P, Berghmans T, Verdebout J-M, et al. If all solid tumor growth depends on angiogenesis, why would anti-angiogenic treatments not be beneficial? Received : 18 November Article PubMed Google Scholar Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, et al. An important family of proteinases essential for the degradation of the extracellular matrix ECM and the basement membrane during angiogenesis include the family of matrix metalloproteinases MMPs Liotta and Stetler-Stevenson,

Anti-angiogenesis clinical trials -

The results of this phase III randomized trial showed that the addition of bevacizumab to capecitabine in second-line therapy improved response rate compared to capecitabine treatment alone However, this combination neither improved progression-free survival PFS median, 4. The E was an open-label, randomized, phase III clinical trial that investigated the efficacy and safety of the combination of paclitaxel and bevacizumab compared to paclitaxel as a first-line treatment for metastatic breast cancer Miller et al.

Findings from the E trial indicated a significantly improved PFS for the combination arm compared to paclitaxel alone, while OS was similar in both treatment arms median, The results from the AVFG and E trials granted accelerated approval of bevacizumab use in the treatment of metastatic breast cancer by the US Food and Drug Administration FDA.

Subsequently, several phase III clinical trials have evaluated bevacizumab with chemotherapy revealing no significant improvement in OS Miles et al. The failure of achieving a survival advantage along with serious tolerability issues created controversy over the real value of bevacizumab treatment in metastatic breast cancer and further brought its approval into question.

Ultimately, the US FDA had revoked the indication of bevacizumab to treat patients with metastatic breast cancer in Sasich and Sukkari, After the withdrawal statement, results from other clinical trials on the use of bevacizumab in breast cancer were published.

The BEATRICE study was an open-label, randomized, phase III clinical trial that assessed the addition of bevacizumab to chemotherapy in adjuvant settings in patients with operable TNBC Cameron et al. Results from the BEATRICE study revealed no improvement of OS compared to patients receiving chemotherapy alone Cameron et al.

Rather, grade III adverse events were increased in the bevacizumab arm Cameron et al. The E study was a double-blind, phase III trial of adjuvant chemotherapy with and without bevacizumab in breast cancer patients with lymph node-positive and high-risk lymph node-negative disease Miller et al. The findings of the study failed to show improvements in invasive disease-free survival or OS upon the addition of bevacizumab to chemotherapy in adjuvant settings in breast cancer patients with high-risk HER2-negative disease Miller et al.

Other studies investigated the value of adding bevacizumab to chemotherapy in the neoadjuvant treatment of breast cancer Bear et al. The primary endpoint in these studies was the pathologic complete response pCR rate.

Overall, findings from these clinical trials showed a favorable response for using bevacizumab with chemotherapy in neoadjuvant treatment in terms of increased pCR rates Bear et al. Recently, Martin et al. examined the addition of bevacizumab to endocrine drugs as first-line treatment in metastatic hormone receptor-positive breast cancer through pooled data analysis from the LEA and CALGB trials Martin et al.

The addition of bevacizumab significantly improved PFS compared to endocrine treatment alone, however, there was no difference in OS between both groups. Besides, a phase II trial of nab-paclitaxel and bevacizumab, followed by maintenance therapy with bevacizumab and erlotinib, for patients with metastatic TNBC was conducted by Symonds et.

Symonds et al. No significant difference was seen for either PFS or OS for patients enrolled and none of them achieved complete response Symonds et al. Ramucirumab Cyramza ® is a monoclonal antibody targeting VEGFR-2 Singh and Parmar, It was first approved by the US FDA in as monotherapy for the treatment of metastatic gastric cancer Casak et al.

Ramucirumab approval was thereafter expanded to combination treatment with chemotherapy for gastric cancer, non-small cell lung cancer NSCLC , and colon cancer. The drug is also approved as monotherapy for hepatocellular carcinoma HCC , and most recently in combination with erlotinib for patients with epidermal growth factor receptor EGFR -positive NSCLC Effing and Gyawali, Few clinical trials investigated ramucirumab treatment in breast cancer.

In phase II, randomized, open-label study, the addition of ramucirumab to capecitabine in previously treated patients with locally advanced and metastatic breast cancer failed to improve PFS and OS compared to capecitabine therapy alone Vahdat et al.

The frequency of adverse effects was increased in the combination group and included headache, anorexia, constipation, epistaxis, and hypertension Vahdat et al.

Another phase II, randomized, open-label clinical trial revealed no difference in survival for the combination of ramucirumab and eribulin versus eribulin monotherapy in patients with advanced breast cancer Yardley et al.

In agreement with findings from previous phase II trials, ramucirumab neither improved PFS nor OS compared to docetaxel treatment median, 9. Higher rates of toxicity were reported in patients receiving ramucirumab treatment Mackey et al.

Tyrosine kinase inhibitors TKIs are small molecules that inhibit the kinase domain of RTKs thus inhibiting receptor activation and downstream signaling Fakhrejahani and Toi, Several angiogenesis inhibitors are small-molecule TKIs.

Sorafenib Nexavar ® is an oral multikinase inhibitor of VEGFR, PDGF receptor PDGFR , and Raf Ben Mousa, The drug is approved for the treatment of renal cell carcinoma RCC Kane et al. The use of sorafenib in breast cancer showed modest efficacy in early clinical trials.

In a randomized, double-blind, placebo-controlled, phase IIb trial, the addition of sorafenib to capecitabine showed higher toxicity and no improvement in OS in patients with locally advanced or metastatic HER2-negative breast cancer Baselga et al.

In another phase IIb, double-blind, randomized, placebo-controlled study, the addition of sorafenib to capecitabin or gemcitabine moderately improved PFS compared to placebo and chemotherapy in breast cancer patients median, 3.

Nevertheless, no significant effect was observed for OS for patients in the sorafenib arm Schwartzberg et al. Despite the modest effects for sorafenib treatment observed in terms of improved PFS, subsequent clinical trials failed to confirm the survival advantage for the drug in breast cancer treatment.

Nevertheless, rates of grade III toxicities were notably higher in the sorafenib arm Baselga et al. In the PASO trial, the safety and efficacy of adding sorafenib to paclitaxel compared to paclitaxel alone were assessed in an open-label, randomized, phase II study design in patients with locally advanced or metastatic HER2-negative breast cancer Decker et al.

Interestingly, a pre-planned efficacy interim analysis showed that patients on paclitaxel monotherapy had a significantly greater PFS and OS compared to patients in the combination arm. Toxicities were higher in the combination arm and the study was therefore discontinued Decker et al.

Similarly, results from the MADONNA study, a multicenter, double-blind, phase II study, revealed a lack of survival advantage upon the addition of sorafenib to docetaxel as first-line treatment in breast cancer patients with metastatic or locally advanced HER2-negative disease Mavratzas et al.

Recently, Ianza et al. showed no difference in survival outcomes for adding sorafenib to letrozole and cyclophosphamide in postmenopausal patients with locally advanced estrogen receptor ER -positive, HER2-negative breast cancer in a phase III trial Ianza et al.

Interestingly, a higher percentage of patients on sorafenib treatment had disease progression Ianza et al. Other clinical studies have constantly supported a lack of survival advantage for the combination of sorafenib and chemotherapy in patients with advanced breast cancer Gradishar et al.

Sunitinib Sutent ® is a novel oral multitargeted TKI of VEGFR-1, VEGFR-2, fetal liver tyrosine kinase receptor 3, c-KIT, PDGFR-α, and PDGFR-β Le Tourneau et al. Sunitinib is FDA approved for the treatment of advanced RCC, gastrointestinal stromal tumors Le Tourneau et al.

Clinical studies for the efficacy of sunitinib in breast cancer treatment have produced disappointing results Yardley et al. In , a multicenter, randomized, open-label, phase III trial was conducted to assess the effect of sunitinib versus capecitabine as a treatment for advanced breast cancer patients with disease recurrence after anthracycline and taxane therapy Barrios et al.

Compared to the capecitabine arm, PFS and OS were shorter for sunitinib median, 2. Sunitinib treatment was also associated with a higher rate and severity of adverse events compared with capecitabine.

Bergh et al. demonstrated no improvement in PFS or OS in breast cancer patients treated with the combination of sunitinib and docetaxel compared to docetaxel alone in an open-label, randomized, phase III trial Bergh et al. Moreover, more adverse events, deaths, and treatment discontinuations were observed in the combination arm Bergh et al.

In a randomized, phase II trial comparing sunitinib to the standard of care in patients with TNBC who relapsed after anthracycline- and taxane-based chemotherapy, no difference between both treatment arms for PFS and OS was observed Curigliano et al. In a randomized, phase III study by Crown et al.

The combination arm failed to improve therapeutic outcomes compared to the capecitabine arm as no statistically significant difference was observed for PFS median, 5.

Vandetanib Caprelsa ® is an oral small-molecule inhibitor of VEGFR-2, VEGFR-3, EGFR, and RET Chau and Haddad, It is approved for the treatment of medullary thyroid carcinoma Chau and Haddad, The efficacy and safety of vandetanib with docetaxel as a second-line treatment for advanced breast cancer was assessed in a double-blind, placebo-controlled, randomized, phase II study Boer et al.

Though well-tolerated, the combination of vandetanib and docetaxel did not improve outcomes compared to placebo and docetaxel Boer et al.

Clemons et al. also showed no difference in PFS or OS for the combination of vandetanib and fulvestrant compared to placebo in postmenopausal patients with metastatic breast cancer in a phase II trial Clemons et al.

Axitinib Inlyta ® is an oral second-generation pan-VEGFR TKI Bellesoeur et al. The drug is approved for the treatment of advanced RCC Tyler, Rugo et al. assessed the efficacy of axitinib plus docetaxel compared to docetaxel and placebo in metastatic breast cancer in a randomized, double-blind, phase II study Rugo et al.

The addition of axitinib to capecitabine did not significantly improve time to progression compared to the placebo arm median, 8. Pazopanib Votrient ® is an oral multitarget TKI of VEGFR, PDGFRs, FGFR, and c-KIT Lee et al. It is approved for the treatment of advanced RCC and advanced soft-tissue sarcoma Nguyen and Shayahi, In , results from a phase II study of single-agent pazopanib in patients with recurrent or metastatic breast cancer revealed promising activity in terms of disease stability and tolerable adverse events Taylor et al.

Nevertheless, subsequent phase II trials for the combination of lapatinib and pazopanib in HER2-positive breast cancer failed to show survival advantage compared to lapatinib alone. The combination also had increased toxicity compared to lapatinib monotherapy Cristofanilli et al.

The addition of pazopanib to chemotherapy in neoadjuvant treatment for HER2-negative locally advanced breast cancer was assessed in a phase II study, however substantial toxicity resulted in a high discontinuation rate of pazopanib Tan et al.

Cediranib Recentin ® is a pan-VEGFR inhibitor Tang et al. It has been assessed in combination with hormonal treatments in breast cancer patients. A randomized, phase II study evaluated cediranib plus fulvestrant in postmenopausal women with hormone-sensitive metastatic breast cancer compared to placebo Hyams et al.

The addition of cediranib to fulvestrant did not improve median PFS versus placebo. Furthermore, the rates of grade III adverse events, discontinuations, and dose reductions were higher in the cediranib arm Hyams et al.

The available experimental evidence, which is not yet definitive, proposes several distinct mechanisms that manifest tumor rescue pathways to anti-angiogenic therapies. Several mechanisms for intrinsic and acquired resistance to angiogenesis inhibitors have now been explored.

Some of these mechanisms are discussed below. The activation of compensatory pro-angiogenic pathways in response to anti-VEGF therapy is a well-established mechanism of acquired resistance in tumors Bergers and Hanahan, ; Ramadan et al. Typically, vasculogenesis is a minor pathway in the development of tumor vasculature at which angiogenesis is the primary pathway.

However, upon the inhibition of angiogenic growth, vasculogenesis may become crucial to maintaining tumor vasculature Brown, Hypoxia-induced by anti-VEGF therapy leads to the recruitment of pro-angiogenic bone marrow-derived cells BMDCs to the tumor microenvironment Lord and Harris, ; Ramadan et al.

BMDCs can restore vascularization of tumors thus enabling them to overcome hypoxia and become resistant to anti-VEGF drugs Bergers and Hanahan, ; Lord and Harris, Several BMDCs have been identified in the tumor microenvironment such as tumor-associated macrophages TAMs , pro-angiogenic monocytic cells, myeloid cells, and Tieexpressing macrophages Lord and Harris, TAMs were associated with high VEGF expression and high microvessel density in ductal breast carcinoma Longatto Filho et al.

Tripathi et al. revealed that TAMs were recruited to tumor microenvironment in an animal model of breast cancer by eotaxin and oncostatin M cytokines Tripathi et al. Blocking these cytokines with neutralizing antibodies reduced tumor vascularization and improved sensitivity to bevacizumab Tripathi et al.

Liu et al. Obesity was associated with increased IL-6 production from adipocytes and myeloid cells within tumors in murine breast cancer model Incio et al. Inhibition of IL-6 normalized tumor vasculature, reduced hypoxia, and restored sensitivity to anti-VEGF therapy.

Heterogeneous pericyte coverage has been described in several types of tumors, at different stages of tumor progression, and even within a single tumor stage Hida et al. The reduction in tumor vascularity induced by anti-VEGF therapy enhances the recruitment of pericytes to maintain blood vessel function and integrity Bergers and Hanahan, Increased pericyte coverage of these blood vessels supports tumor endothelium to survive and function despite the anti-angiogenic drug Bergers and Hanahan, ; Lord and Harris, In addition, pericytes can release pro-angiogenic factors in response to PDGF Lord and Harris, In the breast cancer vasculature, heterogenous pericyte coverage was identified Kim et al.

However, the impact of pericyte on resistance to anti-VEGF therapy in breast tumors is largely unknown. Vasculogenic mimicry and vessel co-option may decrease the dependence on classical angiogenesis by tumors Schneider and Miller, ; Bergers and Hanahan, ; Carmeliet and Jain, These alternative mechanisms render tumors insensitive to anti-angiogenic agents by allowing tumors to obtain the necessary blood supply when classical angiogenesis is limited Schneider and Miller, ; Haibe et al.

Vasculogenic mimicry is associated with aggressive breast cancer phenotypes and poor prognosis Shen et al. Bevacizumab failed to inhibit vasculogenic mimicry in the HCC breast cancer cell line Dey et al. Besides, Sun et al.

showed that the administration of sunitinib induced vasculogenic mimicry in animal models of TNBC which ultimately promoted resistance to sunitinib therapy Sun et al.

Vascular co-option is another mechanism to escape angiogenesis inhibitors and has been shown to drive brain metastasis of breast cancer cells Ramadan et al.

Growing evidence supports the concept of the heterogeneity of the endothelium of vessels involved in angiogenesis Hida et al. Hida et al. showed that tumor blood vessels are heterogeneous and that tumor-associated endothelial cells had relatively large, heterogeneous nuclei, cell aneuploidy, and chromosomal alterations indicative of cytogenetic abnormalities Hida et al.

Altered gene and protein expression profiles in tumor endothelium have also been reported Aird, The heterogeneity of tumor endothelial cells may differ by tumor type, tumor microenvironment, and the stage of tumor growth Hida et al. Grange et al. showed that breast cancer-derived endothelial cells did not undergo normal cell senescence in culture, had increased motility, and constantly expressed markers of endothelial activation and angiogenesis Grange et al.

These endothelial cells were resistant to the cytotoxic activity of chemotherapeutic drugs as compared to normal micro-endothelial cells Grange et al. The functional abnormalities of tumor-associated endothelial cells and the microvascular heterogeneity could explain, at least in part, the reduced efficacy of anti-angiogenic therapy in breast cancer by enabling endothelial cells an increased pro-angiogenic activity to acquire drug resistance Grange et al.

Lack of response to angiogenesis inhibitors may be explained in terms of the stage of progression, treatment history, and genomic constitution that exist in the tumor microenvironment Bergers and Hanahan, An analysis of human breast cancer biopsies demonstrated a plethora of pro-angiogenic factors in late-stage breast cancers including FGF-2, in contrast to earlier-stage tumors which preferentially expressed VEGF Relf et al.

Thus, resistance to anti-VEGF drugs in advanced-stage breast cancer may be explained by the dominance of FGF-2 and other pro-angiogenic factors in such stage of the disease Bergers and Hanahan, Invasive cancers commonly express multiple angiogenic factors and this heterogeneity occurs at an early point in time.

Genetic instabilities in the tumor cells may cause alterations of both the amount and type of pro-angiogenic factors expressed in a tumor which could further promote resistance to anti-angiogenic treatments Schneider and Miller, Cancer stem cells are a subpopulation of cancer cells capable of self-renewal, differentiation, and induction of tumorigenesis, metastasis, and drug resistance Li et al.

The potential of cancer stem cell trans-differentiating into endothelial cells has been reported in a variety of solid tumors Li et al. Bussolati et al. showed that breast cancer stem cells were able to differentiate into the endothelial lineage in the presence of VEGF Bussolati et al.

The stem cells acquired several endothelial markers and organized into capillary-like structures forming vessels in a xenograft animal model Bussolati et al.

Similarly, Wang et al. showed that breast cancer stem cells may trans-differentiate into endothelial cells that can form capillary-like vascular structures in the cell culture system and participate in tumor angiogenesis Wang et al.

An earlier study demonstrated that microRNAa miRNAa expression promoted tumor angiogenesis and metastasis in vivo by mediating endothelial trans-differentiation of breast cancer stem-like cells Tang et al.

Brossa et al. reported the ability of breast cancer stem cells to trans-differentiate to endothelial cells expressing endothelial markers under hypoxic conditions in vitro Brossa et al.

Notably, treatment with the VEGFR inhibitor sunitinib but not the VEGF inhibitor bevacizumab impaired the endothelial differentiation ability of breast cancer stem cells both in vitro and in vivo.

Mechanistically, sunitinib, but not bevacizumab, suppressed HIF-1α required for endothelial differentiation under hypoxic conditions Brossa et al. Together, increasing evidence suggests that cancer stem cell endothelial trans-differentiation supports tumor vascularization and partly contributes to the failure of anti-angiogenic drugs.

The lack of efficacy of the conventional angiogenesis inhibitors necessitates exploring novel angiogenic pathways in breast cancer. Given the heterogeneity of breast cancer and the complexity of angiogenesis, it is unlikely that the identification of a single target such as VEGF would be adequate in the treatment of this disease.

Interleukins ILs are a family of cytokines known to play essential roles in the regulation of several immune cell functions such as differentiation, activation, proliferation, migration, and adhesion Turner et al. Interactions of ILs and their receptors in endothelial cells have been shown to regulate angiogenesis through pro-angiogenic and anti-angiogenic activity Ribatti, Serum IL-6 levels were elevated in breast cancer patients compared to controls Barron et al.

Additionally, serum IL-6 and VEGF correlated positively in breast cancer patients Raghunathachar Sahana et al. Higher expression of IL-6R was demonstrated in clinical specimens for patients with high-grade invasive ductal carcinoma Bharti et al.

A recent study by Hegde et al. showed that a crosstalk between IL-6 and VEGFR-2 signaling pathways exists in myoepithelial and endothelial cells isolated from clinical human breast tumors Hegde et al.

IL-6 epigenetically regulated VEGFR-2 expression through induction of proteasomal degradation of DNA methyltransferase 1 leading to promoter hypomethylation and angiogenic activity Hegde et al.

IL-8 is a pro-inflammatory cytokine that exerts its biologic activity through binding to its CXCR1 and CXCR2 receptors Waugh and Wilson, IL-8 enhanced the proliferation of cancer cells and produced a pro-angiogenic activity Waugh and Wilson, Serum IL-8 levels were significantly higher in breast cancer patients compared with healthy subjects and were associated with advanced disease Benoy et al.

High levels of IL-8 are secreted by stromal cells into the microenvironment of breast cancer patients compared to controls Razmkhah et al. Evidence from preclinical studies showed that IL-8 mediated invasion and angiogenesis of breast cancer cells Lin et al.

Cancer-associated adipocytes express high levels of IL-8 in breast cancer stroma thus promoting the pro-angiogenic effects of breast adipocytes Al-Khalaf et al. In this context, ILexpressing adipocytes increased vascularity of tumor xenografts as indicated by increased expression of CD34, an endothelial cell marker Al-Khalaf et al.

Neutralization of IL-8 or inhibiting its target receptors had been shown to reduce breast cancer growth and angiogenesis Lin et al.

Nannuru et al. showed that silencing of CXCR2 expression reduced tumor vascularity and inhibited spontaneous lung metastasis in an orthotopic animal model of breast cancer Nannuru et al.

Further, CXCR1 blockade with the small molecule inhibitor, repertaxin reduced metastasis in an animal model of breast cancer Ginestier et al. The platelet-derived growth factor PDGF family consists of four gene products PDGF-A, -B, -C, and -D that are combined into five different isoforms: PDGF-AA, -BB, -CC, -DD, and -AB Bartoschek and Pietras, These factors bind and activate their respective RTKs, PDGFR-α, and PDGFR-β.

PDGF family plays a key role in a wide range of oncologic activities essential for cancer growth including angiogenesis, fibrosis, and cellular migration Bartoschek and Pietras, High expression of PDGFs was correlated with an advanced presentation, increased recurrence, and poor survival in patients with invasive breast cancer Jansson et al.

PDGF is an important regulator for the motility of vascular smooth muscle cells induced by breast cancer cells Banerjee et al. Besides, the expression of HIF-1α in invasive breast cancer was significantly associated with angiogenesis and expression of PDGF-BB Bos et al.

Earlier evidence showed that PDGFRs are expressed by breast cancer cells and endothelial cells in metastatic bone lesions in animal models Lev et al. Imatinib remarkably inhibited PDGFR activation in breast cancer cells and tumor-associated endothelial cells and reduced microvessel density in the tumors Lev et al.

Recently, Wang et al. provided evidence from cell culture and animal studies that the downregulation of PDGF-B greatly contributed to the metformin-induced vessel normalization in breast cancer Wang et al.

Fibroblast growth factors FGFs belong to a large family of growth factors that includes 23 members Hui et al. FGFs are key regulators of numerous physiological processes such as angiogenesis, wound healing, and embryonic development. These functions are mediated by the binding of FGFs with their receptors FGFRs , which belong to the RTK family Hui et al.

Growing evidence signifies the oncogenic impact of FGFs and FGFRs to promote cancer development and progression by mediating cancer cell proliferation, survival, epithelial-to-mesenchymal transition, invasion, and angiogenesis Wesche et al.

Chen et al. showed that dipalmitoylphosphatidic acid, a bioactive phospholipid, induced anti-angiogenic activity, and inhibited tumor growth in an experimental xenograft model of breast cancer Chen J. These effects were attributed to transcriptional inhibition of FGF-1 expression leading to the downregulation of HGF Chen J.

In the same context, Cai et al. showed that neutralizing FGF-2 by a disulfide-stabilized diabody inhibited tumor growth and angiogenesis in a mouse model of breast cancer Cai et al. The antitumor activity was associated with a significant decrease in microvessel density and the number of lymphatic vessels Cai et al.

Formononetin, an FGFR-2 inhibitor, demonstrated anti-angiogenic activity in breast cancer in both ex vivo and in vivo angiogenesis assays Wu et al. Besides, formononetin significantly inhibited angiogenesis in vivo by reducing microvessel density and phosphorylated FGFR-2 levels in tumor tissue Wu et al.

Recent evidence showed that FGFpositive tumors are resistant to clinically available drugs targeting VEGF and PDGF Hosaka et al. The resistance is mediated by the ability of FGF-2 to recruit pericytes onto tumor microvessels through a PDGFR-β-dependent mechanism in breast cancer and fibrosarcoma models.

Dual targeting of the VEGF and PDGF produced a superior antitumor effect in FGFpositive breast cancer Hosaka et al. Angiopoietins Angs represent an imperative family of vascular growth factors that produce their biological effects through binding to the RTKs, Tie-1, and Tie-2 Akwii et al.

Angiopoietin-1 Ang-1 and angiopoietin-2 Ang-2 are best characterized for their role in angiogenesis and vascular stability Akwii et al. Ang-1 regulates the organization and maturation of newly formed blood vessels and promotes quiescence and structural integrity of vasculature Brindle et al.

Alternatively, Ang-2 antagonizes the effects of Ang-1 resulting in vessel destabilization Brindle et al. Ramanthan et al. indicated that high Ang-2 gene expression in breast cancer patients was associated with reduced survival Ramanathan et al.

In addition, a strong correlation existed between Angs and VEGF genes in breast cancer tissues Ramanathan et al.

Besides, serum levels of Ang-2 were significantly higher in breast cancer patients compared to healthy control subjects. High Ang-2 serum levels had shorter survival than that of the low Ang-2 expression group Li et al.

Evidence from preclinical models also demonstrated that Ang-2 mediated initial steps of breast cancer metastasis to the brain Avraham et al.

He et al. showed that targeting Ang-2 with miRNAp reduced tumor growth, angiogenesis, and metastasis in animal models He et al. Besides, Wu et al. showed that oral administration of methylseleninic acid reduced microvessel density and increased pericytes coverage by inhibiting Ang-2 in a breast cancer animal model Wu et al.

Dual inhibition of VEGF-A and Ang-2 using a bispecific antibody promoted vascular regression and normalization in a model of metastatic breast cancer Schmittnaegel et al.

Dual inhibition of Ang-1 and TGF-βR2 was also shown to suppress tumor angiogenesis in breast cancer in vivo Flores-Perez et al.

Notch receptors belong to a highly conserved signaling pathway that relies on cell-cell contacts to mediate a response to environmental signals in multicellular animals Aster et al. Four different Notch receptors are expressed in humans, each is encoded by a different gene. In addition, four functional Notch ligands exist and belong to two families: members of the Delta family of ligands; Dll-1 and Dll-4, and members of the Serrate family of ligands; Jag-1 and Jag-2 Aster et al.

In breast cancer, Notch signaling promotes cell proliferation, self-renewal, anti-apoptotic effects, and angiogenesis Aster et al. Notch expression has been associated with the progression and recurrence of breast cancer Mollen et al.

Proia et al. showed that blocking Notch-1 function with a specific antibody inhibited functional angiogenesis and breast cancer growth in animal models Proia et al. HGF is a member of the plasminogen-related growth factor group and is a known angiogenic factor Nakamura and Mizuno, It is primarily expressed and produced by stromal cells, such as fibroblasts in mammary tissues Jiang et al.

The angiogenic actions of HGF are mediated by binding to its RTK, MET on endothelial cells Organ and Tsao, ; Zhang et al. In the activated endothelial cells, MET is upregulated thus modulating cell dissociation, motility, proliferation, and invasion Peruzzi and Bottaro, HGF regulates VEGF expression in tumor cells promoting angiogenic activity Matsumura et al.

Earlier studies showed that targeting HGF with retroviral ribozyme transgene or HGF antagonist reduced the growth and angiogenesis of breast tumors in vivo Jiang et al. Syndecans are transmembrane proteoglycans composed of a core protein and a glycosaminoglycan side chain to which growth factors are attached Szatmari and Dobra, Syndecan-1 is the major syndecan found in epithelial malignancies Szatmari and Dobra, Syndecan-1 ligates with several pro-angiogenic factors such as VEGF, FGFs, Wnt, and HGF, which act as signaling co-receptors Szatmari and Dobra, Expression of syndecan-1 in breast tumors was associated with adverse prognosticators, metastasis, and reduced OS in patients Kind et al.

Besides, stromal syndecan-1 expression increased vessel density and area and promoted the growth and angiogenesis of triple-negative tumors in vivo Maeda et al.

Schönfeld et al. showed that targeting syndecan-1 with an antibody-drug conjugate reduced the growth of TNBC in animal models when combined with chemotherapy Schonfeld et al. An open-label, phase Ib trial evaluating antitumor activity and safety of erdafitinib; a potent and selective pan-FGFR inhibitor, in combination with fulvestrant and palbociclib in patients with metastatic breast cancer is currently recruiting patients NCT The primary objective is to determine safety and tolerability for the combination treatment of erdafitinib with targeted treatments.

Futibatinib is an orally available pan-FGFR inhibitor that is currently being evaluated in a phase II trial as monotherapy and in combination with fulvestrant in patients with locally advanced or metastatic breast cancer harboring FGFR gene amplification NCT The primary outcome of the trial is to determine dose-limiting toxicities during the first two cycles of therapy while secondary outcomes involve the identification of treatment-emergent adverse events TEAEs and objective tumor response.

Rogaratinib is another novel pan-FGFR inhibitor. Rogaratinib showed broad antitumor activity in preclinical studies Grunewald et al. The combination of rogaratinib plus palbociclib and fulvestrant is being assessed in an open-label, multicenter, prospective, phase I dose-escalation clinical trial NCT The primary aims of the study are to assess the recommended phase II dose and the incidence of TEAEs for the combination treatment in patients with metastatic hormone receptor-positive breast cancer who have FGFR-positive tumors.

Additionally, a phase II study is assessing the long-term efficacy and tolerability of rogaratinib in patients who have received the drug in a previous clinical trial and are currently in the continuation phase NCT The selective FGFR-2 inhibitor, RLY, is being evaluated for tolerability and antineoplastic activity in several advanced solid cancers, including the breast NCT The primary outcomes of the study are to determine the maximum tolerated dose of pemigatinib and to assess the pharmacodynamics of the drug.

The I-SPY 2 trial is investigating the effect of trebananib alone or in combination with standard targeted treatments in neoadjuvant settings in patients with breast cancer NCT The main outcome of the trial is to determine the safety and tolerability of NT-I7 in combination with pembrolizumab.

Bintrafusp alfa is a first-in-class bifunctional fusion protein targeting TGF-β and programmed death-ligand 1 PD-L1 Paz-Ares et al. Furthermore, bintrafusp alfa is being assessed as monotherapy in phase II, multicenter, open-label study in participants with TNBC NCT PF, an inhibitor of TGF-βR1, is being evaluated in a phase I dose-escalation study for its safety, tolerability, and pharmacokinetics in patients with advanced solid tumors NCT Table 2 summarizes ongoing clinical trials for selected non-VEGF angiogenic inhibitors in breast cancer.

TABLE 2. Therefore, exploring novel anti-angiogenic therapeutic approaches is of paramount importance for the treatment of aggressive and advanced breast tumors. Such approaches include vascular normalization by targeting pericytes, utilization of miRNAs and extracellular tumor-associated vesicles, using immunotherapeutic drugs, and nanotechnology.

A potential strategy to sensitize tumor endothelium to angiogenesis inhibitors is by targeting pericytes to achieve tumor vascular normalization Lord and Harris, ; Meng et al. Normalization of tumor vasculature prevents cancer cell metastasis, improves the delivery of systemic anticancer therapies, increases the efficacy of local therapies, and enhances recognition by the host immune system.

Pericyte coverage of tumor blood vessels is heterogeneous. In certain tumors, high pericyte coverage of the tumor vasculature causes resistance to anti-angiogenic therapies.

Alternatively, low pericyte coverage detected in the vasculature of certain tumors reduces vascular stability and increases vascular permeability which impairs the delivery of anticancer therapies to tumor cells and allows them to metastasize Meng et al. Earlier studies showed that combining VEGFR and PDGFR inhibitors targeting endothelial cells and pericytes, respectively, improved the efficacy of anti-angiogenic therapy and reduced tumor growth in animal tumor models Bergers et al.

In a xenograft model of breast carcinoma, tumor vascularization was enhanced by increasing the pericyte-endothelium association via a mechanism involving the TGF-β-fibronectin axis Zonneville et al.

In addition, Keskin et al. showed that pericyte targeting in established mouse breast tumors increased Ang-2 expression and that targeting Ang-2 signaling along with pericyte depletion restored vascular stability and decreased tumor growth and metastasis Keskin et al.

Although data from preclinical studies showed that pericyte targeting could be a novel strategy to normalize tumor vasculature, this strategy should be carefully considered as lack of pericyte coverage may disrupt vascular integrity and promote cancer metastasis Lord and Harris, ; Zirlik and Duyster, Assessment of pericyte coverage of tumor vasculature and the identification of the appropriate pericyte-targeted therapy are potential challenges to pericyte targeting Meng et al.

MicroRNAs miRNAs are critical regulators of signaling pathways involved in angiogenesis and cancer metastasis by interacting with the target mRNAs Gallach et al.

To date, there are groups of well-characterized miRNAs implicated in regulating endothelial cell function and angiogenesis, making them attractive targets in tumor angiogenesis Gallach et al. Liang et al. showed that miRNA suppressed breast tumor angiogenesis through targeting HIF-1α and Ang-1 in breast cancer cell lines and animal model.

MiRNA inhibited the proliferation, migration, and tube formation of endothelial cells and decreased the microvessel density Liang et al. Lu et al. reported that miRNAp inhibited tumor invasion and angiogenesis by silencing VEGF-A in breast cancer cells both in vitro and in vivo Lu et al.

MiRNAb inhibited proliferation, migration, and tube formation of endothelial cells. Systemic administration of miRNAb potently suppressed breast tumor growth and vascularization by targeting Akt and downregulating VEGF and c-Myc in breast cancer cells Li et al.

Mimics of miRNA suppressed the proliferation and tube formation of endothelial cells in vitro Wu et al. Moreover, the overexpression of miRNA reduced VEGF and HIF-1α protein levels and suppressed angiogenesis in vivo Wu et al.

Zou et al. showed that miRNA inhibited growth and angiogenesis of TNBC in vivo via post-transcriptional regulation of N-Ras and VEGF Zou et al. Importantly, miRNAs can be transported between cancer cells and stromal cells through extracellular vesicles known to mediate cell-to-cell communication in the tumor microenvironment Kuriyama et al.

Extracellular vesicles are classified into exosomes, microvesicles, and apoptotic bodies based on the size or biogenesis of the vesicles Kuriyama et al. Under hypoxic conditions, tumor cells release extracellular vesicles to a larger extent compared to cells in a normoxic environment Kuriyama et al.

Growing evidence points to the role of tumor-derived extracellular vesicles in tumor angiogenesis of breast cancer. recently reported that extracellular vesicles derived from breast cancer cells are highly enriched with miRNAp which enhanced proliferation and migration of endothelial cells in vitro and angiogenesis and metastasis of breast cancer in vivo Lu et al.

Microvesicles rich in a special VEGF isoform activated VEGFR and induced angiogenesis while being resistant to bevacizumab Feng et al. Exosome-mediated transfer of breast cancer-secreted miRNA efficiently destroyed tight junctions in endothelial monolayers associated with increased vascular permeability Zhou et al.

Few studies showed that extracellular vesicles can be targeted to prevent breast cancer metastasis and restore the activity of anti-angiogenic drugs Zhou et al.

Aslan et al. showed that docosahexaenoic acid decreased the expression of pro-angiogenic genes including HIF-1α, TGF-β, and VEGFR in breast cancer cells and their secreted exosomes Aslan et al. Also, docosahexaenoic acid altered miRNA content in breast cancer cells and their derived exosomes in favor of the inhibition of angiogenesis Aslan et al.

Taken together, miRNAs and extracellular vesicles can be selectively targeted to reduce vascularization in breast cancer providing a novel approach for angiogenesis inhibition Gallach et al.

Normal vasculature is needed for immunosurveillance and efficient detection and killing of cancer cells by immune cells. Disorganized tumor vessels create a selective immune cell barrier limiting the extravasation of immune cells, particularly the cytotoxic T lymphocytes into blood vessels and tumor tissue Yang et al.

Further, hypoxia in the tumor microenvironment promotes lactate accumulation, extracellular acidosis, VEGF overexpression, and VEGFR activation, all of which are known drivers of immune cell tolerance and immunosuppressive status Mendler et al.

Endothelial cells are the first to come into contact with immune cells while infiltrating from the circulation into the tumor tissue Solimando et al. Interestingly, tumor endothelial cells expressed PD-L1 and produced immunosuppressive activity contributing to tumor immune evasion in a mouse model of melanoma Taguchi et al.

Further, leukocyte adhesion was remarkably diminished in tumor vessels Dirkx et al. Tumors secrete angiogenic growth factors that can downregulate endothelial adhesion molecules essential for the interactions with granulocytes, macrophages, and natural killer cells on the vascular endothelium Griffioen, The suppression of these selective adhesion molecules leads to the loss of the adhesive properties of the tumor endothelium thereby impairing immune cell infiltration to tumor tissues.

Solimando et al. showed that junctional adhesion molecule-A JAM-A is an important factor influencing angiogenesis and extra-medullary dissemination in patients with multiple myeloma and its targeting suppressed multiple myeloma-associated angiogenesis both in vitro and in vivo Solimando et al.

Bednarek et al. recently demonstrated that targeting JAM-A with an antagonistic peptide inhibited the adhesion and trans-endothelial migration of breast cancer cells Bednarek et al.

In breast cancer, vascular cell adhesion molecule-1 was aberrantly expressed and mediated angiogenesis and metastasis by binding to its ligand α4β1integrin Sharma et al. Earlier findings also showed that angiogenic stimuli in the microenvironment of breast cancer may influence the expression of endothelial adhesion molecules to prevent leukocyte infiltration to tumor tissue Bouma-Ter Steege et al.

Therefore, selective targeting of adhesion molecules and normalizing tumor vasculature could improve immune cell endothelial adhesion and strengthen the antitumor immune response in epithelial tumors, including breast cancer. A growing body of evidence describes the interplay between immune cells and vasculature in the tumor microenvironment.

The immune response and vascular normalization seem to be mutually regulated Fukumura et al. Normalization of the tumor vasculature improves the infiltration of immune effector cells into tumors enhancing antitumor immune activity Fukumura et al.

Likewise, immunotherapy can promote vascular normalization which further improves the effectiveness of immunotherapeutic drugs and response to anti-angiogenic therapies Huang et al. In preclinical models of breast cancer, immune checkpoint inhibitors induced normalization of tumor vasculature and increased infiltration of immune cells into breast tumors Tian et al.

Together, the combination of anti-angiogenic and immunotherapeutic drugs might be an attractive approach to increase the effectiveness of each class of drugs and reduce the emergence of drug resistance Fukumura et al.

The combination treatment has shown encouraging results in various cancer types Ciciola et al. In a preclinical study, Allen et al. revealed that treatment with a combination of anti-VEGFR-2 and anti-PD-L1 antibodies sensitized tumors to anti-angiogenic therapy and prolonged its efficacy in breast cancer Allen et al.

Li et al. recently demonstrated a dose-dependent synergism for the combined treatment of anti-angiogenic therapy and immune checkpoint blockade Li et al. In this regard, the combination of low-dose anti-VEGFR2 antibody with anti-programmed cell death protein-1 PD-1 therapy normalized tumor vasculature, induced immune cell infiltration, and upregulated PD-1 expression on immune cells in syngeneic breast cancer mouse models.

Additionally, the combined treatment was effective and tolerable in patients with advanced TNBC Li et al. An open-label, randomized, parallel, phase II trial investigated the combination treatment of apatinib, a VEGFR-2 tyrosine kinase inhibitor with the anti-PD-1 monoclonal antibody camrelizumab in patients with advanced TNBC Liu et al.

The results showed that the combination treatment produced favorable therapeutic outcomes in terms of improved objective response rate and PFS which was associated with increased tumor-infiltrating lymphocytes.

The adverse events were manageable and included elevated aminotransferases and hand-foot syndrome Liu et al. Multiple clinical trials of combining anti-angiogenic therapy and immune checkpoint inhibitors are underway Zirlik and Duyster, The nanotechnology-based approach is an emerging strategy for the development of therapies targeting tumor angiogenesis which could improve the current pharmacokinetic profiles of anti-angiogenic drugs and favor their selective accumulation in tumors Banerjee et al.

Radical-containing nanoparticles produced in vitro and in vivo anti-angiogenic activity in a breast cancer model that was mediated by suppressing VEGF in cancer cells Shashni et al. Nanoparticles were also utilized to deliver a combination of therapy for breast cancer to produce anticancer and anti-angiogenic activity Zhao et al.

In a recent study by Gong et al. Table 3 provides a list of novel approaches for targeting vascular growth and angiogenesis in breast cancer. TABLE 3. Breast cancer is a notable example where anti-angiogenic agents had constantly failed to make a significant impact on the survival of patients in clinical settings.

One essential aspect to improve the efficacy of clinically available anti-angiogenic drugs is to better understand the vascular biology of breast cancer at the different stages and molecular types of the disease.

Besides, a greater understanding of the adaptive and intrinsic resistance mechanisms would enhance the proper utilization of angiogenesis inhibitors. Further evaluation for the role of stromal cells within the tumor microenvironment in mediating resistance to anti-angiogenic drugs will improve the efficacy and durability of anti-angiogenic therapy.

Another important facet to consider for the limited activity of angiogenesis inhibitors in breast cancer is the population under examination to allow the identification of breast cancer patients who would benefit most from anti-angiogenic drugs. Furthermore, there are several ongoing efforts to describe novel strategies to inhibit tumor angiogenesis through pericyte targeting, the use of immunotherapy, miRNAs, and the implementation of nanotechnology.

Despite the preclinical success of many of these strategies, limited clinical evidence is available to support their implementation in breast cancer treatment.

NMA conceived the manuscript. All authors listed wrote the manuscript and approved it for publication. The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers.

Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher. Adair Th, M. Google Scholar.

Aird, W. Endothelial Cell Heterogeneity. Cold Spring Harb Perspect. PubMed Abstract CrossRef Full Text Google Scholar. Akino, T. Cytogenetic Abnormalities of Tumor-Associated Endothelial Cells in Human Malignant Tumors.

CrossRef Full Text Google Scholar. Akwii, R. Role of Angiopoietin-2 in Vascular Physiology and Pathophysiology. Cells 8, 1. Al-Khalaf, H. Interleukin-8 Activates Breast Cancer-Associated Adipocytes and Promotes Their Angiogenesis- and Tumorigenesis-Promoting Effects.

Cell Biol 39, 1. Allen, E. Combined Antiangiogenic and Anti-PD-L1 Therapy Stimulates Tumor Immunity through HEV Formation. Transl Med. Andonegui-Elguera, M. An Overview of Vasculogenic Mimicry in Breast Cancer.

Aslan, C. Docosahexaenoic Acid DHA Inhibits Pro-angiogenic Effects of Breast Cancer Cells via Down-Regulating Cellular and Exosomal Expression of Angiogenic Genes and microRNAs. Life Sci. Aster, J. The Varied Roles of Notch in Cancer. Avraham, H. Angiopoietin-2 Mediates Blood-Brain Barrier Impairment and Colonization of Triple-Negative Breast Cancer Cells in Brain.

Balakrishnan, S. Cell Prolif 49, — Banerjee, D. Nanotechnology-mediated Targeting of Tumor Angiogenesis. Cell 3, 3. Banerjee, S. Breast Cancer Cells Secreted Platelet-Derived Growth Factor-Induced Motility of Vascular Smooth Muscle Cells Is Mediated through Neuropilin Carcinog 45, — Barrios, C.

Phase III Randomized Trial of Sunitinib versus Capecitabine in Patients with Previously Treated HER2-Negative Advanced Breast Cancer.

Breast Cancer Res. Barron, G. Circulating Levels of Angiogenesis-Related Growth Factors in Breast Cancer: A Study to Profile Proteins Responsible for Tubule Formation. Bartoschek, M. PDGF Family Function and Prognostic Value in Tumor Biology.

Baselga, J. Sorafenib in Combination with Capecitabine: an Oral Regimen for Patients with HER2-Negative Locally Advanced or Metastatic Breast Cancer. Breast Cancer 17, —e4. Bear, H. Bevacizumab Added to Neoadjuvant Chemotherapy for Breast Cancer.

Bednarek, R. Bell, R. Final Efficacy and Updated Safety Results of the Randomized Phase III BEATRICE Trial Evaluating Adjuvant Bevacizumab-Containing Therapy in Triple-Negative Early Breast Cancer. Bellesoeur, A. Axitinib in the Treatment of Renal Cell Carcinoma: Design, Development, and Place in Therapy.

Drug Des. Devel Ther. Ben Mousa, A. Sorafenib in the Treatment of Advanced Hepatocellular Carcinoma. Saudi J. Benoy, I. Increased Serum Interleukin-8 in Patients with Early and Metastatic Breast Cancer Correlates with Early Dissemination and Survival.

Cancer Res. Bergers, G. Modes of Resistance to Anti-angiogenic Therapy. Cancer 8, — Benefits of Targeting Both Pericytes and Endothelial Cells in the Tumor Vasculature with Kinase Inhibitors.

Bergh, J. First-line Treatment of Advanced Breast Cancer with Sunitinib in Combination with Docetaxel versus Docetaxel Alone: Results of a Prospective, Randomized Phase III Study. Bharti, R. Cancer , — Blumenthal, G. FDA Approval Summary: Sunitinib for the Treatment of Progressive Well-Differentiated Locally Advanced or Metastatic Pancreatic Neuroendocrine Tumors.

Oncologist 17, — Boér, K. Vandetanib with Docetaxel as Second-Line Treatment for Advanced Breast Cancer: a Double-Blind, Placebo-Controlled, Randomized Phase II Study. New Drugs 30, — Bos, R.

Hypoxia-inducible Factor-1alpha Is Associated with Angiogenesis, and Expression of bFGF, PDGF-BB, and EGFR in Invasive Breast Cancer. Histopathology 46, 31— Bottrell, A. An Oncogenic Activity of PDGF-C and its Splice Variant in Human Breast Cancer. Growth Factors 37, — Bouma-Ter Steege, J.

Angiogenic Profile of Breast Carcinoma Determines Leukocyte Infiltration. Brindle, N. Signaling and Functions of Angiopoietin-1 in Vascular protection.

Brossa, A. Sunitinib but Not VEGF Blockade Inhibits Cancer Stem Cell Endothelial Differentiation. Oncotarget 6, — Brown, J. Vasculogenesis: a Crucial Player in the Resistance of Solid Tumours to Radiotherapy. Brufsky, A.

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.

Bussolati, B. Cell Mol Med 13, — Cai, Y. Construction of a Disulfide-Stabilized Diabody against Fibroblast Growth Factor-2 and the Inhibition Activity in Targeting Breast Cancer.

Cancer Sci. Cameron, D. Adjuvant Bevacizumab-Containing Therapy in Triple-Negative Breast Cancer BEATRICE : Primary Results of a Randomised, Phase 3 Trial. Lancet Oncol. Carmeliet, P. Molecular Mechanisms and Clinical Applications of Angiogenesis.

Nature , — Casak, S. FDA Approval Summary: Ramucirumab for Gastric Cancer. Chau, N. Vandetanib for the Treatment of Medullary Thyroid Cancer. Chen, J. Cell Mol Med 22, — Chen, W. Organotropism: New Insights into Molecular Mechanisms of Breast Cancer Metastasis.

NPJ Precis Oncol. Ciciola, P. Combining Immune Checkpoint Inhibitors with Anti-angiogenic Agents. Clavarezza, M. Phase II Open-Label Study of Bevacizumab Combined with Neoadjuvant Anthracycline and Taxane Therapy for Locally Advanced Breast Cancer.

Breast 22, — Clemons, M. Randomised, Phase II, Placebo-Controlled, Trial of Fulvestrant Plus Vandetanib in Postmenopausal Women with Bone Only or Bone Predominant, Hormone-Receptor-Positive Metastatic Breast Cancer MBC : the OCOG ZAMBONEY Study. Cristofanilli, M. Crown, J.

Phase III Trial of Sunitinib in Combination with Capecitabine versus Capecitabine Monotherapy for the Treatment of Patients with Pretreated Metastatic Breast Cancer.

Curigliano, G. Randomized Phase II Study of Sunitinib versus Standard of Care for Patients with Previously Treated Advanced Triple-Negative Breast Cancer.

Dai, X. Breast Cancer Intrinsic Subtype Classification, Clinical Use and Future Trends. Darweesh, R. Gold Nanoparticles and Angiogenesis: Molecular Mechanisms and Biomedical Applications.

Nanomedicine 14, — Decker, T. A Randomized Phase II Study of Paclitaxel Alone versus Paclitaxel Plus Sorafenib in Second- and Third-Line Treatment of Patients with HER2-Negative Metastatic Breast Cancer PASO. BMC Cancer 17, Dey, N.

Evading Anti-angiogenic Therapy: Resistance to Anti-angiogenic Therapy in Solid Tumors. Transl Res. Dirkx, A. Tumor Angiogenesis Modulates Leukocyte-Vessel wall Interactions In Vivo by Reducing Endothelial Adhesion Molecule Expression.

PubMed Abstract Google Scholar. Donnem, T. Vessel Co-option in Primary Human Tumors and Metastases: an Obstacle to Effective Anti-angiogenic Treatment? Cancer Med. Dudley, A.

Tumor Endothelial Cells. Effing, S. Assessing the Risk-Benefit Profile of Ramucirumab in Patients with Advanced Solid Tumors: A Meta-Analysis of Randomized Controlled Trials. EClinicalMedicine 25, El-Kenawi, A. Angiogenesis Inhibitors in Cancer Therapy: Mechanistic Perspective on Classification and Treatment Rationales.

Erber, R. Combined Inhibition of VEGF and PDGF Signaling Enforces Tumor Vessel Regression by Interfering with Pericyte-Mediated Endothelial Cell Survival Mechanisms. FASEB J. Eroğlu, A. Vascular Endothelial Growth Factor VEGF -C, VEGF-D, VEGFR-3 and D Expressions in Primary Breast Cancer: Association with Lymph Node Metastasis.

Evans, C. HIF-mediated Endothelial Response during Cancer Progression. Fakhrejahani, E. Antiangiogenesis Therapy for Breast Cancer: an Update and Perspectives from Clinical Trials. Tumor Angiogenesis: Pericytes and Maturation Are Not to Be Ignored.

Feng, Q. A Class of Extracellular Vesicles from Breast Cancer Cells Activates VEGF Receptors and Tumour Angiogenesis. Flores-Pérez, A. Dual Targeting of ANGPT1 and TGFBR2 Genes by miR Controls Angiogenesis in Breast Cancer.

Folkman, J. Tumor Angiogenesis: Therapeutic Implications. Fukumura, D. Enhancing Cancer Immunotherapy Using Antiangiogenics: Opportunities and Challenges. Gallach, S. MicroRNAs: Promising New Antiangiogenic Targets in Cancer. Gerhardt, H. It is used to treat glioblastoma and cancers of the lung , kidney , breast , colon and rectum.

Other angiogenesis inhibitor drugs work on a different part of the process, by stopping VEGF receptors from sending signals to blood vessel cells. These drugs are known as tyrosine kinase inhibitors TKI. Sunitinib Sutent ® is an example of a tyrosine kinase inhibitor.

For this reason, these drugs are typically used in combination with chemotherapy or other treatments. Angiogenesis inhibitors are particularly effective for treating liver cancer , kidney cancer and neuroendocrine tumors.

Since they act on blood vessel formation and not the tumor itself, the side effects of angiogenesis inhibitors are different than traditional chemotherapy drugs.

My Chart. Donate Today. Request an Appointment Request an Appointment New Patients Current Patients Referring Physicians. Manage Your Risk Manage Your Risk Manage Your Risk Home Tobacco Control Diet Body Weight Physical Activity Skin Safety HPV Hepatitis. Family History Family History Family History Home Genetic Testing Hereditary Cancer Syndromes Genetic Counseling and Testing FAQs.

Donate Donate Donate Home Raise Money Honor Loved Ones Create Your Legacy Endowments Caring Fund Matching Gifts. Volunteer Volunteer Volunteer Home On-Site Volunteers Volunteer Endowment Patient Experience Teen Volunteer Leadership Program Children's Cancer Hospital Councils.

Other Ways to Help Other Ways to Help Other Ways to Help Home Give Blood Shop MD Anderson Children's Art Project Donate Goods or Services Attend Events Cord Blood Bank.

Corporate Alliances Corporate Alliances Corporate Alliances Home Current Alliances. For Physicians. Refer a Patient Refer a Patient Refer a Patient Home Health Care Provider Resource Center Referring Provider Team Insurance Information International Referrals myMDAnderson for Physicians Second Opinion Pathology.

Clinical Trials Clinical Trials Clinical Trials Home. Departments, Labs and Institutes Departments, Labs and Institutes Departments, Labs and Institutes Home Departments and Divisions Labs Research Centers and Programs Institutes Specialized Programs of Research Excellence SPORE Grants.

Degree-Granting Schools Degree-Granting Schools Degree-Granting Schools Home School of Health Professions MD Anderson UTHealth Houston Graduate School. Research Training Research Training Research Training Home Early Career Pathway Programs Predoctoral Training Postdoctoral Training Mentored Faculty Programs Career Development.

Outreach Programs Outreach Programs Outreach Programs Home Project ECHO Observer Programs Comparative Effectiveness Training CERTaIN.

Jump To:. Angiogenesis Inhibitors. Possible side effects include: High blood pressure hypertension Wounds that are slow to heal Increased risk of internal bleeding Perforation of the intestine rare. We're here for you.

Thank you for visiting nature. You are using a browser Anti-angiogenesid with Anti-angiogenesis clinical trials Anti-angiogenesi for CSS. Anti-angiogenesis clinical trials Anti-aging skincare the best Anti-angiogwnesis, we recommend you use a more triials to date browser or turn 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.

Author: Faujas

3 thoughts on “Anti-angiogenesis clinical trials

  1. Ich entschuldige mich, aber meiner Meinung nach irren Sie sich. Es ich kann beweisen. Schreiben Sie mir in PM, wir werden umgehen.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com