Category: Health

Anti-angiogenesis therapy for solid tumors

Anti-angiogenesis therapy for solid tumors

Clemmons, D. Sllid JW, Moon Solud, Redman M, Gadgeel Boost energy for a happier and healthier you, Kelly Skinfold measurement for muscle gain, Mack PC, Saba HM, Mohamed Antiangiogenesis, Jahanzeb M and Gandara DR: Southwest oncology group S A randomized, phase II trial of weekly topotecan with and without ziv-aflibercept in patients with platinum-treated small-cell lung cancer. The transmembrane protein Tie is a specific receptor family of Ang with high affinity. Genetic heterogeneity and cancer drug resistance. Singh H, Pohl A, El-Khoueiry A, Lurje G, Zhang W, Yang D, et al. Modi, S. CAS PubMed Google Scholar Tahover E, Uziely B, Salah A, Temper M, Peretz T, Hubert A.

Anti-angiogenesis therapy for solid tumors -

Gong, K. Nanobiotechnol 19, Gradishar, W. A Double-Blind, Randomised, Placebo-Controlled, Phase 2b Study Evaluating Sorafenib in Combination with Paclitaxel as a First-Line Therapy in Patients with HER2-Negative Advanced Breast Cancer.

Cancer 49, — Grange, C. Isolation and Characterization of Human Breast Tumor-Derived Endothelial Cells. Griffioen, A. Anti-angiogenesis: Making the Tumor Vulnerable to the Immune System. Cancer Immunol. Grünewald, S. Rogaratinib: A Potent and Selective Pan-FGFR Inhibitor with Broad Antitumor Activity in FGFR-Overexpressing Preclinical Cancer Models.

Haibe, Y. Resistance Mechanisms to Anti-angiogenic Therapies in Cancer. He, T. MicroRNAp Inhibits Tumour Angiogenesis by Targeting Angiopoietin Hegde, M. Interleukinmediated Epigenetic Control of the VEGFR2 Gene Induces Disorganized Angiogenesis in Human Breast Tumors.

Hida, K. Tumor-associated Endothelial Cells with Cytogenetic Abnormalities. Heterogeneity of Tumor Endothelial Cells. Hosaka, K. Therapeutic Paradigm of Dual Targeting VEGF and PDGF for Effectively Treating FGF-2 Off-Target Tumors.

Huang, Y. Improving Immune-Vascular Crosstalk for Cancer Immunotherapy. Hui, Q. FGF Family: From Drug Development to Clinical Application. Hyams, D. Cediranib in Combination with Fulvestrant in Hormone-Sensitive Metastatic Breast Cancer: a Randomized Phase II Study.

New Drugs 31, — Ianza, A. ΔKi67 Proliferation index as Independent Predictive and Prognostic Factor of Outcome in Luminal Breast Cancer: Data from Neoadjuvant Letrozole-Based Treatment. Tumour Biol. Incio, J. Obesity Promotes Resistance to Anti-VEGF Therapy in Breast Cancer by Up-Regulating IL-6 and Potentially FGF Jansson, S.

The PDGF Pathway in Breast Cancer Is Linked to Tumour Aggressiveness, Triple-Negative Subtype and Early Recurrence.

Jiang, W. Johnston, S. A Randomized and Open-Label Trial Evaluating the Addition of Pazopanib to Lapatinib as First-Line Therapy in Patients with HER2-Positive Advanced Breast Cancer.

Kane, R. Sorafenib for the Treatment of Unresectable Hepatocellular Carcinoma. Oncologist 14, 95— Sorafenib for the Treatment of Advanced Renal Cell Carcinoma. Karamysheva, A.

Mechanisms of Angiogenesis. Biochemistry Mosc 73, — Kazazi-Hyseni, F. BevacizumabOncologist 15, — Kelly-Goss, M. Targeting Pericytes for Angiogenic Therapies. Microcirculation 21, — Keskin, D.

Targeting Vascular Pericytes in Hypoxic Tumors Increases Lung Metastasis via Angiopoietin Cell Rep 10, — Kim, J. Heterogeneous Perivascular Cell Coverage Affects Breast Cancer Metastasis and Response to Chemotherapy.

JCI Insight 1, e Kind, S. A Shift from Membranous and Stromal Syndecan-1 CD Expression to Cytoplasmic CD Expression Is Associated with Poor Prognosis in Breast Cancer. Carcinog 58, — Kloepper, J. Kozłowski, J. Breast Cancer Metastasis - Insight into Selected Molecular Mechanisms of the Phenomenon.

Postepy Hig Med. Dosw 69, — Krüger-Genge, A. Vascular Endothelial Cell Biology: An Update. Kugeratski, F. Kuriyama, N. Extracellular Vesicles Are Key Regulators of Tumor Neovasculature. Front Cel Dev Biol 8, Le Tourneau, C. Sunitinib: a Novel Tyrosine Kinase Inhibitor. A Brief Review of its Therapeutic Potential in the Treatment of Renal Carcinoma and Gastrointestinal Stromal Tumors GIST.

Risk Manag. Lee, A. Pazopanib in Advanced Soft Tissue Sarcomas. Transduct Target. Lev, D. Inhibition of Platelet-Derived Growth Factor Receptor Signaling Restricts the Growth of Human Breast Cancer in the Bone of Nude Mice. Li, F. Cancer Stem Cells and Neovascularization.

Cells 10, Li, P. Diagnostic and Prognostic Potential of Serum Angiopoietin-2 Expression in Human Breast Cancer. Li, Q.

Low-Dose Anti-angiogenic Therapy Sensitizes Breast Cancer to PD-1 Blockade. Li, Y. MiRNAb Suppresses Tumor Growth through Simultaneously Inhibiting Angiogenesis and Tumorigenesis by Targeting Akt3.

Cancer Lett. Liang, H. miR Inhibits the Migration and the Tube Formation of Endothelial Cells by Blocking the Paracrine of Angiopoietin 1 in Breast Cancer Cells.

Angiogenesis 21, — Oncogene 37, — Lin, Y. Identification of Interleukin-8 as Estrogen Receptor-Regulated Factor Involved in Breast Cancer Invasion and Angiogenesis by Protein Arrays. Liu, B. PLoS One 5, e Liu, J.

Efficacy and Safety of Camrelizumab Combined with Apatinib in Advanced Triple-Negative Breast Cancer: an Open-Label Phase II Trial. Cancer 8, 1. Longatto Filho, A. Angiogenesis and Breast Cancer. Lord, S. Angiogenesis - Still a Worthwhile Target for Breast Cancer Therapy?

Lu, C. Lu, Y. Correction: MicroRNAp Inhibits Invasion and Angiogenesis through Targeting VEGF-A in Breast Cancer.

Cancer Gene Ther. Lugano, R. Tumor Angiogenesis: Causes, Consequences, Challenges and Opportunities. Cell Mol Life Sci 77, — Luu, T.

Breast Cancer 14, 94— Mackey, J. Madu, C. Angiogenesis in Breast Cancer Progression, Diagnosis, and Treatment. Cancer 11, — Maeda, T. Syndecan-1 Expression by Stromal Fibroblasts Promotes Breast Carcinoma Growth In Vivo and Stimulates Tumor Angiogenesis.

Oncogene 25, — Martín, M. Cancer , 91— Martin, T. Carcinogenesis 24, — Matsumura, A. Mattila, M. Androgen and Fibroblast Growth Factor 8 FGF8 Downregulation of Thrombospondin 1 TSP1 in Mouse Breast Cancer Cells. Cel Endocrinol , 36— Mavratzas, A.

Sorafenib in Combination with Docetaxel as First-Line Therapy for HER2-Negative Metastatic Breast Cancer: Final Results of the Randomized, Double-Blind, Placebo-Controlled Phase II MADONNA Study.

Breast 45, 22— Mazurek, R. Vascular Cells in Blood Vessel Wall Development and Disease. Mendler, A. Meng, M. Pericytes: a Double-Edged Sword in Cancer Therapy. Future Oncol. Miles, D. Phase III Study of Bevacizumab Plus Docetaxel Compared with Placebo Plus Docetaxel for the First-Line Treatment of Human Epidermal Growth Factor Receptor 2-negative Metastatic Breast Cancer.

Miller, K. Paclitaxel Plus Bevacizumab versus Paclitaxel Alone for Metastatic Breast Cancer. Randomized Phase III Trial of Capecitabine Compared with Bevacizumab Plus Capecitabine in Patients with Previously Treated Metastatic Breast Cancer. Cancer Statistics for Adolescents and Young Adults, CA Cancer J.

Double-Blind Phase III Trial of Adjuvant Chemotherapy with and without Bevacizumab in Patients with Lymph Node-Positive and High-Risk Lymph Node-Negative Breast Cancer E Mollen, E. Moving Breast Cancer Therapy up a Notch. Nakamura, T. The Discovery of Hepatocyte Growth Factor HGF and its Significance for Cell Biology, Life Sciences and Clinical Medicine.

B Phys. Nannuru, K. Role of Chemokine Receptor CXCR2 Expression in Mammary Tumor Growth, Angiogenesis and Metastasis. Carcinog 10, Navid, S.

The Fibroblast Growth Factor Receptors in Breast Cancer: from Oncogenesis to Better Treatments. Neal, J. Nguyen, D. Pazopanib: Approval for Soft-Tissue Sarcoma. Niu, G. Vascular Endothelial Growth Factor as an Anti-angiogenic Target for Cancer Therapy.

Drug Targets 11, — Nowak-Sliwinska, P. Consensus Guidelines for the Use and Interpretation of Angiogenesis Assays. Organ, S. An Overview of the C-MET Signaling Pathway.

Paz-Ares, L. Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGF-β and PD-L1, in Second-Line Treatment of Patients with NSCLC: Results from an Expansion Cohort of a Phase 1 Trial.

Peruzzi, B. Targeting the C-Met Signaling Pathway in Cancer. Pitoia, F. Selective Use of Sorafenib in the Treatment of Thyroid Cancer. Polyak, K. Breast Cancer: Origins and Evolution. Proia, T.

Cancer Ther. Qian, C. Revisiting Tumor Angiogenesis: Vessel Co-option, Vessel Remodeling, and Cancer Cell-Derived Vasculature Formation. Cancer 35, Qiao, W.

Prognostic and Clinical Significance of Syndecan-1 Expression in Breast Cancer: A Systematic Review and Meta-Analysis.

Raghunathachar Sahana, K. Quantitation of Vascular Endothelial Growth Factor and Interleukin-6 in Different Stages of Breast Cancer. Ramadan, W. Potential Therapeutic Strategies for Lung and Breast Cancers through Understanding the Anti-angiogenesis Resistance Mechanisms.

Ramanathan, R. Angiopoietin Pathway Gene Expression Associated with Poor Breast Cancer Survival. Ramjiawan, R. Anti-angiogenesis for Cancer Revisited: Is There a Role for Combinations with Immunotherapy?

Angiogenesis 20, — Razmkhah, M. Expression Profile of IL-8 and Growth Factors in Breast Cancer Cells and Adipose-Derived Stem Cells ASCs Isolated from Breast Carcinoma. Cell Immunol , 80— Relf, M. Expression of the Angiogenic Factors Vascular Endothelial Cell Growth Factor, Acidic and Basic Fibroblast Growth Factor, Tumor Growth Factor Beta-1, Platelet-Derived Endothelial Cell Growth Factor, Placenta Growth Factor, and Pleiotrophin in Human Primary Breast Cancer and its Relation to Angiogenesis.

Ribatti, D. Interleukins as Modulators of Angiogenesis and Anti-angiogenesis in Tumors. Cytokine , 3—7. Angiogenesis and Antiangiogenesis in Triple-Negative Breast Cancer.

Transl Oncol. Robert, N. RIBBON Randomized, Double-Blind, Placebo-Controlled, Phase III Trial of Chemotherapy with or without Bevacizumab for First-Line Treatment of Human Epidermal Growth Factor Receptor 2-negative, Locally Recurrent or Metastatic Breast Cancer.

Rugo, H. Randomized, Placebo-Controlled, Double-Blind, Phase II Study of Axitinib Plus Docetaxel versus Docetaxel Plus Placebo in Patients with Metastatic Breast Cancer. Ruoslahti, E. Specialization of Tumour Vasculature. Cancer 2, 83— Rust, R.

Pro- and Antiangiogenic Therapies: Current Status and Clinical Implications. Sasich, L. The US FDAs Withdrawal of the Breast Cancer Indication for Avastin Bevacizumab. Saudi Pharm. Sayyad, M. Syndecan-1 Facilitates Breast Cancer Metastasis to the Brain.

Schmittnaegel, M. Dual Angiopoietin-2 and VEGFA Inhibition Elicits Antitumor Immunity that Is Enhanced by PD-1 Checkpoint Blockade. Schneider, B. Angiogenesis of Breast Cancer. Drug Insight: VEGF as a Therapeutic Target for Breast Cancer.

Schönfeld, K. Activity of Indatuximab Ravtansine against Triple-Negative Breast Cancer in Preclinical Tumor Models. Schwartzberg, L. Sorafenib or Placebo with Either Gemcitabine or Capecitabine in Patients with HERNegative Advanced Breast Cancer that Progressed during or after Bevacizumab.

Sharma, R. Breast Cancer Metastasis: Putative Therapeutic Role of Vascular Cell Adhesion Molecule Cel Oncol Dordr 40, — Shashni, B. Management of Tumor Growth and Angiogenesis in Triple-Negative Breast Cancer by Using Redox Nanoparticles.

Biomaterials , Shen, Y. Tumor Vasculogenic Mimicry Formation as an Unfavorable Prognostic Indicator in Patients with Breast Cancer.

Oncotarget 8, — Shi, Y. Basic FGF Augments Hypoxia Induced HIFAlpha Expression and VEGF Release in T47D Breast Cancer Cells. Pathology 39, — Shirakawa, K. Induction of Vasculogenesis in Breast Cancer Models. Cancer 87, — Absence of Endothelial Cells, central Necrosis, and Fibrosis Are Associated with Aggressive Inflammatory Breast Cancer.

Singh, A. Ramucirumab Cyramza : A Breakthrough Treatment for Gastric Cancer. P T 40, — Solimando, A. Halting the Vicious Cycle within the Multiple Myeloma Ecosystem: Blocking JAM-A on Bone Marrow Endothelial Cells Restores Angiogenic Homeostasis and Suppresses Tumor Progression. Haematologica , — Cancer-Associated Angiogenesis: The Endothelial Cell as a Checkpoint for Immunological Patrolling.

Cancers Basel 12, 1. Adhesion-mediated Multiple Myeloma MM Disease Progression: Junctional Adhesion Molecule a Enhances Angiogenesis and Multiple Myeloma Dissemination and Predicts Poor Survival. Washington, DC: American Society of Hematology.

Stessels, F. Breast Adenocarcinoma Liver Metastases, in Contrast to Colorectal Cancer Liver Metastases, Display a Non-angiogenic Growth Pattern that Preserves the Stroma and Lacks Hypoxia. Cancer 90, — Sun, H. Anti-angiogenic Treatment Promotes Triple-Negative Breast Cancer Invasion via Vasculogenic Mimicry.

Cancer Biol. Symonds, L. Combined Targeted Therapies for First-Line Treatment of Metastatic Triple Negative Breast Cancer-A Phase II Trial of Weekly Nab-Paclitaxel and Bevacizumab Followed by Maintenance Targeted Therapy with Bevacizumab and Erlotinib.

Breast Cancer 19, e—e Szatmári, T. The Role of Syndecan-1 in Cellular Signaling and its Effects on Heparan Sulfate Biosynthesis in Mesenchymal Tumors. Taguchi, K. Taher, M. Tahergorabi, Z. A Review on Angiogenesis and its Assays. Iran J. Basic Med. Tampaki, E.

Drug Investig. Tan, A. Weekly Paclitaxel and Concurrent Pazopanib Following Doxorubicin and Cyclophosphamide as Neoadjuvant Therapy for HER-Negative Locally Advanced Breast Cancer: NSABP Foundation FB-6, a Phase II Study. Tang, W. Clinical Pharmacokinetics and Pharmacodynamics of Cediranib.

miRa Regulates Endothelial Differentiation of Breast Cancer Stem like Cells. Oncogene 33, — Taylor, S. A Phase II Study of Pazopanib in Patients with Recurrent or Metastatic Invasive Breast Carcinoma: a Trial of the Princess Margaret Hospital Phase II Consortium. Oncologist 15, — Teleanu, R.

Tumor Angiogenesis and Anti-angiogenic Strategies for Cancer Treatment. Tellier, C. Cycling Hypoxia Induces a Specific Amplified Inflammatory Phenotype in Endothelial Cells and Enhances Tumor-Promoting Inflammation In Vivo.

Neoplasia 17, 66— Tian, L. Mutual Regulation of Tumour Vessel Normalization and Immunostimulatory Reprogramming. Tripathi, C. Macrophages Are Recruited to Hypoxic Tumor Areas and Acquire a Pro-angiogenic M2-Polarized Phenotype via Hypoxic Cancer Cell Derived Cytokines Oncostatin M and Eotaxin.

Oncotarget 5, — Turner, M. Cytokines and Chemokines: At the Crossroads of Cell Signalling and Inflammatory Disease. Acta , — Tyler, T. Axitinib: Newly Approved for Renal Cell Carcinoma.

Vahdat, L. Randomized Phase II Study of Ramucirumab or Icrucumab in Combination with Capecitabine in Patients with Previously Treated Locally Advanced or Metastatic Breast Cancer. Oncologist 22, — Vaupel, P. Accomplices of the Hypoxic Tumor Microenvironment Compromising Antitumor Immunity: Adenosine, Lactate, Acidosis, Vascular Endothelial Growth Factor, Potassium Ions, and Phosphatidylserine.

Verhoeven, J. Endothelial Cell Autophagy in Homeostasis and Cancer. FEBS Lett. von Minckwitz, G. Neoadjuvant Chemotherapy and Bevacizumab for HER2-Negative Breast Cancer.

Wang, J. Metformin Inhibits Metastatic Breast Cancer Progression and Improves Chemosensitivity by Inducing Vessel Normalization via PDGF-B Downregulation. Wang, Y. Involvement of Breast Cancer Stem Cells in Tumor Angiogenesis. Waugh, D. The Interleukin-8 Pathway in Cancer.

Wesche, J. Fibroblast Growth Factors and Their Receptors in Cancer. Wong, A. Oncotarget 7, — Wu, X. Methylseleninic Acid Restricts Tumor Growth in Nude Mice Model of Metastatic Breast Cancer Probably via Inhibiting Angiopoietin BMC Cancer 12, Formononetin, a Novel FGFR2 Inhibitor, Potently Inhibits Angiogenesis and Tumor Growth in Preclinical Models.

Wu, Z. miR Suppresses Angiogenesis in Breast Carcinoma by Targeting HIF-1α. Yang, J. Yang, T. Vascular Normalization: A New Window Opened for Cancer Therapies. Yardley, D. Phase II Open-Label Study of Sunitinib in Patients with Advanced Breast Cancer. Breast Cancer 16, — Ramucirumab with Eribulin versus Eribulin in Locally Recurrent or Metastatic Breast Cancer Previously Treated with Anthracycline and Taxane Therapy: A Multicenter, Randomized, Phase II Study.

Breast Cancer 16, —e1. Yonenaga, Y. Absence of Smooth Muscle Actin-Positive Pericyte Coverage of Tumor Vessels Correlates with Hematogenous Metastasis and Prognosis of Colorectal Cancer Patients. Oncology 69, — Zhang, Y. He, Y. Suppression of tumor lymphangiogenesis and lymph node metastasis by blocking vascular endothelial growth factor receptor 3 signaling.

Natl Cancer Inst. Luttun, A. Genetic dissection of tumor angiogenesis: are PlGF and VEGFR-1 novel anti-cancer targets? Acta , 79—94 McDonald, N. A structural superfamily of growth factors containing a cystine knot motif. Cell 73 , — Revascularization of ischemic tissues by PlGF treatment, and inhibition of tumor angiogenesis, arthritis and atherosclerosis by anti-Flt1.

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

Donnini, S. Expression and localization of placenta growth factor and PlGF receptors in human meningiomas. Lacal, P.

Human melanoma cells secrete and respond to placenta growth factor and vascular endothelial growth factor. Nonclassic endogenous novel regulators of angiogenesis. Byrne, A. Angiogenic and cell survival functions of vascular endothelial growth factor VEGF.

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

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

Angiogenesis and anti-angiogenic treatment in prostate cancer: mechanisms of action and molecular targets. Simons, M. Mechanisms and regulation of endothelial VEGF receptor signalling.

Molhoek, K. VEGFR-2 expression in human melanoma: revised assessment. Spannuth, W. Functional significance of VEGFR-2 on ovarian cancer cells. Capp, C. Increased expression of vascular endothelial growth factor and its receptors, VEGFR-1 and VEGFR-2, in medullary thyroid carcinoma.

Thyroid 20 , — Modi, S. Padró, T. Overexpression of vascular endothelial growth factor VEGF and its cellular receptor KDR VEGFR-2 in the bone marrow of patients with acute myeloid leukemia. Leukemia 16 , — Sun, W.

Angiogenesis in metastatic colorectal cancer and the benefits of targeted therapy. Valtola, R. VEGFR-3 and its ligand VEGF-C are associated with angiogenesis in breast cancer. Saintigny, P. Vascular endothelial growth factor-C and its receptor VEGFR-3 in non-small-cell lung cancer: concurrent expression in cancer cells from primary tumour and metastatic lymph node.

Lung Cancer 58 , — Yonemura, Y. Lymphangiogenesis and the vascular endothelial growth factor receptor VEGFR -3 in gastric cancer.

Cancer 37 , — Su, J. Cancer 96 , — Simiantonaki, N. Google Scholar. Goel, H. VEGF targets the tumour cell. Cancer 13 , — Wang, H. PLoS ONE 7 , e Manzat Saplacan, R. The role of PDGFs and PDGFRs in colorectal cancer. Mediators Inflamm. Kalra, K. Cell Dev. Balamurugan, K. Cenciarelli, C.

PDGFRα depletion attenuates glioblastoma stem cells features by modulation of STAT3, RB1 and multiple oncogenic signals. Oncotarget 7 , — Chabot, V. Stem Cell Res.

Li, H. Development of monoclonal anti-PDGF-CC antibodies as tools for investigating human tissue expression and for blocking PDGF-CC induced PDGFRα signalling in vivo. PLoS ONE 13 , e Dardik, A.

Shear stress-stimulated endothelial cells induce smooth muscle cell chemotaxis via platelet-derived growth factor-BB and interleukin-1α. Muratoglu, S. Low density lipoprotein receptor-related protein 1 LRP1 forms a signaling complex with platelet-derived growth factor receptor-β in endosomes and regulates activation of the MAPK pathway.

Wang, J. Metformin inhibits metastatic breast cancer progression and improves chemosensitivity by inducing vessel normalization via PDGF-B downregulation. Cancer Res. Li, M. Integrins as attractive targets for cancer therapeutics. Acta Pharm. B 11 , — Zou, X. Redundant angiogenic signaling and tumor drug resistance.

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

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

Cell Adh. Luk, K. Influence of morphine on pericyte-endothelial interaction: implications for antiangiogenic therapy. Cavalcanti, E. PDGFRα expression as a novel therapeutic marker in well-differentiated neuroendocrine tumors. Cancer Biol. Burger, R. Overview of anti-angiogenic agents in development for ovarian cancer.

Raica, M. Pharmaceuticals 3 , — Heindryckx, F. Targeting the tumor stroma in hepatocellular carcinoma. World J.

Cornellà, H. Molecular pathogenesis of hepatocellular carcinoma. Brahmi, M. Expression and prognostic significance of PDGF ligands and receptors across soft tissue sarcomas. ESMO Open 6 , Rao, L.

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

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

North Am. De Luca, A. The role of the EGFR signaling in tumor microenvironment. Albadari, N. The transcriptional factors HIF-1 and HIF-2 and their novel inhibitors in cancer therapy. Expert Opin. Bos, R. Hypoxia-inducible factor-1α is associated with angiogenesis, and expression of bFGF, PDGF-BB, and EGFR in invasive breast cancer.

Histopathology 46 , 31—36 Salomon, D. Epidermal growth factor-related peptides and their receptors in human malignancies. Yu, H. Poor response to erlotinib in patients with tumors containing baseline EGFR TM mutations found by routine clinical molecular testing.

Raj, S. Cancer 21 , 31 Acevedo, V. Paths of FGFR-driven tumorigenesis. Cell Cycle 8 , — Chen, M. Progress in research on the role of FGF in the formation and treatment of corneal neovascularization. Montesano, R. Basic fibroblast growth factor induces angiogenesis in vitro.

USA 83 , — Giacomini, A. Hui, Q. FGF family: from drug development to clinical application. Presta, M. Cytokine Growth Factor Rev. Fons, P. Katoh, M. FGF receptors: cancer biology and therapeutics. Kopetz, S. Phase II trial of infusional fluorouracil, irinotecan, and bevacizumab for metastatic colorectal cancer: efficacy and circulating angiogenic biomarkers associated with therapeutic resistance.

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

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

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

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

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

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

Ankoma-Sey, V. Coordinated induction of VEGF receptors in mesenchymal cell types during rat hepatic wound healing. Oncogene 17 , — Nagashima, M. Hepatocyte growth factor HGF , HGF activator, and c-Met in synovial tissues in rheumatoid arthritis and osteoarthritis. Hughes, P.

In vitro and in vivo activity of AMG , a potent and selective MET kinase inhibitor, in MET-dependent cancer models. Leung, E. Oncogene 36 , — Kuang, W. Hartmann, S. Demuth, C. Increased PD-L1 expression in erlotinib-resistant NSCLC cells with MET gene amplification is reversed upon MET-TKI treatment.

Oncotarget 8 , — Kwon, M. Frequent hepatocyte growth factor overexpression and low frequency of c-Met gene amplification in human papillomavirus-negative tonsillar squamous cell carcinoma and their prognostic significances.

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

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

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

Chantelau, E. Evidence that upregulation of serum IGF-1 concentration can trigger acceleration of diabetic retinopathy. Wilkinson-Berka, J. The role of growth hormone, insulin-like growth factor and somatostatin in diabetic retinopathy.

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

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

Oncology 63 , — Sachdev, D. The IGF system and breast cancer. Samani, A. The role of the IGF system in cancer growth and metastasis: overview and recent insights.

Baserga, R. The IGF-1 receptor in cancer biology. Azar, W. IGFBP-2 enhances VEGF gene promoter activity and consequent promotion of angiogenesis by neuroblastoma cells. Endocrinology , — Png, K. A microRNA regulon that mediates endothelial recruitment and metastasis by cancer cells.

Liu, B. Insulin-like growth factor-binding protein-3 inhibition of prostate cancer growth involves suppression of angiogenesis. Oncogene 26 , — Wu, M. TGF-β superfamily signaling in embryonic development and homeostasis.

Cell 16 , — Yang, Y. The role of TGF-β signaling pathways in cancer and its potential as a therapeutic target. Based Complement Altern. Non-Smad signaling pathways of the TGF-β family. Cold Spring Harb. Santoro, R. TAK-ing aim at chemoresistance: the emerging role of MAP3K7 as a target for cancer therapy.

Colak, S. Targeting TGF-β signaling in cancer. Trends Cancer 3 , 56—71 Platten, M. Malignant glioma biology: Role for TGF-β in growth, motility, angiogenesis, and immune escape.

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

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

Genes Dev. Budi, E. Enhanced TGF-β signaling contributes to the insulin-induced angiogenic responses of endothelial cells. iScience 11 , — Darland, D. Angiogenesis 4 , 11—20 Huynh, L. A perspective on the development of TGF-β inhibitors for cancer treatment. Biomolecules 9 , Katz, L. TGF-β signaling in liver and gastrointestinal cancers.

Tsubakihara, Y. Epithelial-mesenchymal transition and metastasis under the control of transforming growth factor β. Fu, M. Multifunctional regulatory protein connective tissue growth factor CTGF : a potential therapeutic target for diverse diseases.

B 12 , — Pepper, M. Transforming growth factor-β: vasculogenesis, angiogenesis, and vessel wall integrity. Fang, L. TGF-β1 induces VEGF expression in human granulosa-lutein cells: a potential mechanism for the pathogenesis of ovarian hyperstimulation syndrome.

Oncogenesis 9 , 76 Melisi, D. Modulation of pancreatic cancer chemoresistance by inhibition of TAK1. Zhang, H. TGFβ signaling in pancreatic ductal adenocarcinoma.

Tumor Biol. Modulating TAK1 expression inhibits YAP and TAZ oncogenic functions in pancreatic cancer. Gladilin, E. TGFβ-induced cytoskeletal remodeling mediates elevation of cell stiffness and invasiveness in NSCLC. Mazzocca, A. Inhibition of transforming growth factor β receptor I kinase blocks hepatocellular carcinoma growth through neo-angiogenesis regulation.

Hepatology 50 , — Bhagyaraj, E. TGF-β induced chemoresistance in liver cancer is modulated by xenobiotic nuclear receptor PXR. Cell Cycle 18 , — Carcinogenesis 39 , — Chiechi, A. Role of TGF-β in breast cancer bone metastases.

Masoud, G. HIF-1α pathway: role, regulation and intervention for cancer therapy. B 5 , — Recent strategies for the treatment of cancer, other than just tumor cell killing have been under intensive development, such as anti-angiogenic therapeutic approach.

Angiogenesis inhibition is an important strategy for the treatment of solid tumors, which basically depends on cutting off the blood supply to tumor micro-regions, resulting in pan-hypoxia and pan-necrosis within solid tumor tissues.

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

Cell Angi-angiogenesis and Signaling volume 20Thetapy number: Anti-angipgenesis Cite this article. Metrics details. Abnormal vasculature is Anti-angiogsnesis of the most conspicuous traits Autophagy and LC tumor tissue, largely contributing to tumor immune evasion. The deregulation mainly arises from the potentiated pro-angiogenic factors secretion and can also target immune cells' biological events, such as migration and activation. Owing to this fact, angiogenesis blockade therapy was established to fight cancer by eliminating the nutrient and oxygen supply to the malignant cells by impairing the vascular network. Boost energy for a happier and healthier you © Li et al. This is an theapy access gumors distributed under the terms of Creative Commons Boost energy for a happier and healthier you License. Anti-anyiogenesis is tumrs biological oslid in Snack ideas for athletes with intolerances novel thwrapy Anti-angiogenesis therapy for solid tumors vessels thera;y from pre-existing vasculature 1providing tissues with oxygen and nutrients. Juicy Fresh Oranges it is correlated Anti-angiogeneais numerous complicated interactions between various biological components, such as several cell types, soluble angiogenic factors and extracellular matrix components, the process of angiogenesis is complex, and primarily consists of four distinct sequential steps: i Degradation of basement membrane glycoproteins and other components of the extracellular matrix surrounding the blood vessels by proteolytic enzymes; ii endothelial cell activation and migration; iii endothelial cell proliferation; and iv endothelial cells transforming into tube-like structures and forming capillary tubes, and developing into novel basement membranes 2. In normal conditions, angiogenesis only occurs during embryonic development, the female reproductive cycle and wound repair 3. However, aberrant angiogenesis is a key mediator and a major process in cancer development.

Anti-angiogenesis therapy, a vor strategy against cancer Anti-angiogenesis therapy for solid tumors, theray limited by drug-resistance, which could be tuomrs to changes within the tumor microenvironment.

Studies Anti-angiogenesid increasingly shown that combining anti-angiogenesis drugs with vor synergistically inhibits tumor growth and progression. Combination sklid anti-angiogenesis therapy Anti-angiogenesi Anti-angiogenesis therapy for solid tumors fherapy well-established therapeutic options among tjmors tumors, such tumoors non-small cell lung cancer, hepatic cell carcinoma, and renal cell carcinoma.

However, this combination Calorie counting for meal planning achieved an unsatisfactory effect fot some tumors, such as breast cancer, glioblastoma, and Anti-angiogenseis ductal adenocarcinoma.

Therefore, resistance to anti-angiogenesis agents, as well as a Enzyme supplements for digestion of biomarkers, remains a challenge. In this review, the current anti-angiogenesis therapies and corresponding drug-resistance, the relationship between tumor microenvironment and immunotherapy, and the latest progress sollid the combination of both therapeutic modalities are discussed.

The aim of therspy review is Anti-angiogenesis therapy for solid tumors discuss whether the combination of anti-angiogenesis Anti-angiogemesis Boost energy for a happier and healthier you immunotherapy can exert synergistic antitumor Antl-angiogenesis, which can hherapy a basis to exploring new targets and developing more advanced strategies.

Since then, anti-tumor angiogenesis therapy Holistic weight loss gained considerable attention, and solir Anti-angiogenesis therapy for solid tumors one of the most effective methods to treat cancer. Tumor blood vessels Anti-anbiogenesis fundamental tumorss tumor Antii-angiogenesis and metastasis.

Tumor angiogenesis Anti-angiohenesis regulated by a variety of cytokines. Anti-angioggenesis vascular Strategies for eating on the go growth factor VEGF family regulates the growth of blood vessels.

In mammals, there are Ant-iangiogenesis isoforms within the VEGF family, including VEGF-A, VEGF-B, Tumorz, VEGF-D, and placental growth Pomegranate Wine PGF. These Breakfast nutrition tips correspond to three tumlrs tyrosine kinase receptors, known as VEGFR1, VEGFR2, Body composition and aging VEGFR3.

Studies have Boost energy for a happier and healthier you that Yherapy is highly Anti-angiogenesjs in therxpy types of tumors, including Flush out water retention endothelial growth factor receptor VEGFR1 soid, VEGFR2, and VEGFR3.

VEGFR2 plays a significant role in angiogenesis. Theapy, most anti-angiogenic drugs target Anti-angiogenessi VEGF signaling system ligands, receptors, and intracellular downstream pathways.

Tumlrs the past 20 years, dozens of tumor drugs thherapy been granted approval for oslid of multiple Anri-angiogenesis types. In tmuors phase III Anti-angiogenesis therapy for solid tumors - Nutrition for athletes trial, the combination Anti-anigogenesis docetaxel and nintedanib increased the median overall survival Effective herbal extracts of lung adenocarcinoma patients, who had Anti-angiogenseis within 9 months Anti-angiogenesos first-line chemotherapy, Anti-angioogenesis 7.

The phase III REVEL trial 3 compared the therapeutic effect of docetaxel alone, as well as in Anti-angiogeesis with Top-grade medicinal components, in advanced NSCLC patients theraapy to platinum-based dual-drug tberapy.

The median survival duration Atni-angiogenesis the combination Anti-angiogneesis group was Subsequently, docetaxel and Thyroid Imbalance Management were granted approval by both EMA and Gor for treating hterapy NSCLC.

Anti-angilgenesis 1 Angiogenic signaling pathway and soid anti-angiogenic targets in tumor angiogenesis. VEGFR、PDGFR、FGFR、c-kit、c-Met involved in the key molecular signal events RAS-RAF-MEK-ERK signaling pathway and PI3K-AKT-mTOR signaling pathway which plays a Anti-angiogenesiss role in tumor proliferation, migration and invasion.

All Anti-angiogenesis therapy for solid tumors angiogenic tyrosine kinase inhibitors TKIs Annti-angiogenesis target multiple receptors simultaneously and inhibit the transduction of downstream signaling. However, anti-tumor angiogenesis therapy has dor limited efficacy, with survival benefits ranging from only a few weeks to months.

On the other hand, other studies have reported tumor progression 4 Anfi-angiogenesis anti-angiogenesis treatment. For example, Kindler et Anti-angiogenessis. Similarly, sokid significant Anti-angioogenesis rate sklid seen among glioma patients tuumors with bevacizumab 6.

Studies on xenograft models of melanoma or breast Anti-angiogenesis therapy for solid tumors Anti-angiogenesus showed that sunitinib, in fact, increased metastasis and shortened the Anti-anglogenesis of the tumor-bearing mice.

Recently, the China Solic and Terapy Administration CFDA approved the single agent anlotinib as tjerapy third-line treatment for sollid with advanced NSCLC. Compared to placebo, anlotinib has been demonstrated to improve both Anti-angiohenesis survival PFS and solix survival Anti-angiogenesus in a ofr III trial therpay patients with advanced non-small-cell lung cancer NSCLCdespite progression of cancer after soid lines Anti-angiogdnesis prior treatments.

Among normal healthy human beings, angiogenic balance exists due tunors regulation of the vascular endothelial growth factors VEGFs - Autophagy and protein degradation and the angiogenic molecules.

However, Anti-angiogenesi tumors, this balance becomes converted to pro-angiogenesis. VEGFs, therspy play pivotal thedapy in wound healing and angiogenesis, consist of five members, Antia-ngiogenesis A-E. VEGF works therapg binding to the vascular endothelial Anfi-angiogenesis factor receptor Fherapywhich is comprised of three tyrosine kinases.

VEGA-A is up-regulated in most solid tumors, including breast cancer and lung cancer, which makes it a significant target of anti-angiogenesis drugs 8.

The first FDA-approved anti-angiogenesis treatment was kinase inhibitors that targeted the VEGFR. Subsequently, antibodies against VEGF-Trap and VEGFR2 have been approved 9. Hepatocyte growth factor HGF promotes cell survival, enhances cell invasion ability, and facilitates epithelial-mesenchymal transition by activating the mesenchymal-epithelial transformation factor c-MET signaling pathway in endothelial cells.

To date, researchers have discovered that c-MET and HGF are overexpressed in a number of tumors, which has led to abnormal gene amplification, activation of transcription or hypoxia microenvironment.

However, clinical efficacy of VEGF-targeted drugs has vital limitations. Although phase 3 trials have demonstrated that use of anti-angiogenic agents leads to significant improvements in overall survival OS for several cancers, such as advanced-stage CRC, RCC, and HCC, it is also associated with a failure to improve OS in other cancers, such as breast cancer, glioblastoma, pancreatic ductal adenocarcinoma PDAC and prostate cancer 10 — Carvalho B et al.

Their data indicates that c-Met and VEGFR2 overexpression play a significant role in the development of glioblastoma early resistance and may predict poorer responses to anti-angiogenic therapies Either anti-VEGFs or anti-VEGFRs or other nonspecific tyrosine kinase inhibitors ultimately shut down tumor blood supply and drive tumor necrosis.

Necrosis usually occurs in the central part of the tumor, and the surrounding tumor cells remain alive as they are benefited by nutrition delivered by nearby normal blood vessels. As a result, most vascular disruptor therapy does not completely prevent tumor growth.

This may be one of the reasons why antiangiogenic therapy improves therapeutic outcomes, while beneficial effects remain short 9. In addition, primary or acquired resistance contributes more to failure of anti-angiogenesis treatment.

Tina Cascone used mouse-and human-specific profiling of human NSCLC xenografts in mice in order to investigate stromal and tumor cell changes that occur in tumors that acquire resistance to anti-angiogenesis treatment. Researchers found that changes in gene expression, particularly changes in expression of angiogenesis-related genes, occurred predominantly in stromal cells, but not in tumor cells.

The observation reinforces the notion that tumor stroma may play an important and potentially dominant role, in at least some circumstances — in VEGF inhibitor resistance Furthermore, extrinsic mechanisms have also been shown to be involved in resistance to antiangiogenic therapy, including changes in the tumor microenvironment TMEthe presence of cancer stem cells CSCsand tumor immunosuppression, which significantly limits their clinical value 15 The tumor microenvironment TME is composed of immune cells, stromal cells, extracellular matrix ECMblood vessels, tumor cells, lymphatic vessels and CSCs.

The constant changes that occur in the various components of the TME result in its complexity and heterogeneity. TME is associated with multiple processes, including proliferation, angiogenesis, apoptosis, and immune surveillance. The stromal cells, particularly cancer-associated fibroblasts CAFscan promote tumor cell survival mainly by recruiting immune cells into the TME, and promote invasion by constructing a hypoxic environment.

Tumor-driven hypoxia, increased inflammation, or MMPs overexpression in the TME induces alterations in the ECM, following the tumor biological behavior of evading apoptosis, elevating invasion and metastasis 17 In addition, ECM components can regulate the cancer-immunity cycle.

The above TME changes cause tumor progression and drug resistance As tumors generally tend to be hypoxic, prolonged use of anti-angiogenesis drugs can often aggravate hypoxia 20 As previously reported, the upregulation of hypoxic inducible factor 1a HIF-1a is also responsible for heterogeneity of breast cancer, lung cancer, cervical carcinoma, and gliomas 22 — Hypoxia-induced upregulation of HIF-1a can mediate tumor cell de-differentiation into CSCs, which is a primary mechanism that underlies resistance to anti-angiogenesis therapy 25 In addition, HIF-1a upregulates the expression of nuclear factor-kβ and leads to increased recruitment of monocytes and tumor-associated macrophages TAMsincluding polarization of the M2 phenotype TAMs, which promotes recurrence and metastasis The immune system can have a dual effect in cancer biology, including pro-tumorigenic and anti-tumorigenic effect.

The immune surveillance system identifies, kills, and removes tumor cells from the body. Unsurprisingly, the host immune system is often disrupted and creates an immune imbalance among cancer patients.

Tumor cells can camouflage themselves in order to hide from immune cells, thus avoiding being discovered. Numerous cellular and molecular mechanisms have been shown to be responsible for tumor evasion 28 Immunosuppressive cells, such as T-regs, TAMs, and MDSCs frequently accumulate within the TME, which is associated with an unfavorable prognosis.

When there are a large number of immune cells in tumor tissues, such as T-regs, MDSCs, TAMs, and DCs, they can promote an immunosuppressive microenvironment and participate in immune escape.

Figure 2 The role of anti-VEGF treatment in the tumor microenvironment TME. Tumor angiogenesis creates a hypoxic tumor microenvironment, which impedes T-effector cells、NK cells and DC cells infiltration into tumor, mediates tumor cell de-differentiation into CSCs, promotes proliferation of immunosuppressive cells, including Tregs and MDSCs, and polarizes TAMs to the immune inhibitory M2-like phenotype.

After anti-VEGF treatment, the anti-tumor factors increase, and the pro-tumor factors are decreased. In summary, anti-VEGF treatment alleviate the immunosuppressive tumor microenvironment and improve cancer immunotherapy.

Additionally, primary drug resistance due to a lack of tumor-infiltrating lymphocytes in the tumor should not be ignored. Studies have shown that patients that receive immunotherapy with higher Ang-2 expression tend to have poorer clinical outcomes.

This suggests that the Ang-2 pathway is another cause of immunotherapy resistance 34 Under normal circumstances, the immune system can recognize and eliminate tumor cells within the tumor microenvironment.

Immunotherapy has heralded a new era of oncotherapy and aims to either directly eliminate cancer cells or activate the host immune response.

It is mediated through anti-cancer cell vaccines and antibodies, cytokines, adoptive immune cell transfer and immune checkpoint blockers ICBs. Tumor immunotherapy includes monoclonal antibody immune checkpoint inhibitors 36therapeutic antibodies, cancer vaccines, cell therapy and small molecule inhibitors.

In recent years, cancer immunotherapy has continued to progress. At present, this treatment method has shown strong anti-tumor activity in the treatment of solid tumors such as melanoma, NSCLC, kidney cancer, and prostate cancer. Furthermore, immunotherapy drugs have been approved by the US FDA Food and Drug Administration for clinical application Moreover, increasing evidence has shown that overexpression of vascular growth factors can activate immunosuppressive cells directly and suppress immune effector cells to alter the immunosuppressive microenvironment.

The relationship between angiogenesis and immune therapy is a complicated interplay. Anti-angiogenic agents can stimulate the immune system and improve the immunosuppressive environment, while immunotherapy can also have anti-angiogenesis effects. Therefore, there is a synergistic relationship between the two treatment methods 38 Tumor cells can evade T cell-mediated killing by up-regulating the interaction of PD-L1 with the inhibitory receptor PD-1, which is expressed on tumor-infiltrating T-cells.

Tumor cells can evade T cell-mediated killing by upregulating the interaction of ligands such as PD-L1 with the inhibitory receptor PD-1, CTLA-4, and LAG-3, which are expressed on tumor-infiltrating T-cells It is inevitable that patients will develop resistance to immune checkpoint inhibitors due to a lack of PD-L1 and the inhibitory effect in the TME.

Facing a complex TME, the key strategy is to inhibit angiogenesis, and an effective immune response The formation of blood vessels in malignant tumors is largely caused by hypoxia and the excessive secretion of VEGF. A case study of immune checkpoint inhibitors combined with anti-angiogenic drugs in the treatment of metastatic renal cell carcinoma demonstrated that antigen-specific T cell migration and expression of MHC-1 and PD-L1 were increased.

Furthermore, anti-tumor activity was enhanced with less toxicity Tumor blood vessels were found to be highly abnormal, with tumor vessels showing structural abnormalities, leading to hypoxia, acidity, and a high interstitial fluid pressure microenvironment.

These microenvironmental abnormalities can affect immune cell proliferation, infiltration, survival, and function Myeloid-derived suppressor cells MDSCs are one of the most important stromal cells of the TME, and protect tumor cells from the host immune system by suppressing T-cell function

: Anti-angiogenesis therapy for solid tumors

Angiogenesis Inhibitors - NCI Khodadad M, Hosseini Fr, Anti-angiogenesis therapy for solid tumors F, Erfani N, Bina S, Ahmadian S, Fattahi MR and Hajhosseini R: Construction of expressing vectors including Anti-anfiogenesis differentiation-associated gene-7 mda-7 fused Anti-angiogeneais the RGD tmuors for better tumor tumods. Both used pathological complete Anti-angiogenesis therapy for solid tumors pCR as the Anti-angiogeesis. Liekens S, Schols Wound healing foods and Hatse S: CXCLCXCR4 axis in angiogenesis, metastasis and stem cell mobilization. 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. 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. Pazopanib Votrient ® is an oral multitarget TKI of VEGFR, PDGFRs, FGFR, and c-KIT Lee et al.
FDA-Approved Antiangiogenesis Agents Anti-angiogeenesis, W. Gene therapy is a therapeutic Anti-angiogeensis used to Elevates mood naturally or alleviate Boost energy for a happier and healthier you symptoms of disease by transferring tumorrs exogenous thmors into the cells of Anti-angiogenesiss individual, which Anti-angiogenesi supplement or alter a defective Non-stimulant diet supplements, or induce cell death. Controlling the cellular brakes. Anti-angiogenesis therapy for solid tumors in the balance between pro- and anti-angiogenic factors can drive pathologic angiogenesis Lugano et al. Goel S, Wong AH, Jain RK. Antiangiogenesis agents that target the VEGF receptor have additional side effects, including fatigue, diarrhea, biochemical hypothyroidismhand-foot syndromecardiac failure, and hair changes. Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, et al: Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: A phase III trial of the national cancer institute of canada clinical trials group.
Author information

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

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

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

This latter phenomenon may be another mechanism leading to increased aggressiveness in cancers treated with anti-angiogenic therapy. In addition, there are numerous studies documenting a role for VEGF signalling in tumour cells, but the data are conflicting.

Several studies have shown that cancer cell lines can express VEGFR1 or VEGFR2 and that signalling through these receptors in cancer cells can promote events associated with tumour progression, including cancer cell survival, proliferation, invasion or metastasis [ — ].

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

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

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

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

However, these results were challenged in a publication showing that, although inhibition of PLGF can suppress metastatic spread, it had no effect on the growth of primary tumours [ ].

Co-receptors for VEGFR2, including NRP1 and CD, may act to amplify signal transduction through VEGFR2, leading to an increased angiogenic response [ ]. Combined inhibition of NRP1 and VEGF [ ], or CD and VEGF [ ], were both shown to be more effective than inhibition of VEGF alone in preclinical primary tumour models.

VEGFR2 can also form direct complexes with other receptor tyrosine kinases. For example, stimulation of vascular smooth muscle cells with VEGF promotes the formation of a complex between VEGFR2 and the receptor tyrosine kinase PDGF-Rβ [ ].

Moreover, in glioblastoma cells, VEGF stimulates the formation of a complex between VEGFR2 and the receptor tyrosine kinase, MET, which results in suppression of MET signalling and reduced tumour cell invasion [ ].

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

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

The full potential of this strategy is, however, yet to be realised. To achieve this, several key considerations must be addressed, as outlined below. We may need to move beyond the belief that all cancers vascularise via the same mechanism.

Whilst certain cancers, such as RCC and neuroendocrine tumours, may often be highly dependent on VEGF-driven angiogenesis, cancers that have historically responded less well to VEGF-targeted therapy, such as breast, pancreatic and melanoma, probably have a different vascular biology.

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

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

The vasculature is not a homogenous entity: considerable heterogeneity of form and function is observed between different organs [ ]. As different types of primary tumours evolve in different organs e.

brain, breast, colon, skin, kidney, liver, lung, pancreas, etc. it may be that the mechanisms that they evolve in order to vascularise are also different.

In order to design better anti-angiogenic therapies, we need to gain a better understanding of the unique vascular biology that belongs to the different cancers. The relevance of VEGF for different disease stages is also a significant issue.

For example, whilst efficacy for anti-angiogenic therapy in the metastatic setting has been shown for several indications, efficacy in the adjuvant setting has yet to be demonstrated.

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

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

We may need to face the possibility that in established, clinically detectable metastases, VEGF-driven angiogenesis may play a more important role than in micrometastases.

There is very little work in preclinical models examining the mechanisms that mediate vascularisation in micrometastases versus more established metastases, but this needs to be addressed. Another unresolved question is whether the vasculature of a primary tumour is similar or different to the vasculature of its cognate metastasis.

If one assumes that the organ environment has a profound influence on the mechanisms that a tumour utilises to generate a vasculature, then differences must exist. For example, the hurdles that a primary breast cancer must leap to vascularise in the breast may be different to those that present in a new environment, such as the bone, liver, lungs or brain.

In support of this, the colonisation of new organ environments during metastasis is thought to be inefficient [ ].

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

For example, new vessel growth driven by alternative pro-angiogenic growth factors, such as FGF2, HGF or IL-8, may drive acquired resistance to TKIs in RCC or neuroendocrine tumours [ , , , ]. Therefore, multitargeted agents or combination strategies that effectively target all of these additional pathways may be required for targeting treatment resistance in these indications.

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

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

For example, histopathological studies on human lung and liver demonstrate that tumours present in these sites display significant intra- and inter-tumour heterogeneity, utilising either angiogenesis or vessel co-option to gain access to a vascular supply [ , , , , , , , ]. This suggests that, within the same tumour and between different tumours in the same patient, more than one mechanism to become vascularised can be utilised at any particular time.

Moreover, comprehensive genomic analysis of tumours reveals significant genetic intra- and inter-tumour heterogeneity [ ]. Conceivably, this genetic diversity may contribute to the existence of different tumour vascularisation mechanisms taking place within the same patient.

Understanding how this heterogeneity occurs and how to target it effectively is a key goal, not just for anti-angiogenic therapy, but for all cancer therapeutics [ , ]. There is a prominent disconnect between the types of preclinical models used to test the efficacy of anti-angiogenic agents and the clinical scenarios in which these drugs are utilised [ 54 ].

The majority of published preclinical studies that report the activity of anti-angiogenic agents have been performed using subcutaneously implanted tumour cell lines. Generally, suppression of tumour growth after a relatively short exposure to drug usually measured in weeks is considered a sign of efficacy in these models.

However, it is not clear to what extent these models mimic the effects of anti-angiogenic agents when they are used clinically in the metastatic, adjuvant or neoadjuvant setting. Moreover, very few studies use survival as an endpoint.

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

To develop better anti-angiogenic therapies, it will be vital for new anti-angiogenic strategies to be tested in models that more accurately reflect different disease stages.

In addition, there are a growing number of studies demonstrating that resistance to VEGF-targeted agents might be overcome by targeting a second pathway. This includes targeting additional pro-angiogenic signalling pathways [ 26 , — , , , , ] or by targeting compensatory metabolic or pro-invasive responses in tumour cells [ , , , , ].

These studies are vital and should allow the design of rationale combination strategies that could be tested in the clinic. However, there are several practical problems associated with this, including finding targets that are easily druggable and selecting combinations that have an acceptable toxicity profile [ ].

A consideration of these practicalities at the preclinical phase may accelerate the selection of new strategies that can be practically and rapidly translated to the clinic. As we have seen, the biology determining response and resistance to anti-angiogenic therapy is complex.

It is perhaps therefore unsurprising that predictive biomarkers for this class of agent remain elusive.

To identify which patients will benefit from these therapies, mechanism-driven biomarkers are required that can account for the dynamic and complex underlying biology. Importantly, as more and more promising biomarkers are uncovered, a further challenge will be to standardise methods of biomarker assessment across centres so that they can be validated prospectively and, eventually, utilised routinely.

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

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

germline SNPs, d circulating markers acquired at baseline and during therapy, and e functional imaging data acquired both at baseline and during therapy. Moreover, in a world where multiple targeted agents are now potentially available for tailored treatment, the decision to use anti-angiogenic therapy will need to be weighed against the use of other potentially effective treatment options for each patient.

Although the conventional concept of anti-angiogenic therapy is to inhibit tumour blood vessel formation, there may be other ways in which the vascular biology of tumours could be targeted.

Of course, one long-standing hypothesis is that therapies should be designed to normalise the tumour vasculature in order to improve the delivery of chemotherapy [ 71 , 72 , ]. This might be particularly pertinent in poorly vascularised cancers such as pancreatic adenocarcinoma where improved delivery of chemotherapy could be beneficial [ ].

Moreover, vascular normalisation may have additional beneficial effects for controlling oedema or tumour oxygenation [ 74 , 75 ]. In addition, it is now known that blood vessels are not merely passive conduits for the delivery of oxygen and nutrients.

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

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

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

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

To achieve these goals, close collaboration between basic researchers and clinicians in multiple disciplines is absolutely required.

Folkman J Tumor angiogenesis: therapeutic implications. N Engl J Med 21 — CAS PubMed Google Scholar. Carmeliet P, Jain RK Molecular mechanisms and clinical applications of angiogenesis.

Nature — CAS PubMed Central PubMed Google Scholar. Leite de Oliveira R, Hamm A, Mazzone M Growing tumor vessels: more than one way to skin a cat—implications for angiogenesis targeted cancer therapies.

Mol Aspects Med 32 2 — PubMed Google Scholar. Ellis LM, Hicklin DJ VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer 8 8 — Kerbel RS Tumor angiogenesis. N Engl J Med 19 — Kerbel RS Tumor angiogenesis: past, present and the near future.

Carcinogenesis 21 3 — Carmeliet P et al Branching morphogenesis and antiangiogenesis candidates: tip cells lead the way. Nat Rev Clin Oncol 6 6 — Olsson AK et al VEGF receptor signalling—in control of vascular function. Nat Rev Mol Cell Biol 7 5 — Escudier B et al Sorafenib in advanced clear-cell renal-cell carcinoma.

N Engl J Med 2 — Escudier B et al Sorafenib for treatment of renal cell carcinoma: final efficacy and safety results of the phase III treatment approaches in renal cancer global evaluation trial.

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

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

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

N Engl J Med 4 — Raymond E et al Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 6 — Hurwitz H et al Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 23 — Giantonio BJ et al Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin FOLFOX4 for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E J Clin Oncol 25 12 — Saltz LB et al Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study.

J Clin Oncol 26 12 — Cunningham D et al Bevacizumab plus capecitabine versus capecitabine alone in elderly patients with previously untreated metastatic colorectal cancer AVEX : an open-label, randomised phase 3 trial. Lancet Oncol 14 11 — Fischer C et al FLT1 and its ligands VEGFB and PlGF: drug targets for anti-angiogenic therapy?

Nat Rev Cancer 8 12 — Li X et al VEGF-B: a survival, or an angiogenic factor? Cell Adh Migr 3 4 — PubMed Central PubMed Google Scholar. Zhang F et al VEGF-B is dispensable for blood vessel growth but critical for their survival, and VEGF-B targeting inhibits pathological angiogenesis.

Proc Natl Acad Sci USA 15 — Fischer C et al Anti-PlGF inhibits growth of VEGF R -inhibitor-resistant tumors without affecting healthy vessels. Cell 3 — Van Cutsem E et al Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen.

J Clin Oncol 30 28 — Carrato A et al Fluorouracil, leucovorin, and irinotecan plus either sunitinib or placebo in metastatic colorectal cancer: a randomized, phase III trial. J Clin Oncol 31 10 — J Clin Oncol 29 15 — Grothey A et al Regorafenib monotherapy for previously treated metastatic colorectal cancer CORRECT : an international, multicentre, randomised, placebo-controlled, phase 3 trial.

Sandler A et al Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 24 — Reck M et al Phase III trial of cisplatin plus gemcitabine with either placebo or bevacizumab as first-line therapy for nonsquamous non-small-cell lung cancer: AVAil.

J Clin Oncol 27 8 — Reck M et al Overall survival with cisplatin-gemcitabine and bevacizumab or placebo as first-line therapy for nonsquamous non-small-cell lung cancer: results from a randomised phase III trial AVAiL. Ann Oncol 21 9 — Ann Oncol 24 1 — Perren TJ et al A phase 3 trial of bevacizumab in ovarian cancer.

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

J Clin Oncol 30 17 — Miller KD et al Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer. J Clin Oncol 23 4 — Miller K et al Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer.

Miles DW et al Phase III study of bevacizumab plus docetaxel compared with placebo plus docetaxel for the first-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol 28 20 — Robert NJ et al RIBBON randomized, double-blind, placebo-controlled, phase III trial of chemotherapy with or without bevacizumab for first-line treatment of human epidermal growth factor receptor 2-negative, locally recurrent or metastatic breast cancer.

J Clin Oncol 29 10 — Brufsky AM et al RIBBON a randomized, double-blind, placebo-controlled, phase III trial evaluating the efficacy and safety of bevacizumab in combination with chemotherapy for second-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer.

J Clin Oncol 29 32 — Crown JP et al Phase III trial of sunitinib in combination with capecitabine versus capecitabine monotherapy for the treatment of patients with pretreated metastatic breast cancer.

J Clin Oncol 31 23 — Bergh J et al First-line treatment of advanced breast cancer with sunitinib in combination with docetaxel versus docetaxel alone: results of a prospective, randomized phase III study. J Clin Oncol 30 9 — Robert NJ et al Sunitinib plus paclitaxel versus bevacizumab plus paclitaxel for first-line treatment of patients with advanced breast cancer: a phase III, randomized, open-label trial.

Clin Breast Cancer 11 2 — Barrios CH et al Phase III randomized trial of sunitinib versus capecitabine in patients with previously treated HER2-negative advanced breast cancer. Breast Cancer Res Treat 1 — Kim KB et al BEAM: a randomized phase II study evaluating the activity of bevacizumab in combination with carboplatin plus paclitaxel in patients with previously untreated advanced melanoma.

J Clin Oncol 30 1 — Flaherty KT et al Phase III trial of carboplatin and paclitaxel with or without sorafenib in metastatic melanoma.

J Clin Oncol 31 3 — Hauschild A et al Results of a phase III, randomized, placebo-controlled study of sorafenib in combination with carboplatin and paclitaxel as second-line treatment in patients with unresectable stage III or stage IV melanoma.

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

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

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

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

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

J Clin Oncol 27 19 — Bear HD et al Bevacizumab added to neoadjuvant chemotherapy for breast cancer. von Minckwitz G et al Neoadjuvant chemotherapy and bevacizumab for HER2-negative breast cancer. Google Scholar. Grunhagen D et al The history of adoption of hepatic resection for metastatic colorectal cancer: — Crit Rev Oncol Hematol 86 3 — Nordlinger B et al Combination of surgery and chemotherapy and the role of targeted agents in the treatment of patients with colorectal liver metastases: recommendations from an expert panel.

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

Ann Oncol 22 9 — Gruenberger T, Arnold D, Rubbia-Brandt L Pathologic response to bevacizumab-containing chemotherapy in patients with colorectal liver metastases and its correlation with survival.

Surg Oncol 21 4 — Loupakis F et al Histopathologic evaluation of liver metastases from colorectal cancer in patients treated with FOLFOXIRI plus bevacizumab. Br J Cancer 12 — Kaye SB Bevacizumab for the treatment of epithelial ovarian cancer: will this be its finest hour?

J Clin Oncol 25 33 — Jain RK Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat Med 7 9 — Jain RK Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science — Van der Veldt AA et al Rapid decrease in delivery of chemotherapy to tumors after anti-VEGF therapy: implications for scheduling of anti-angiogenic drugs.

Cancer Cell 21 1 — Kamoun WS et al Edema control by cediranib, a vascular endothelial growth factor receptor-targeted kinase inhibitor, prolongs survival despite persistent brain tumor growth in mice. J Clin Oncol 27 15 — Batchelor TT et al Improved tumor oxygenation and survival in glioblastoma patients who show increased blood perfusion after cediranib and chemoradiation.

Proc Natl Acad Sci USA 47 — Shaked Y et al Rapid chemotherapy-induced acute endothelial progenitor cell mobilization: implications for antiangiogenic drugs as chemosensitizing agents. Cancer Cell 14 3 — Alishekevitz D, et al. Mol Cancer Ther 13 1 — Smith NR, et al.

Clin Cancer Res 19 24 — Rugo HS Inhibiting angiogenesis in breast cancer: the beginning of the end or the end of the beginning? Rossari JR et al Bevacizumab and breast cancer: a meta-analysis of first-line phase III studies and a critical reappraisal of available evidence. J Oncol Chen HX, Cleck JN Adverse effects of anticancer agents that target the VEGF pathway.

Nat Rev Clin Oncol 6 8 — Hutson TE et al Targeted therapies for metastatic renal cell carcinoma: an overview of toxicity and dosing strategies. Oncologist 13 10 — Dienstmann R et al Toxicity as a biomarker of efficacy of molecular targeted therapies: focus on EGFR and VEGF inhibiting anticancer drugs.

Oncologist 16 12 — Schuster C et al Clinical efficacy and safety of bevacizumab monotherapy in patients with metastatic melanoma: predictive importance of induced early hypertension. PLoS ONE 7 6 :e Rini BI et al Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib.

J Natl Cancer Inst 9 — Osterlund P et al Hypertension and overall survival in metastatic colorectal cancer patients treated with bevacizumab-containing chemotherapy.

Br J Cancer 4 — Mancuso MR et al Rapid vascular regrowth in tumors after reversal of VEGF inhibition. J Clin Invest 10 — Griffioen AW et al Rapid angiogenesis onset after discontinuation of sunitinib treatment of renal cell carcinoma patients.

Clin Cancer Res 18 14 — Wolter P et al Flare-up: an often unreported phenomenon nevertheless familiar to oncologists prescribing tyrosine kinase inhibitors.

Acta Oncol 48 4 — Desar IM et al The reverse side of the victory: flare up of symptoms after discontinuation of sunitinib or sorafenib in renal cell cancer patients.

A report of three cases. Acta Oncol 48 6 — Grothey A et al Bevacizumab beyond first progression is associated with prolonged overall survival in metastatic colorectal cancer: results from a large observational cohort study BRiTE. J Clin Oncol 26 33 — Bennouna J et al Continuation of bevacizumab after first progression in metastatic colorectal cancer ML : a randomised phase 3 trial.

Lancet Oncol 14 1 — Rini BI et al Phase II study of axitinib in sorafenib-refractory metastatic renal cell carcinoma. J Clin Oncol 27 27 — Rini BI et al Antitumor activity and biomarker analysis of sunitinib in patients with bevacizumab-refractory metastatic renal cell carcinoma.

J Clin Oncol 26 22 — Di Lorenzo G et al Phase II study of sorafenib in patients with sunitinib-refractory metastatic renal cell cancer.

Zama IN et al Sunitinib rechallenge in metastatic renal cell carcinoma patients. Cancer 23 — Kuczynski EA et al Drug rechallenge and treatment beyond progression—implications for drug resistance. Nat Rev Clin Oncol 10 10 — Tang TC et al Development of a resistance-like phenotype to sorafenib by human hepatocellular carcinoma cells is reversible and can be delayed by metronomic UFT chemotherapy.

Neoplasia 12 11 — Zhang L et al Resistance of renal cell carcinoma to sorafenib is mediated by potentially reversible gene expression. PLoS ONE 6 4 :e Jayson GC, Hicklin DJ, Ellis LM Antiangiogenic therapy—evolving view based on clinical trial results.

Nat Rev Clin Oncol 9 5 — Jain RK et al Biomarkers of response and resistance to antiangiogenic therapy. Jubb AM, Harris AL Biomarkers to predict the clinical efficacy of bevacizumab in cancer. Lancet Oncol 11 12 — Hegde PS et al Predictive impact of circulating vascular endothelial growth factor in four phase III trials evaluating bevacizumab.

Clin Cancer Res 19 4 — J Clin Oncol 31 14 — Miles DW et al Biomarker results from the AVADO phase 3 trial of first-line bevacizumab plus docetaxel for HER2-negative metastatic breast cancer. Br J Cancer 5 — Van Cutsem E et al Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a biomarker evaluation from the AVAGAST randomized phase III trial.

Tran HT et al Prognostic or predictive plasma cytokines and angiogenic factors for patients treated with pazopanib for metastatic renal-cell cancer: a retrospective analysis of phase 2 and phase 3 trials.

Lancet Oncol 13 8 — Collinson F et al Predicting response to bevacizumab in ovarian cancer: a panel of potential biomarkers informing treatment selection. Clin Cancer Res 19 18 — Maru D, Venook AP, Ellis LM Predictive biomarkers for bevacizumab: are we there yet?

Clin Cancer Res 19 11 — Lambrechts D et al VEGF pathway genetic variants as biomarkers of treatment outcome with bevacizumab: an analysis of data from the AViTA and AVOREN randomised trials. Lancet Oncol 13 7 — Beuselinck B, et al.

Acta Oncol 53 1 — Clin Cancer Res 18 24 — Hahn OM et al Dynamic contrast-enhanced magnetic resonance imaging pharmacodynamic biomarker study of sorafenib in metastatic renal carcinoma.

J Clin Oncol 26 28 — Flaherty KT et al Pilot study of DCE-MRI to predict progression-free survival with sorafenib therapy in renal cell carcinoma. Cancer Biol Ther 7 4 — Han KS et al Pretreatment assessment of tumor enhancement on contrast-enhanced computed tomography as a potential predictor of treatment outcome in metastatic renal cell carcinoma patients receiving antiangiogenic therapy.

Cancer 10 — Fournier LS et al Metastatic renal carcinoma: evaluation of antiangiogenic therapy with dynamic contrast-enhanced CT. Radiology 2 — Smith AD, et al. Urol Oncol 7 — Nathan PD et al CT response assessment combining reduction in both size and arterial phase density correlates with time to progression in metastatic renal cancer patients treated with targeted therapies.

Cancer Biol Ther 9 1 — van der Veldt AA et al Choi response criteria for early prediction of clinical outcome in patients with metastatic renal cell cancer treated with sunitinib. Krajewski KM et al Comparison of four early posttherapy imaging changes EPTIC; RECIST 1. Eur Urol — Smith AD et al Morphology, Attenuation, Size, and Structure MASS criteria: assessing response and predicting clinical outcome in metastatic renal cell carcinoma on antiangiogenic targeted therapy.

AJR Am J Roentgenol 6 — Smith AD, Lieber ML, Shah SN Assessing tumor response and detecting recurrence in metastatic renal cell carcinoma on targeted therapy: importance of size and attenuation on contrast-enhanced CT.

AJR Am J Roentgenol 1 — Vasudev NS et al Changes in tumour vessel density upon treatment with anti-angiogenic agents: relationship with response and resistance to therapy.

Chun YS et al Association of computed tomography morphologic criteria with pathologic response and survival in patients treated with bevacizumab for colorectal liver metastases. JAMA 21 — Bergers G, Hanahan D Modes of resistance to anti-angiogenic therapy. Cancer Res 72 8 — Helfrich I et al Resistance to antiangiogenic therapy is directed by vascular phenotype, vessel stabilization, and maturation in malignant melanoma.

J Exp Med 3 — Bergers G et al Benefits of targeting both pericytes and endothelial cells in the tumor vasculature with kinase inhibitors.

J Clin Invest 9 — Erber R et al Combined inhibition of VEGF and PDGF signaling enforces tumor vessel regression by interfering with pericyte-mediated endothelial cell survival mechanisms. FASEB J 18 2 — Welti JC et al Contrasting effects of sunitinib within in vivo models of metastasis.

Angiogenesis 15 4 — Tong RT et al Vascular normalization by vascular endothelial growth factor receptor 2 blockade induces a pressure gradient across the vasculature and improves drug penetration in tumors.

Cancer Res 64 11 — Shaheen RM et al Tyrosine kinase inhibition of multiple angiogenic growth factor receptors improves survival in mice bearing colon cancer liver metastases by inhibition of endothelial cell survival mechanisms.

Cancer Res 61 4 — Winkler F et al Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell 6 6 — Shojaei F et al Bv8 regulates myeloid-cell-dependent tumour angiogenesis.

Cascone T et al Upregulated stromal EGFR and vascular remodeling in mouse xenograft models of angiogenesis inhibitor-resistant human lung adenocarcinoma.

J Clin Invest 4 — Li JL et al DLL4-Notch signaling mediates tumor resistance to anti-VEGF therapy in vivo. Cancer Res 71 18 — Casanovas O et al Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors.

Cancer Cell 8 4 — Welti JC et al Fibroblast growth factor 2 regulates endothelial cell sensitivity to sunitinib. Oncogene 30 10 — Cancer Res 70 24 — Huang D et al Interleukin-8 mediates resistance to antiangiogenic agent sunitinib in renal cell carcinoma.

Cancer Res 70 3 — Crawford Y et al PDGF-C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti-VEGF treatment.

Cancer Cell 15 1 — di Tomaso E et al PDGF-C induces maturation of blood vessels in a model of glioblastoma and attenuates the response to anti-VEGF treatment. PLoS ONE 4 4 :e Kopetz S et al Phase II trial of infusional fluorouracil, irinotecan, and bevacizumab for metastatic colorectal cancer: efficacy and circulating angiogenic biomarkers associated with therapeutic resistance.

J Clin Oncol 28 3 — Porta C et al Changes in circulating pro-angiogenic cytokines, other than VEGF, before progression to sunitinib therapy in advanced renal cell carcinoma patients. Oncology 84 2 — Johnson PJ et al Brivanib versus sorafenib as first-line therapy in patients with unresectable, advanced hepatocellular carcinoma: results from the randomized phase III BRISK-FL study.

J Clin Oncol 31 28 — Llovet JM et al Brivanib in patients with advanced hepatocellular carcinoma who were intolerant to sorafenib or for whom sorafenib failed: results from the randomized phase III BRISK-PS study.

Motzer R Phase 3 trial of dovitinib vs sorafenib in patients with metastatic renal cell carcinoma after 1 prior VEGF pathway-targeted and 1 prior mTOR inhibitor therapy.

Presented at European cancer congress ; September 27—October 1, ; Amsterdam, The Netherlands. Abstract LBA Kalluri R, Zeisberg M Fibroblasts in cancer.

Nat Rev Cancer 6 5 — Coussens LM, Werb Z Inflammation and cancer. De Palma M, Lewis CE Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell 23 3 — De Palma M, Naldini L Role of haematopoietic cells and endothelial progenitors in tumour angiogenesis.

Biochim Biophys Acta 1 — Bissell MJ, Radisky D Putting tumours in context. Nat Rev Cancer 1 1 — Joyce JA, Pollard JW Microenvironmental regulation of metastasis. Nat Rev Cancer 9 4 — Ebos JM, Lee CR, Kerbel RS Tumor and host-mediated pathways of resistance and disease progression in response to antiangiogenic therapy.

Clin Cancer Res 15 16 — Shojaei F et al Role of myeloid cells in tumor angiogenesis and growth. Trends Cell Biol 18 8 — McMillin DW, Negri JM, Mitsiades CS The role of tumour-stromal interactions in modifying drug response: challenges and opportunities.

Nat Rev Drug Discov 12 3 — Reynolds AR Potential relevance of bell-shaped and u-shaped dose-responses for the therapeutic targeting of angiogenesis in cancer. Dose Response 8 3 — Nat Biotechnol 25 8 — Chung AS et al An interleukinmediated paracrine network promotes tumor resistance to anti-angiogenic therapy.

Nat Med 19 9 — Shaked Y et al Therapy-induced acute recruitment of circulating endothelial progenitor cells to tumors. Lyden D et al Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nat Med 7 11 — Cancer Cell 6 4 — Bailey AS et al Myeloid lineage progenitors give rise to vascular endothelium.

Proc Natl Acad Sci USA 35 — Yu JL et al Effect of p53 status on tumor response to antiangiogenic therapy. Yu JL et al Heterogeneous vascular dependence of tumor cell populations. Am J Pathol 4 — McIntyre A et al Carbonic anhydrase IX promotes tumor growth and necrosis in vivo and inhibition enhances anti-VEGF therapy.

Clin Cancer Res 18 11 — Nardo G et al Glycolytic phenotype and AMP kinase modify the pathologic response of tumor xenografts to VEGF neutralization. Cancer Res 71 12 — Hu YL et al Hypoxia-induced autophagy promotes tumor cell survival and adaptation to antiangiogenic treatment in glioblastoma.

Cancer Res 72 7 — Rapisarda A, Melillo G Overcoming disappointing results with antiangiogenic therapy by targeting hypoxia. Nat Rev Clin Oncol 9 7 — Xu J et al Colorectal cancer cells refractory to anti-VEGF treatment are vulnerable to glycolytic blockade due to persistent impairment of mitochondria.

Mol Cancer Ther 12 5 — Cancer Res 62 7 — Dome B et al Alternative vascularization mechanisms in cancer: pathology and therapeutic implications. Am J Pathol 1 :1— Donnem T et al Vessel co-option in primary human tumors and metastases: an obstacle to effective anti-angiogenic treatment?

Cancer Med 2 4 — Ribatti D et al Microvascular density, vascular endothelial growth factor immunoreactivity in tumor cells, vessel diameter and intussusceptive microvascular growth in primary melanoma. Oncol Rep 14 1 — Nico B et al Intussusceptive microvascular growth in human glioma.

Clin Exp Med 10 2 — Straume O et al Prognostic importance of glomeruloid microvascular proliferation indicates an aggressive angiogenic phenotype in human cancers.

Cancer Res 62 23 — Folberg R, Maniotis AJ Vasculogenic mimicry. APMIS 7—8 — Wang R et al Glioblastoma stem-like cells give rise to tumour endothelium. Ricci-Vitiani L et al Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells.

Cheng L et al Glioblastoma stem cells generate vascular pericytes to support vessel function and tumor growth. Cell 1 — Kilarski WW et al Biomechanical regulation of blood vessel growth during tissue vascularization. Nat Med 15 6 — Cancer Res 71 1 — de Groot JF et al Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice.

Neuro Oncol 12 3 — Pezzella F et al Non-small-cell lung carcinoma tumor growth without morphological evidence of neo-angiogenesis.

Am J Pathol 5 — Cancer cells lacking blood circulation expanded 1—2 mm 3 in diameter and afterward halted, but when put in a location where angiogenesis was feasible, they expanded to more than 2 mm 3.

Given that tumors become necrotic or even apoptotic in the absence of a circulatory supply [ 8 ], it has strongly been validated that angiogenesis is a critical component in cancer development. Tumors differ significantly in the patterns and characteristics of the angiogenic vascular system, as well as their sensitivity to anti-angiogenic treatment [ 9 ].

Cancer cells control the angiogenic programming of neoplastic tissues through collaboration with a range of tumor-associated stromal cells as well as their bioactive products, which include cytokines and growth hormones, the extracellular matrix, as well as secreted microvesicles [ 10 ].

Apart from cancer immunotherapy or other pioneering approaches such as chemotherapy and radiotherapy, which have resulted in a significant advance in cancer treatment [ 11 , 12 ], another potential treatment approach is anti-angiogenesis, which aims to impair the vasculature and deprive the tumor of oxygen and nutrition [ 13 ].

This is accomplished mostly by targeting the pro-angiogenic factors-induced signaling pathway, which is prominent in the tumor microenvironment under hypoxic conditions [ 14 ]. VEGF family members are the regulator of angiogenesis both under normal circumstances and in a disease condition.

This family consists of VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, and placenta growth factor PlGF , which binds with divergent affinities and specificities to tyrosine kinase receptors VEGFR 1,-2, and -3 [ 16 , 17 ]. The interfaces between VEGF-A and VEGFR 2 exceed angiogenesis, while VEGF-C and D preferentially make connections with VEGFR-3 [ 18 ].

The improved expression of VEGF inspires tumourigenesis by potentiating the epithelial-mesenchymal transition EMT activation. In addition to VEGF receptor tyrosine kinases, the neuropilins NRPs , potent co-receptors for class 3 semaphorins, are crucial for exerting the impacts of VEGF on cancer cells as a result of their capability to affect the activities of growth factor receptors and integrins [ 19 ].

Meanwhile, multitargeted small-molecule TKI can target multiple receptor sites simultaneously. The main targets included vascular endothelial growth factor receptor VEGFR , platelet-derived growth factor receptor PDGFR , fibroblast growth factor receptor FGFR , c-Kit, and c-Met.

Anti-angiogenic TKIs block the kinase activity of receptors and transduction of downstream signals involved in the proliferation, migration, and survival [ 22 ]. However, monotherapy with an anti-angiogenic drug has shown minimal therapeutic advantages for most cancer patients [ 23 ]. Thereby, it has been suggested and also evidenced that combining anti-angiogenic medicines with other strategies, comprising immune checkpoint inhibitors ICIs , chemotherapy, human epidermal growth factor receptor 2 HER2 -targeted therapies, adoptive cell transfer ACT , cancer vaccines, and also radiotherapy may have a synergistic anti-tumor impact [ 24 ].

This review highlights current knowledge and clinical developments of anti-angiogenesis combination treatment, either alone or in conjunction with other modalities, focusing on last decade in vivo reports. The central role of VEGF in tumor angiogenesis. The VEGF induces angiogenesis in tumor cells following interaction with responding receptor, VEGFR2, on tumor cells and subsequently by activating various signaling axes.

Several successive stages throughout tumor angiogenesis may be emphasized. The vessel wall of mature capillaries comprises an endothelial cell lining, a basement membrane, and a layer of cells termed pericytes that partly surround the endothelium [ 25 ].

Pericytes share the same basement membrane as endothelial cells and sometimes come into touch with them. Tumor-derived angiogenic agents attach to endothelial cell receptors, initiating the angiogenesis process.

VEGF, fibroblast growth factors FGF , tumour necrosis factor α TNFα , transforming growth factor TGF-β , and angiopoietin Ang are the most well-known angiogenic cytokines and growth factors [ 26 , 27 ]. When endothelial cells are encouraged to develop, proteases, heparanase, as well as other digestive enzymes are secreted, which break down the underlying membrane that surrounds the artery [ 28 , 29 ].

Matrix metalloproteinases MMPs , a class of metalloendopeptidase produced by tumor cells and supportive cells, allow for the degradation of the basement membrane as well as the extracellular matrix surrounding pre-existing capillaries, typically postcapillary venules [ 30 , 31 ].

The breakdown of the extracellular matrix also enables the discharge of pro-angiogenic factors out from the matrix. Endothelial cell connections change, cell extensions cross through the gap produced, and the recently created sprout develops towards the source of the stimulation [ 32 ].

Endothelial cells enter the matrix and start migrating and proliferating inside the tumor mass. Freshly created endothelial cells arrange into hollow tubes and produce a new basement membrane for vascular stability at this site [ 33 ]. The blood flow inside the tumor is formed by freshly shaped fused blood vessels.

Significant interactions between cell-associated surface proteins and the extracellular matrix promote the development of the lumen during canalization. Hybrid oligosaccharides galectin-2, platelet endothelial cell adhesion molecule-1 PECAM-1 or CD31 , and VE-cadherin are among the surface proteins discovered in this interaction [ 34 , 35 ].

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

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

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

These findings suggest that cytokines have a variety of roles in tumorigenesis as well as development. Numerous interleukin 1 IL-1 family members stimulate tumor angiogenesis [ 38 ]. Through the activity of nuclear factor-kappa B NF-κB , p38 mitogen-activated protein kinase MAPK signaling, and Janus kinase JAK , IL-1 signaling stimulates angiogenesis by upregulating VEGF as well as angiogenesis-related molecules [ 39 , 40 ].

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

Researchers discovered that co-treatment with hypoxia-inducible factor 1 HIF-1 inhibitors and bevacizumab had a greater anticancer impact than therapy with bevacizumab separately in glioma xenografts [ 42 ]. HIF-1 is an upstream regulator of many angiogenic factors that may directly stimulate angiogenic factor transcription to enhance tumor angiogenesis [ 43 ].

Furthermore, various hypoxia-induced lncRNAs may enhance tumor angiogenesis by influencing angiogenic factor expression [ 44 ]. As angiogenic factors abound in the tumor microenvironment, treating cancer cells with medicines that target several angiogenic agents may result in improved outcomes.

In contrast, tumor-secreted cytokines largely stimulate a proangiogenic and protumorigenic phenotype of the tumor-associated inflammatory infiltrate. Recently, Wang et al.

The contrast effects of immune cells found in TME on tumor progress. While TH2 and M2 macrophages convince tumor angiogenesis, TH1 and M1 macrophage suppress tumor angiogenesis by secreting a diversity of cytokines. Upon successful preclinical studies Table 1 , a myriad of clinical trials have been accomplished or are ongoing to determine the safety, feasibility, and efficacy of anti-angiogenic agents therapy in cancer patients alone or in combination with other therapeutic means Table 2.

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

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

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

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

In June , it was approved with fluorouracil 5-FU -based therapy for second-line mCRC. Also, it has been indicated for NSCLC plus chemotherapy , breast cancer, glioblastoma, ovarian cancer plus chemotherapy , and also cervical cancer [ 51 ]. Another well-known angiogenesis inhibitor, axitinib, has gained approval from FDA for use as a treatment for renal cell carcinoma RCC since January and also has shown promising outcomes in pancreatic cancer plus gemcitabine [ 52 , 53 ].

Moreover, since , it is used for neuroendocrine tumors NET of gastrointestinal GI or lung origin with unresectable, locally advanced, or metastatic disease [ 55 ].

In November , cabozantinib, a small molecule inhibitor of the tyrosine kinases c-Met and VEGFR2, was approved for thyroid cancer [ 56 ] and also in April was accepted as second-line treatment for RCC [ 57 ].

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

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

Besides, since April , the ramucirumab, a direct VEGFR2 antagonist, is indicated as a single-agent treatment for advanced gastric cancer or gastro-esophageal junction GEJ adenocarcinoma after treatment with fluoropyrimidine- or platinum-containing chemotherapy [ 63 ].

Further, ramucirumab in combination with docetaxel has gained approval for treatment of metastatic NSCLC [ 64 ]. Ramucirumab also is used for mCRC since [ 65 ] and HCC since [ 66 ] therapy.

Also, regorafenib, an orally-administered inhibitor of multiple kinases, has been indicated for the treatment of patients with advanced HCC who were previously treated with sorafenib [ 67 ].

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

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

Finally, tyrosine kinase inhibitor vandetanib is employed to treat medullary thyroid cancer in adults who are ineligible for surgery [ 72 , 73 ].

Despite their total tumor growth reduction, therapeutic anti-angiogenic agents were linked to enhanced local invasiveness as well as distant metastasis.

These events seem to be significant factors to resistance to anti-angiogenesis treatments. They were originally reported in various preclinical models by Paez-Ribes and coworkers [ 74 ]. Based on the literature, anti-angiogenic treatment may increase tumor invasiveness.

RCC cells, for example, showed increased proliferation and an invasive character after being treated with bevacizumab [ 75 ]. Likewise, glioblastoma cells in mice models were more invasive after VEGF suppression [ 74 ].

Sunitinib treatment also has been found to cause vascular alterations such as decreased adherens junction protein expression, reduced basement membrane, pericyte coverage, and increased leakiness [ 76 , 77 ].

These phenotypic alterations were found in both normal and tumor organ arteries, indicating that they promote tumor cell local intravasation and extravasation, culminating in metastatic colonization [ 78 ]. Angiogenesis blockade therapy may lead to vascular regression and resultant intra-tumoral hypoxia.

Various investigations have been fulfilled to assess an enhancement in hypoxic areas in primary tumors upon angiogenesis blockade therapy [ 76 , 79 ]. Further investigation also exposed an attendant augmentation in HIF-1a expression during treatment.

HIF-1a and hypoxia are recognized drivers of epithelial-mesenchymal transition EMT , a process that induced tumor metastasis. Significant improvement in the expression and activities of EMT-related genes e.

Moreover, loss of the epithelial marker, E-cadherin, and the stimulation of the mesenchymal marker, vimentin, has been evidenced following anti-angiogenic treatment [ 80 ].

Hypoxic milieu also largely promotes VEGF expression by the upstream transcription factor HIF-1a [ 81 ]. HIF-1, in turn, inspires tumors to achieve more angiogenic and invasive competencies, culminating in metastasis [ 82 ]. In fact, hypoxia and EMT bring about increased invasiveness and metastasis of tumors mainly caused by up-regulation of c-Met, Twist, and HIF-1a [ 83 , 84 ].

Conversely, semaphorin 3A Sema3A , a well-known endogenous anti-angiogenic molecule, is substantially down-regulated in tumors, ensuring provoked invasiveness and metastasis [ 85 ].

Ang-Tie signaling system is a vascular-specific receptor tyrosine kinases RTK pathway complicated in modifying the vascular permeability and blood vessel formation and remodeling by potent angiogenic growth factors, Ang-1 and Ang-2 [ 86 ].

Molecular analysis has confirmed that activation of the Ang-Tie pathway as a result of the connection between Ang-1 and Tie2 receptor on the M2 subpopulation of monocytes, hematopoietic stem cells HSCs , and endothelial cells ECs of blood and lymphatic vessels elicits maturation or stabilization of blood vessels [ 80 ].

Besides, Ang-2 suppresses this pathway, eventually sustaining remodeling or generation of vascular sprouts upon exposure to VEGF [ 87 ].

Ang-2 up-regulation has been noticed in multiple types of tumors and is likely involved in resistance versus anti-VEGF therapy [ 88 , 89 ]. For instance, there is clear evidence signifying that enhanced serum Ang-2 levels are in association with an undesired response to bevacizumab therapy in CRC patients [ 90 ].

Studies in lung adenocarcinoma patients revealed that elevated levels of VEGFA and Ang-2 is valued prognostic biomarkers and double targeting of VEGFA and Ang-2 can improve therapeutic outcome [ 91 ]. As well, up-regulation and compensatory mechanisms of other growth factors, in particular basic fibroblast growth factor bFGF , are thought to contribute to the stimulation of the resistance to VEGF targeted therapies.

Improved level of the bFGF has strongly been evidenced in the chronic inflammation area, after tissue injury, as well as human cancers bevacizumab [ 92 ]. Upon bevacizumab treatment in glioblastoma tumor models, Okamoto et al. showed the increased levels of the bFGF and PDGF expression in the endothelial cells, pericytes, and also tumor cells, in turn, caused robust resistance to bevacizumab [ 94 ].

Also, cancer patients with up-regulated bFGF in serum usually show no desired response to sunitinib, indicating the necessity of co-targeting VEGF and bFGF pathways concurrently [ 95 , 96 ].

Increased metastasis and invasiveness in response to anti-angiogenesis therapy vary according to treatment type, dosage, and schedule. Sunitinib and anti-VEGF antibody monotherapy showed varied effects on mice tumor models, according to Singh et al.

reports [ 77 ]. While sunitinib therapy increased tumor cell aggressiveness, anti-VEGF antibody treatment did not [ 77 ]. Chung et al. also corroborated these findings by comparing the effectiveness of several RTK inhibitors and antibody treatments in mouse models [ 97 ].

Though imatinib, sorafenib, or sunitinib increased lung metastasis after 66c14 cell injection, employing an anti-VEGFR2 antibody reduced the development of lung nodules [ 97 ]. Overall, reports show that the increased metastasis and invasiveness caused by angiogenesis blockade therapy depend highly on the treatment type.

Anti-angiogenic drug dosage and delivery schedules may also potentially cause resistance. Sunitinib at high doses accelerated tumor development and facilitated metastasis to the lung and liver, resulting in decreased survival [ 74 , 98 ].

Although sorafenib had comparable outcomes, sunitinib produced conflicting findings in various trials. High-dose sunitinib therapy before systemic injection of tumor cells enhanced the metastatic potential of lung cancer cells, but not RCC cells.

Recently, scientists have concentrated on the role of immune checkpoint molecules, such as cytotoxic T-lymphocyte antigen-4 CTLA-4 and programmed cell death protein 1 PD-1 , largely participating in tumor cell escape from immune surveillance as their capacity to obstruct T cell activation [ 99 , ].

Hence, immune checkpoint inhibitors ICIs have been evolved for suppressing these immune checkpoint molecules [ ].

FDA-approved ICIs comprise the nivolumab, cemiplimab, and pembrolizumab, atezolizumab, avelumab, durvalumab, and also ipilimumab [ ]. Atezolizumab has been approved for use in combination with bevacizumab, paclitaxel, and carboplatin as the first-line treatment of patients with NSCLC [ ]. PD-L1 expression is regulated by various factors, such as inflammatory and oncogenic signaling, leading to the varied significances of PD-L1 positivity.

Recent reports exhibited that combination therapy with anti-angiogenic agents and ICIs could elicit synergistic anti-tumor effects in preclinical models as well as humans Table 3. Meanwhile, co-administration of anti-PD-1 and anti-VEGFR2 monoclonal antibodies mAbs in the Colon adenocarcinoma mice model gave rise to the potent inhibition of tumor growth synergistically without overt toxicity [ ].

VEGFR2 blockade therapy negatively regulated tumor neovascularization, as evidenced by the attenuated frequencies of microvessels, whereas PD-1 inhibition exerted no effect on tumor angiogenesis. PD-1 mAbs improved T cell infiltration into tumors and promoted local immune response, as documented via the improvement in various proinflammatory cytokine expressions.

Such events signified that concurrent suppression of PD-1 and VEGFR2 might inspire synergistic in vivo anti-tumor influences by dissimilar mechanisms [ ]. Further, in a mouse model of small-cell lung cancer SCLC , co-administration of anti-VEGF and anti-PD-L1 mAbs resulted in a more prominent therapeutic outcome than mono therapy with each agent [ ].

Notably, the depleted T-cell phenotype following anti-PD-L1 therapy was revoked through the addition of anti-VEGF blockade therapy. Analysis revealed that VEGFA expression improves the expression of the inhibitory receptor TIM-3 on T cells, representative of an immunosuppressive action of VEGF in patients with SCLC during PD-1 blockade therapy.

Thereby, it seems that VEGFA inhibition may entice T cell activation at higher levels, facilitating T cell-mediated anti-tumor immunity [ ]. Similarly, combination therapy with sunitinib and PD-L1 blocked therapy prolonged overall survival OS of treated RCC mice models in comparison to mono therapy with either drug [ ].

Besides, in the triple-negative breast cancer TNBC mice model, PD-L1 blocking was highly effective as an adjuvant monotherapy. However, its co-administration with paclitaxel chemotherapy with or without VEGF blocked therapy showed superiority over neoadjuvant therapy [ ].

In , Schmittnaegel et al. also noticed that dual Ang-2 and VEGFA inhibition induced antitumor immunity that was promoted by PD-1 blockade therapy in breast cancer, pancreatic neuroendocrine tumor, and melanoma [ ]. They showed that Ang-2 and VEGFA blockade by a bispecific antibody A2V caused vascular regression, tumor necrosis along with improved antigen presentation by intratumoral phagocytes [ ].

Recent clinical trials have also shown that bevacizumab plus atezolizumab could induce synergistic influence on the median OS of patients with RCC [ ], and also in combination with nivolumab could elicit modest efficacy in ovarian cancer patients [ ]. Also, co-administration of PD-L1 inhibitor avelumab with axitinib resulted in improved objective response rate ORR in HCC [ ] and also RCC [ ] patients, with acceptable safety profile.

Also, combination therapy with axitinib and pembrolizumab enhanced median progression-free survival PFS in sarcoma patients more evidently than axitinib or pembrolizumab monotherapy.

The most common treatment-related unwanted events were autoimmune colitis, pneumothorax, transaminitis, seizures, hemoptysis, and hypertriglyceridemia [ ]. Besides, co-administration of regorafenib plus nivolumab resulted in significant antitumor impacts in patients with gastric cancer and CRC [ ].

Moreover, co-administration of nivolumab plus sunitinib or pazopanib showed a significant anti-tumor effect in advanced RCC patients [ , ].

Conversely, other trials revealed that combined use of regorafenib plus nivolumab [ ] and also ramucirumab plus pembrolizumab [ ] had no remarkable therapeutic merits in CRC patients [ ] and patients with advanced biliary tract cancer BTC [ ], respectively.

Therapeutic cancer vaccines ease tumor regression, remove minimal residual disease MRD , entice durable antitumor memory, and also averts non-specific or adverse events [ , ]. Till, FDA has approved three cancer vaccines, comprising Bacillus Calmette-Guérin BCG lives, sipuleucel-T, and also talimogene laherparepvec T-VEC respectively for patients with early-stage bladder cancer, prostate cancer as well as melanoma [ ].

In the melanoma mice model, Bose and coworkers found that a treatment regimen comprising a 7-day course of axitinib 0. These desired outcomes are probably exerted by a decrease in myeloid-derived suppressor cells MDSC and Treg frequencies in the tumor concomitant with induction and recruitment of CTLs in TME [ ].

Also, addition of the axitinib to oncolytic herpes simplex virus oHSV expressing murine IL12 G47Δ-mIL12 triggered improved OS in both immunodeficient and immunocompetent orthotopic glioblastoma mice models than mice receiving monotherapy [ ]. Notably, the addition of the ICI did not promote efficacy in mice models [ ].

As well, combination therapy with sunitinib and vesicular stomatitis virus VSV brought about the eradication of prostate, breast, and kidney malignant tumors in mice, while monotherapy with VSV or sunitinib did not [ ].

Importantly, enhancement in median viral titers by fold following combination therapy indicated that this regimen could potentiate oncolytic virotherapy permitting the recovery of tumor-bearing animals.

In RCC and NSCLC mice model, co-injection of reovirus and sunitinib more potently attenuated tumor burden supporting improved OS, and also reduced the population of immune suppressor cells in tumors compared with monotherapy with reovirus [ ].

Thereby, it appears that this regimen can be a rational and effective strategy ready for clinical testing against RCC and NSCLC. Also, Tan and coworkers showed that the bevacizumab improved viral distribution and also tumor hypoxia and promoted the population of apoptotic cells and thus stimulated a synergistic antitumor impact when used in combination with oHSV in TNBC murine models [ ].

Combining bevacizumab with OHSV expressing vasculostatin RAMBO also demonstrated great anti-tumor capacities in glioma xenografts [ ]. Correspondingly, intratumorally administration of RAMBO 1 week after tumor inoculation, and intraperitoneally administration of bevacizumab twice a week reduced migration as well as invasion of glioma cells [ ].

Co-treated mice also experienced improved OS and dampened tumor invasion than those treated with bevacizumab alone [ ]. In another study, combining tumor antigen-loaded DCs vaccination and anti-angiogenic molecule lenalidomide synergistically potentiated antitumor immunity in the mice colon cancer model, largely provided by suppressing the establishment of immune suppressive cells and also activation of effector cells, such as natural killer NK cells [ ].

This regimen similarly caused a robust reduction in tumor growth and malignant cell spread in lymphoma [ ] and also myeloma [ ] xenografts by similar mechanisms. Further, lenalidomide in combination with a fusion DNA lymphoma vaccine reduced the systemic population of MDSC and Treg in tumor-bearing mice and also led to the decreased tumor burden [ ].

In addition, the combination therapy supported the incidence of the higher rates of the antitumor T cells, providing further rationale for clinical application [ ]. Currently, a clinical trial was conducted to address the safety and efficacy of combination therapy with sipuleucel-T as a cellular prostate cancer vaccine with bevacizumab in 22 prostate cancer patients [ ].

Combination therapy persuaded immune reactions and also alleviated prostate-specific antigen PSA in participants with biochemically recurrent prostate cancer [ ].

In contrast, co-administration of bevacizumab plus MA multi-peptide vaccine adjuvanted with poly-ICLC polyinosinic-polycytidylic acid stabilized with polylysine and carboxymethylcellulose did not show superiority over monotherapy with each agent in terms of alteration in OS and PFS in glioblastoma patients [ ].

However, a phase II study evaluating the safety and efficacy of bevacizumab in combination with ERC, advanced immunotherapy based on freshly extracted tumor cells and lysates, revealed that this regimen could prolong the OS in patients who received ERC plus bevacizumab compared to bevacizumab monotherapy 12 months versus 7.

Besides, evaluation of the safety, tolerability, and anti-myeloma activity of the PVX, a novel tetra-peptide vaccine with 3 of the 4 antigens XBP1 [2 splice variants] and CD with or without lenalidomide was accomplished in MM patients by Nooka et al.

They showed that the PVX vaccine was well tolerated, accompanied by mild injection site reactions and constitutional symptoms. Meanwhile, 5 of 12 patients showed clinical response to combination therapy [ ]. Importantly, CRC patients presented complete pathological remission following treatment with bevacizumab, oxaliplatin plus leucovorin and 5-fluorouracil FOLFOX-4 , surgery, and the oncolytic virus Rigvir [ ].

In consistence with previous findings, it appears that angiogenesis blockade therapy could promote viral delivery through targeting the TME [ ]. Adoptive cell therapy ACT with using TILs or genetically-modified T cells expressing novel T cell receptors TCR or chimeric antigen receptors CAR T cells or CAR-NK cells is another plan to convince the immune system to stimulate recognition of the maligned cells and then their eradication [ , ].

Notwithstanding, tumor vasculature usually obstructs the tumor-specific T cells infiltration, averting anti-tumor immunity. In the B16 melanoma mice model, co-administration of anti-VEGF mAb to ACT abrogated tumor progress and improve OS [ ]. Similarly, anti-angiogenic therapy could also improve the antitumor functions of cytokine-induced killer cells CIK cells cells by normalizing tumor vasculature and alleviating the hypoxic TME, as shown in NSCLC xenografts [ ].

Meanwhile, Shi et al. evaluated the therapeutic benefits of combination therapy with recombinant human endostatin rh-endostatin and CIK cells in NSCLC murine model. They exhibited that rh-endostatin normalized tumor vasculature and attenuated hypoxic regions in the TME [ ].

The rh-endostatin markedly potentiated the administrated CIK cells homing and also reduced immune suppressive cells frequency in the tumor tissue. On the other hand, the used regimen instigated a higher level of TILs in tumor tissue [ ]. Further, GD2-redirected CAR T cells plus bevacizumab displayed a remarkable anti-tumor effect in an orthotopic xenograft model of human neuroblastoma [ ].

Co-administration of bevacizumab or ganglioside GD2-CAR T cells or both by single systemic injection supported higher rates of CAR T cells infiltration into tumor tissue accompanied with improved IFN-γ levels in TME.

Additionally, the analysis presented that PD-L1 blockade therapy might augment the efficacy of this regimen [ ]. Likewise, epithelial cell adhesion molecule EpCAM redirected CAR NK cells injection resulted in CRC cell regression in animal models, which was potentiated when used in combination with regorafenib [ ].

These findings delivered a novel plan for the treatment of CRC and also other solid tumors. Anti-angiogenic agents as noticed can transiently stimulate a functional normalization of the disorganized labyrinth of vessels, sustaining the therapeutic efficacy of coadministered chemotherapeutic agents.

Notwithstanding, durable angiogenesis suppression usually fences tumor uptake of chemotherapeutic drugs, and so accomplishment of further studies in this context are urgently required [ ]. Correspondingly, designing intermittent treatment schedules is of paramount significance [ ].

A study in 9L glioma cell-bearing rats showed that coadministration of axitinib with metronomic cyclophosphamide potently suppressed tumor progress, whereas multiple treatment cycles were needed by monotherapy with metronomic cyclophosphamide to abrogate tumor growth [ ].

Unfortunately, the abridged tumor infiltration of 4-OH-CPA resulted in a reduction in cyclophosphamide-mediated 9L cell elimination [ ]. Such events in turn underlined lacking tumor complete regression by applied combined regimen, reflecting the importance of the optimization of drug scheduling and dosages.

In another study, co-administration of the bevacizumab plus cisplatin and paclitaxel concurrently also induced reduced tumor growth as well as improved OS in ovarian cancer xenografts [ ]. Also, monotherapy with bevacizumab suppressed ascites formation, accompanied by the partial impact on tumor burden [ ].

TNP, an angiogenesis inhibitor, plus cisplatin inhibited the liver metastasis of human pancreatic carcinoma [ ]. Indeed, liver metastasis percentages reduced from While monotherapy with each agent did not modify tumor growth in vivo, the addition of TNP to cisplatin strikingly reduced tumor growth [ ].

Of course, it seems that TNP may entice a decrease in glioma tumor uptake of some chemotherapeutic drugs, such as temozolomide, by affecting the tumor vasculature as a result of its pharmacodynamic effect [ ].

As cited, more comprehensive studies are required to define how these combinations can efficiently be utilized. Another study also exhibited that the addition of the TNP to cisplatin chemotherapy reduced the microvascular density of bladder cancer in a murine model [ ].

Nonetheless, TNP has no significant influence on the cisplatin impact versus bladder cancer as determined by apoptosis and cell proliferation [ ]. Besides, Bow and coworkers demonstrated that local delivery of angiogenesis-inhibitor minocycline could potentiate the anti-tumor efficacy of radiotherapy RT and oral temozolomide, as evidenced by enhanced OS in a rodent glioma model [ ].

These findings offered further evidence for the idea that angiogenesis inhibitors in combination with conventional therapeutic modalities could promote OS in glioblastoma patients [ ].

Moreover, the addition of the novel anti-angiogenic agent, SU, to paclitaxel supported improved PFS accompanied with some mild to modest adverse events e. However, the regimen led to the occurrences of thromboembolic events and prophylactic anticoagulation, suggesting that careful consideration must be taken.

Besides, TSU when used plus carboplatin and paclitaxel showed a manageable safety profile in NSCLC patients [ ]. Furthermore, combining TNP and paclitaxel was well tolerated with no significant pharmacokinetic interaction between them in NSCLC patients [ ].

Further, several clinical trials have verified the efficacy of combination therapy with anti-angiogenic agent and conventional therapy in patients with ovarian cancer [ , ], CRC [ , ], NSCLC [ ], MCL [ ] and also MM [ ]. For instance combination therapy with bevacizumab and paclitaxel plus carboplatin prolonged the median OS in participants with platinum-sensitive recurrent ovarian cancer [ ].

Finally, axitinib combined with cisplatin and gemcitabine [ ] and also bevacizumab plus paclitaxel and carboplatin [ ] induced significant anti-tumor effect in NSCLC patients, as documented by improved OS and PFS. In addition, the Ziv-aflibercept in combination with 5-fluorouracil, leucovorin, and irinotecan FOLFIRI significantly promoted OS in a phase III study of patients with metastatic CRC previously treated with an oxaliplatin-based regimen [ ].

However, Ziv-aflibercept in combination with cisplatin and pemetrexed did not significantly affect OS and PFS in patients with previously untreated NSCLC cancer [ ]. A list of trials based on combination therapy with angiogenesis inhibitors plus chemotherapy or chemoradiotherapy has been offered Table 4.

RT crucially contributes to the multimodality treatment of cancer. Current evolving in RT have chiefly complicated improvements in dose delivery [ ].

Upcoming developments in tumor therapeutics will probably include the combination of RT with targeted therapies. Meanwhile, preliminary results of anti-angiogenic agents in combination with RT have produced encouraging consequences [ ]. Further, there are clear proofs that suggest that well-vascularized and perfused tumors mainly exhibit desired response to RT [ , ].

Studies have shown that the addition of the angiogenesis-inhibitor minocycline to radiotherapy and oral temozolomide could result in prolonged OS in a murine glioma model [ ].

Anti-angiogenesis therapy using anginex in combination with RT also supported tumor control in squamous cell carcinoma SCC xenografts accompanied by reducing oxygen levels in tumor tissue [ ]. Observation showed that the applied regimen modified the amount of functional vasculature in tumors and also augmented radiation-elicited tumor eradication [ ].

Likewise, robust hindrance of tumor proliferation was achieved from the addition of the angiogenesis inhibitor TNP to RT in SCC xenografts more evidently than monotherapy with each approach [ ].

Also, it was speculated that exclusive investigation of each tumor neovascularization competence can be imperative before deciding the angiogenesis blockade treatment [ ]. In contrast, the addition of TNP to RT attenuated the tumor control probability in murine mammary carcinoma [ ].

Such unanticipated consequence could be ensured from the partial reserve of reoxygenation by TNP, as no remarkable alteration was shown between the RT plus TNP and RT alone under hypoxic conditions [ ].

A potent anti-angiogenesis agent, liposomal honokiol, also elicited significant anti-tumor influence by stimulating apoptosis and also suppressing angiogenesis when used plus RT in Lewis lung cancer LLC xenografts [ ].

Liposomal honokiol, in fact, could ameliorate tumor cell radiosensitivity in vivo, offering that RT plus liposomal honokiol can engender better anti-tumor efficacy in a myriad of tumors, such as lung cancer, SCC, and CRC [ , , ].

In , Yang et al. evaluated the safety and efficacy of that combination therapy with axitinib plus RT in advanced HCC patients. They exhibited that the regimen was well tolerated with an axitinib MTD of 3 mg twice daily [ ]. Besides, the addition of the bevacizumab to adjuvant radiotherapy was associated with the manageable safety profile in breast cancer patients [ ].

Likewise, erlotinib in combination with bevacizumab as well as capecitabine-based definitive chemoradiation CRT showed acceptable safety in unresectable pancreatic cancer patients [ ]. As well 2 of 9 participants showed complete response to intervention [ ].

Of course, large-scale trials on this newer therapeutic mean seem justified. Albeit there are some reports which show that combining anti-angiogenic therapy with RT had no therapeutic advantages.

For instance, in rectal carcinoma patients, combination therapy with bevacizumab and capecitabine plus RT revealed no merits in terms of improved PFS or OS in the short or long term during a phase 2 clinical trial NCT [ ]. As a result of some divergences results related to anti-angiogenic agents as well as their modest responses, we must determine and categorize a spectrum of biomarkers, screening the patients of possible responders [ ].

Additionally, such biomarkers are urgently required to can monitor disease development and angiogenic actions of tumors following exposure with treatment angiogenesis inhibitors.

There are some reports showing that angiogenesis inhibitors could not support therapeutic effect in previously treated metastatic breast cancer [ ]. These undesired events are likely related to the secretion of pro-angiogenic factors from resistant malignant tissue [ ].

The finding outlines the importance of determining biomarkers to predict the efficacy of VEGF-targeted therapies. Much effort has been spent in this regard and resulted in the finding several biomarkers comprising dynamic measurements such as variations in systemic blood pressure , circulating markers such as VEGF serum levels , genotypic markers such as VEGF polymorphism , blood cells frequencies such as progenitor cells , tissue markers such as IFP and also imaging parameters [such as estimating capillary permeability employing magnetic resonance imaging MRI ] [ ].

Recent studies have revealed that there is a negative correlation between OS with serum lactate dehydrogenase LDH and neutrophil levels in CRC patients who received bevacizumab plus standard chemotherapy [ ].

Besides, enhanced IL-8 levels were associated with shorter PFS, while low Ang-2 serum levels were related to improved OS in tumor patients undergoing angiogenesis blockade therapy [ 90 ].

Circulating endothelial cells CEC also has been determined as a robust indicator for the outcome of treatment with bevacizumab. On the other hand, greater intra-tumoral expression of VEGFR-3 may predict better response, while overexpression of VEGFR1 mainly indicates poor survival [ ].

Other studies in RCC patients upon treatment with sorafenib also revealed that high baseline levels of VEGF were related to poor prognosis [ ], while serum levels of circulating neutrophil gelatinase-associated lipocalin NGAL and VEGF were powerfully supported prolonged PFS in RCC patients receiving sunitinib [ ].

In contrast to the classical hypothesis of vascular regression, the central aim of conventional anti-angiogenic treatments is tumor vascular normalization and maturity.

This event, in turn, offered enhanced tumor access to chemotherapeutic drugs and underlays more efficient cancer immunotherapy. As cited, survival benefits of angiogenesis blockade therapy are compromised by cancer resistance to theses agent, and thereby provoke interest in evolving more effective means to combine anti-angiogenic drugs with other conventional therapeutics.

To date, a large number of clinical trials have evaluated the safety and therapeutic merits of angiogenesis blockade therapy alone or in combination with other modalities in cancer panties Fig.

Although combination therapy regimen mainly caused significant efficacy in cancer patients, intervention-related toxicities hurdle their application in clinic. For instance, bevacizumab therapy could sustain ischemic heart disease.

Indeed, CRC patients receiving bevacizumab may experience considerably augmented possibility of cardiac ischemia [ ]. In addition, it has been proved that combination therapy with angiogenesis inhibitors and chemotherapeutic agents may attenuate antitumor effects of chemotherapy.

Hence, further rigorous investigations are warranted to circumvent the cited problems. Moreover, determining the suitable dose and sequence is of paramount importance to optimize the effectiveness, toxicity, and tolerability of the combination therapy.

Thanks to the involvement of a myriad of cytokines and growth factors and the resultant interplay and compensation among them, co-targeting various growth factors is urgently required. The recognition and potent suppression of downstream kinases and strategic signaling biomolecules where several angiogenic pathways converge may defeat current difficulties motivated via the variety of angiogenic ligands and receptors and should be the emphasis of upcoming investigations.

For instance, dual EGFR inhibition erlotinib and cetuximab combined with bevacizumab is a safe and well-tolerated combination, demonstrating antitumor activity in patients with solid tumors [ ]. BQ13esides, continued treatment with conventional anti-angiogenic agents is related to toxicity and drug resistance.

These conditions offer a robust justification for novel plans to improve the efficacy of mAbs targeting tumor vasculature, such as antibody—drug conjugates ADCs and peptide-drug conjugates PDCs , offering a new avenue to exert anti-angiogenic effects on cancerous cells.

Clinical trials based on cancer therapy by anti-angiogenic agents registered in ClinicalTrials. gov October The schematic exemplifies clinical trials utilizing anti-angiogenic agents depending on the study status A , study phase B , study location C , and condition D in cancer patients.

Folkman J. Annu Rev Med. CAS PubMed Google Scholar. Senger DR, Davis GE. Cold Spring Harb Perspect Biol. PubMed PubMed Central Google Scholar. Aguilar-Cazares D, Chavez-Dominguez R, Carlos-Reyes A, Lopez-Camarillo C, Hernadez de la Cruz ON, Lopez-Gonzalez JS.

Contribution of angiogenesis to inflammation and cancer. Front Oncol. Kerbel RS. Tumor angiogenesis. N Engl J Med. CAS PubMed PubMed Central Google Scholar. Reinmuth N, Parikh AA, Ahmad SA, Liu W, Stoeltzing O, Fan F, et al.

Biology of angiogenesis in tumors of the gastrointestinal tract. Microsc Res Tech. Rajendran JG, Krohn KA. Imaging hypoxia and angiogenesis in tumors. Radiol Clin. Google Scholar. Muthukkaruppan VR, Kubai L, Auerbach R. Tumor-induced neovascularization in the mouse eye J Natl Cancer Inst.

Antiangiogenic therapy of transgenic mice impairs de novo tumor growth. Proc Natl Acad Sci U S A. Teleanu RI, Chircov C, Grumezescu AM, Teleanu DM.

Tumor angiogenesis and anti-angiogenic strategies for cancer treatment. J Clin Med. CAS Google Scholar. Al-Abd AM, Alamoudi AJ, Abdel-Naim AB, Neamatallah TA, Ashour OM. Anti-angiogenic agents for the treatment of solid tumors: potential pathways, therapy and current strategies—a review.

J Adv Res. Dudzinski SO, Cameron BD, Wang J, Rathmell JC, Giorgio TD, Kirschner AN. Combination immunotherapy and radiotherapy causes an abscopal treatment response in a mouse model of castration resistant prostate cancer.

J Immunother Cancer. Lee JJ, Chu E. Adjuvant chemotherapy for stage II colon cancer: the debate goes on. J Oncol Pract. Shahneh FZ, Baradaran B, Zamani F, Aghebati-Maleki L. Tumor angiogenesis and anti-angiogenic therapies.

Hum Antib. Sharma PS, Sharma R, Tyagi T. Curr Cancer Drug Targets. Pro-angiogenic peptides in biomedicine. Arch Biochem Biophys. PubMed Google Scholar. Li X, Eriksson U. Novel VEGF family members: VEGF-B, VEGF-C and VEGF-D.

Int J Biochem Cell Biol. Tomanek RJ, Holifield JS, Reiter RS, Sandra A, Lin JJ. Role of VEGF family members and receptors in coronary vessel formation.

Dev Dyn. 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. Zachary I. Neuropilins: role in signalling, angiogenesis and disease.

Chem Immunol Allergy. Mercurio AM. Int J Mol Sci. CAS PubMed Central Google Scholar. Finley SD, Popel AS. Predicting the effects of anti-angiogenic agents targeting specific VEGF isoforms. AAPS J. Qin S, Li A, Yi M, Yu S, Zhang M, Wu K. Recent advances on anti-angiogenesis receptor tyrosine kinase inhibitors in cancer therapy.

J Hematol Oncol. Dey N, De P, Brian L-J. Evading anti-angiogenic therapy: resistance to anti-angiogenic therapy in solid tumors. Am J Transl Res. Liang P, Ballou B, Lv X, Si W, Bruchez MP, Huang W, et al.

Monotherapy and combination therapy using anti-angiogenic nanoagents to fight cancer. Adv Mater. Ribatti D, Nico B, Crivellato E.

The role of pericytes in angiogenesis. Int J Dev Biol. Wang X, Ma W, Han S, Meng Z, Zhao L, Yin Y, et al. Sci Rep. Kim JH, Kim S-K, Wang K-C. Moyamoya disease update. Tokya: Springer; Jayatilleke KM, Hulett MD. Heparanase and the hallmarks of cancer.

J Transl Med. Vempati P, Popel AS, Mac GF. Extracellular regulation of VEGF: isoforms, proteolysis, and vascular patterning. Cytokine Growth Factor Rev.

Rundhaug JE. Matrix metalloproteinases, angiogenesis, and cancer: commentary re: AC Lockhart et al. Cancer Res. Clin Cancer Res. Matrix metalloproteinases and angiogenesis. J Cell Mol Med.

Tang Y, Nakada MT, Kesavan P, McCabe F, Millar H, Rafferty P, et al. Extracellular matrix metalloproteinase inducer stimulates tumor angiogenesis by elevating vascular endothelial cell growth factor and matrix metalloproteinases.

Can Res. Lamalice L, Le Boeuf F, Huot J. Endothelial cell migration during angiogenesis. Circ Res. Colomb F, Wang W, Simpson D, Zafar M, Beynon R, Rhodes JM, et al. Galectin-3 interacts with the cell-surface glycoprotein CD MCAM, MUC18 and induces secretion of metastasis-promoting cytokines from vascular endothelial cells.

J Biol Chem. Tatum JL, Hoffman JM. Angiogenesis imaging methodology: AIM for clinical trials. Voron T, Marcheteau E, Pernot S, Colussi O, Tartour E, Taieb J, et al.

Control of the immune response by pro-angiogenic factors. Sheu B-C, Chang W-C, Cheng C-Y, Lin H-H, Chang D-Y, Huang S-C. Cytokine regulation networks in the cancer microenvironment. Front Biosci. Fahey E, Doyle SL. IL-1 family cytokine regulation of vascular permeability and angiogenesis.

Front Immunol. Mountain DJ, Singh M, Menon B, Singh K. Am J Physiol Cell Physiol. Carmi Y, Voronov E, Dotan S, Lahat N, Rahat MA, Fogel M, et al.

The role of macrophage-derived IL-1 in induction and maintenance of angiogenesis. J Immunol. Huang Q, Duan I, Qian X, Fan J, Lv Z, Zhang X, et al. IL promotes angiogenic factors IL-6, IL-8, and Vegf production via Stat1 in lung adenocarcinoma.

Rapisarda A, Hollingshead M, Uranchimeg B, Bonomi CA, Borgel SD, Carter JP, et al. Increased antitumor activity of bevacizumab in combination with hypoxia inducible factor-1 inhibition.

Mol Cancer Ther. Yang Y, Sun M, Wang L, Jiao B. HIFs, angiogenesis, and cancer. J Cell Biochem. Niu Y, Bao L, Chen Y, Wang C, Luo M, Zhang B, et al. HIF2-induced long noncoding RNA RAB11B-AS1 promotes hypoxia-mediated angiogenesis and breast cancer metastasis.

Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers Basel. Wang L, He T, Liu J, Tai J, Wang B, Chen Z, et al. Pan-cancer analysis reveals tumor-associated macrophage communication in the tumor microenvironment.

Exp Hematol Oncol. Tumor angiogenesis: therapeutic implications. McCormack PL, Keam SJ. Sandler A. Bevacizumab in non-small cell lung cancer. Wu H-C, Huang C-T, Chang D-K.

Anti-angiogenic therapeutic drugs for treatment of human cancer. J Cancer Mol. Mukherji S. Bevacizumab avastin. Am J Neuroradiol. Kelly RJ, Rixe O. Axitinib AG Small Mol Oncol. Article Google Scholar. Spano JP, Chodkiewicz C, Maurel J, Wong R, Wasan H, Barone C, et al.

Efficacy of gemcitabine plus axitinib compared with gemcitabine alone in patients with advanced pancreatic cancer: an open-label randomised phase II study. Houghton PJ. Singh S, Carnaghi C, Buzzoni R, Pommier RF, Raderer M, Tomasek J, et al.

Everolimus in neuroendocrine tumors of the gastrointestinal tract and unknown primary. Elisei R, Schlumberger MJ, Müller SP, Schöffski P, Brose MS, Shah MH, et al. Cabozantinib in progressive medullary thyroid cancer. J Clin Oncol. Chanzá NM, Xie W, Bilen MA, Dzimitrowicz H, Burkart J, Geynisman DM, et al.

Cabozantinib in advanced non-clear-cell renal cell carcinoma: a multicentre, retrospective, cohort study. Lancet Oncol. PubMed Central Google Scholar. Armoiry X, Aulagner G, Facon T. Lenalidomide in the treatment of multiple myeloma: a review.

J Clin Pharm Ther. Talati C, Sallman D, List A, editors. Lenalidomide: myelodysplastic syndromes with del 5q and beyond.

Seminars in hematology. Amsterdam: Elsevier; Cabanillas ME, Habra MA. Lenvatinib: role in thyroid cancer and other solid tumors. Cancer Treat Rev. Leonetti A, Leonardi F, Bersanelli M, Buti S. Clinical use of lenvatinib in combination with everolimus for the treatment of advanced renal cell carcinoma.

Ther Clin Risk Manag. Keisner SV, Shah SR. Poole RM, Vaidya A. Ramucirumab: first global approval. Garon EB, Cao D, Alexandris E, John WJ, Yurasov S, Perol M. A randomized, double-blind, phase III study of docetaxel and ramucirumab versus docetaxel and placebo in the treatment of stage IV non—small-cell lung cancer after disease progression after 1 previous platinum-based therapy REVEL : Treatment Rationale and Study Design.

Clin Lung Cancer. Verdaguer H, Tabernero J, Macarulla T. Ramucirumab in metastatic colorectal cancer: evidence to date and place in therapy. Ther Adv Med Oncol. Syed YY. Ramucirumab: a review in hepatocellular carcinoma. Bruix J, Qin S, Merle P, Granito A, Huang Y-H, 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. The Lancet. Abdelgalil AA, Alkahtani HM, Al-Jenoobi FI.

Profiles of drug substances, excipients and related methodology, vol. Le Tourneau C, Raymond E, Faivre S. Sunitinib: a novel tyrosine kinase inhibitor. A brief review of its therapeutic potential in the treatment of renal carcinoma and gastrointestinal stromal tumors GIST.

Palumbo A, Facon T, Sonneveld P, Blade J, Offidani M, Gay F, et al. Thalidomide for treatment of multiple myeloma: 10 years later. Blood J Am Soc Hematol. Patel A, Sun W. Ziv-aflibercept in metastatic colorectal cancer.

Biol Targets Ther. Commander H, Whiteside G, Perry C. Wells SA Jr, Robinson BG, Gagel RF, Dralle H, Fagin JA, Santoro M, et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial. Pàez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Viñals F, et al.

Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. Grepin R, Guyot M, Jacquin M, Durivault J, Chamorey E, Sudaka A, et al. Maione F, Capano S, Regano D, Zentilin L, Giacca M, Casanovas O, et al.

Semaphorin 3A overcomes cancer hypoxia and metastatic dissemination induced by antiangiogenic treatment in mice. J Clin Investig. Singh M, Couto SS, Forrest WF, Lima A, Cheng JH, Molina R, et al.

Anti-VEGF antibody therapy does not promote metastasis in genetically engineered mouse tumour models. J Pathol. Welti JC, Powles T, Foo S, Gourlaouen M, Preece N, Foster J, et al.

Contrasting effects of sunitinib within in vivo models of metastasis. Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway. Haibe Y, Kreidieh M, El Hajj H, Khalifeh I, Mukherji D, Temraz S, et al.

Resistance mechanisms to anti-angiogenic therapies in cancer. Jiang B-H, Agani F, Passaniti A, Semenza GL. V-SRC induces expression of hypoxia-inducible factor 1 HIF-1 and transcription of genes encoding vascular endothelial growth factor and enolase 1: involvement of HIF-1 in tumor progression.

Pereira ER, Frudd K, Awad W, Hendershot LM. Endoplasmic reticulum ER stress and hypoxia response pathways interact to potentiate hypoxia-inducible factor 1 HIF-1 transcriptional activity on targets like vascular endothelial growth factor VEGF. Luo D, Wang Z, Wu J, Jiang C, Wu J.

The role of hypoxia inducible factor-1 in hepatocellular carcinoma. BioMed Res Int. Article PubMed PubMed Central Google Scholar. Kitajima Y, Miyazaki K. The critical impact of HIF-1a on gastric cancer biology.

Maione F, Molla F, Meda C, Latini R, Zentilin L, Giacca M, et al. Semaphorin 3A is an endogenous angiogenesis inhibitor that blocks tumor growth and normalizes tumor vasculature in transgenic mouse models. Fagiani E, Christofori G.

Angiopoietins in angiogenesis. Cancer Lett. Hanahan D. Signaling vascular morphogenesis and maintenance. Eklund L, Saharinen P. Angiopoietin signaling in the vasculature. Exp Cell Res.

New approaches to antiangiogenesis therapy of solid tumors | Nature Genetics Breast Cancer therappy, —e4. CAS PubMed PubMed Central Anti-angiogenesis therapy for solid tumors Scholar Chanzá NM, Tumord W, Bilen Anti-anggiogenesis, Dzimitrowicz H, Burkart Anti-amgiogenesis, Geynisman DM, et al. Pan Mood-lifting exercises et al Blocking neuropilin-1 function has an additive Anti-angiogenesus with anti-VEGF to inhibit Anti-angiogenesis therapy for solid tumors growth. Decades of experimental evidence have shown that solid tumor growth is dependent on angiogenic formation of new blood vessels [ 3 ]. Kilarski WW et al Biomechanical regulation of blood vessel growth during tissue vascularization. Upon successful preclinical studies Table 1a myriad of clinical trials have been accomplished or are ongoing to determine the safety, feasibility, and efficacy of anti-angiogenic agents therapy in cancer patients alone or in combination with other therapeutic means Table 2. Proc Natl Acad Sci USA 43 —

Video

Tumour immunology and immunotherapy

Author: Bashakar

3 thoughts on “Anti-angiogenesis therapy for solid tumors

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com