REFERENCES
2. Guerra A, Belinha J, Mangir N, MacNeil S, Natal Jorge R. Simulation of the process of angiogenesis: quantification and assessment of vascular patterning in the chicken chorioallantoic membrane. Comput Biol Med 2021;136:104647.
3. Naito H, Iba T, Takakura N. Mechanisms of new blood-vessel formation and proliferative heterogeneity of endothelial cells. Int Immunol 2020;32:295-305.
5. Folkman J, Long DM Jr, Becker FF. Growth and metastasis of tumor in organ culture. Cancer 1963;16:453-67.
6. de Heer EC, Jalving M, Harris AL. HIFs, angiogenesis, and metabolism: elusive enemies in breast cancer. J Clin Invest 2020;130:5074-87.
7. Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci 2020;77:1745-70.
9. Melincovici CS, Boşca AB, Şuşman S, et al. Vascular endothelial growth factor (VEGF)-key factor in normal and pathological angiogenesis. Rom J Morphol Embryol 2018;59:455-67.
10. Bai Y, Bai L, Zhou J, Chen H, Zhang L. Sequential delivery of VEGF, FGF-2 and PDGF from the polymeric system enhance HUVECs angiogenesis in vitro and CAM angiogenesis. Cell Immunol 2018;323:19-32.
11. Armani G, Pozzi E, Pagani A, et al. The heterogeneity of cancer endothelium: the relevance of angiogenesis and endothelial progenitor cells in cancer microenvironment. Microvasc Res 2021;138:104189.
12. Huang XL, Khan MI, Wang J, et al. Role of receptor tyrosine kinases mediated signal transduction pathways in tumor growth and angiogenesis-New insight and futuristic vision. Int J Biol Macromol 2021;180:739-52.
13. Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 1989;246:1306-9.
14. Montesano R, Vassalli JD, Baird A, Guillemin R, Orci L. Basic fibroblast growth factor induces angiogenesis in vitro. Proc Natl Acad Sci U S A 1986;83:7297-301.
15. Fett JW, Strydom DJ, Lobb RR, et al. Isolation and characterization of angiogenin, an angiogenic protein from human carcinoma cells. Biochemistry 1985;24:5480-6.
16. Schreiber AB, Winkler ME, Derynck R. Transforming growth factor-alpha: a more potent angiogenic mediator than epidermal growth factor. Science 1986;232:1250-3.
17. Roberts AB, Sporn MB, Assoian RK, et al. Transforming growth factor type beta: rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro. Proc Natl Acad Sci U S A 1986;83:4167-71.
18. Fràter-Schröder M, Risau W, Hallmann R, Gautschi P, Böhlen P. Tumor necrosis factor type alpha, a potent inhibitor of endothelial cell growth in vitro, is angiogenic in vivo. Proc Natl Acad Sci U S A 1987;84:5277-81.
19. Ishikawa F, Miyazono K, Hellman U, et al. Identification of angiogenic activity and the cloning and expression of platelet-derived endothelial cell growth factor. Nature 1989;338:557-62.
20. Bussolino F, Ziche M, Wang JM, et al. In vitro and in vivo activation of endothelial cells by colony-stimulating factors. J Clin Invest 1991;87:986-95.
21. Zhao J, Chen L, Shu B, et al. Granulocyte/macrophage colony-stimulating factor influences angiogenesis by regulating the coordinated expression of VEGF and the Ang/Tie system. PLoS One 2014;9:e92691.
22. Zheng Q, Li X, Cheng X, et al. Granulocyte-macrophage colony-stimulating factor increases tumor growth and angiogenesis directly by promoting endothelial cell function and indirectly by enhancing the mobilization and recruitment of proangiogenic granulocytes. Tumour Biol 2017;39:1010428317692232.
23. Zhou Y, Tu C, Zhao Y, Liu H, Zhang S. Placental growth factor enhances angiogenesis in human intestinal microvascular endothelial cells via PI3K/Akt pathway: potential implications of inflammation bowel disease. Biochem Biophys Res Commun 2016;470:967-74.
24. Kitadai Y, Takahashi Y, Haruma K, et al. Transfection of interleukin-8 increases angiogenesis and tumorigenesis of human gastric carcinoma cells in nude mice. Br J Cancer 1999;81:647-53.
25. Kaga T, Kawano H, Sakaguchi M, Nakazawa T, Taniyama Y, Morishita R. Hepatocyte growth factor stimulated angiogenesis without inflammation: differential actions between hepatocyte growth factor, vascular endothelial growth factor and basic fibroblast growth factor. Vascul Pharmacol 2012;57:3-9.
26. Gospodarowicz D, Bialecki H, Thakral T. The angiogenic activity of the fibroblast and epidermal growth factor. Experimental Eye Research 1979;28:501-14.
27. Yokoi A, Mccrudden KW, Huang J, et al. Human epidermal growth factor receptor signaling contributes to tumor growth via angiogenesis in her2/neu-expressing experimental Wilms’ tumor. Journal of Pediatric Surgery 2003;38:1569-73.
28. Palinski W, Monti M, Camerlingo R, et al. Lysosome purinergic receptor P2X4 regulates neoangiogenesis induced by microvesicles from sarcoma patients. Cell Death Dis 2021;12:797.
29. Pavlakovic H, Havers W, Schweigerer L. Multiple angiogenesis stimulators in a single malignancy: implications for anti-angiogenic tumour therapy. Angiogenesis 2001;4:259-62.
30. 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 2011;2:1097-105.
31. Muller YA, Li B, Christinger HW, Wells JA, Cunningham BC, de Vos AM. Vascular endothelial growth factor: crystal structure and functional mapping of the kinase domain receptor binding site. Proc Natl Acad Sci U S A 1997;94:7192-7.
32. Shibuya M. Role of VEGF-flt receptor system in normal and tumor angiogenesis. Adv Cancer Res 1995;67:281-316.
33. Li L, Liu H, Xu C, et al. VEGF promotes endothelial progenitor cell differentiation and vascular repair through connexin 43. Stem Cell Res Ther 2017;8:237.
34. Zhu D, Li Y, Zhang Z, et al. Recent advances of nanotechnology-based tumor vessel-targeting strategies. J Nanobiotechnology 2021;19:435.
35. El-Kenawi AE, El-Remessy AB. Angiogenesis inhibitors in cancer therapy: mechanistic perspective on classification and treatment rationales. Br J Pharmacol 2013;170:712-29.
36. Mundel TM, Kalluri R. Type IV collagen-derived angiogenesis inhibitors. Microvasc Res 2007;74:85-9.
37. Abdollahi A, Hahnfeldt P, Maercker C, et al. Endostatin’s antiangiogenic signaling network. Molecular Cell 2004;13:649-63.
38. Ranjit PM, Anuradha C, Vishnupriya S, Girijasankar G, Girish K, Chowdary YA. Endogenous angiogenesis inhibitor endostatin: an overview. Asian Journal of Pharmaceutical and Clinical Research 2012;5:1-8. Available from: https://xueshu.baidu.com/usercenter/paper/show?paperid=cdfa6789882c0b11995e7c9aed8209c7&site=xueshu_se&hitarticle=1. [Last accessed on 26 Apr 2022].
39. Kerbel R, Folkman J. Clinical translation of angiogenesis inhibitors. Nat Rev Cancer 2002;2:727-39.
41. Rao N, Lee YF, Ge R. Novel endogenous angiogenesis inhibitors and their therapeutic potential. Acta Pharmacol Sin 2015;36:1177-90.
43. Frezzetti D, Gallo M, Maiello MR, et al. VEGF as a potential target in lung cancer. Expert Opin Ther Targets 2017;21:959-66.
44. Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011;473:298-307.
45. Xu G. Angiogenesis inhibition in the in vivo antineoplastic effect of manumycin and paclitaxel against anaplastic thyroid carcinoma. Journal of Clinical Endocrinology & Metabolism 2001;86:1769-77.
46. Xu N, Bai Y, Luo T, Duan P. Low-dose paclitaxel combined with thalidomide inhibits angiogenesis in mice bearing S180 sarcoma. Chinese Journal of Clinical Oncology 2020;47:7-11. (in Chinese) Available from: https://xueshu.baidu.com/usercenter/paper/show?paperid=1j620ed0bf510m10dv0a08y0km339097&site=xueshu_se&hitarticle=1. [Last accessed on 26 Apr 2022].
47. Zhang QY, Sun L, Wang ZH. Effect of ginsenoside Rg3 combined with cyclophosphamide on angiogenesis in rats with EMT-6 breast cancer.Chinese Journal of Clinical Rehabilitation 2004. Available from: https://xueshu.baidu.com/usercenter/paper/show?paperid=ac39f44b384ddaf8af60bee674f81bbe&site=xueshu_se&hitarticle=1. [Last accessed on 26 Apr 2022].
48. Ezoe K, Murata N, Yabuuchi A, et al. Long-term adverse effects of cyclophosphamide on follicular growth and angiogenesis in mouse ovaries. Reprod Biol 2014;14:238-42.
49. Ciardiello F, Caputo R, Bianco R, et al. Inhibition of growth factor production and angiogenesis in human cancer cells by ZD1839 (Iressa), a selective epidermal growth factor receptor tyrosine kinase inhibitor. Clin Cancer Res 2001;7:1459-65.
50. Hirata A, Ogawa S, Kometani T, et al. ZD1839 (Iressa) induces antiangiogenic effects through inhibition of epidermal growth factor receptor tyrosine kinase. Cancer Res 2002;62:2554-60.
51. Fallahi P, Ferrari SM, Galdiero MR, et al. Molecular targets of tyrosine kinase inhibitors in thyroid cancer. Semin Cancer Biol 2022;79:180-96.
52. Papadimitriou M, Papadimitriou CA. Antiangiogenic tyrosine kinase inhibitors in metastatic colorectal cancer: focusing on regorafenib. Anticancer Res 2021;41:567-82.
53. Dan H, Lei X, Huang X, Ma N, Xing Y, Shen Y. CM082, a novel VEGF receptor tyrosine kinase inhibitor, can inhibit angiogenesis in vitro and in vivo. Microvasc Res 2021;136:104146.
54. Dai MD, Wang YL, Fan J, et al. DW14383 is an irreversible pan-FGFR inhibitor that suppresses FGFR-dependent tumor growth in vitro and in vivo. Acta Pharmacol Sin 2021;42:1498-506.
55. Yang JC, Haworth L, Sherry RM, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med 2003;349:427-34.
56. Willett CG, Boucher Y, di Tomaso E, et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med 2004;10:145-7.
57. Heinemann V, Hoff PM. Bevacizumab plus irinotecan-based regimens in the treatment of metastatic colorectal cancer. Oncology 2010;79:118-28.
58. Cui F, Chen JZ, Wan C, Chen B, Luo RC, Zheng H. Clinical research of bevacizumab in combination with irinotecan, fluorouracil and leucovorin for advanced metastatic colorectal cancer. Zhonghua Wei Chang Wai Ke Za Zhi 2009;12:374-7.
59. Kabbinavar F, Hurwitz HI, Fehrenbacher L, et al. Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastatic colorectal cancer. J Clin Oncol 2003;21:60-5.
60. Dank M, Budi L, Piko B, et al. First-line bevacizumab-paclitaxel in 220 patients with metastatic breast cancer: results from the AVAREG study. Anticancer Res 2014;34:1275-80.
61. Johnson DH, Fehrenbacher L, Novotny WF, et al. Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreated locally advanced or metastatic non-small-cell lung cancer. J Clin Oncol 2004;22:2184-91.
62. Soria JC, Mauguen A, Reck M, et al. Meta-analysis of Bevacizumab in Advanced NSCLC Collaborative Group. Systematic review and meta-analysis of randomised, phase II/III trials adding bevacizumab to platinum-based chemotherapy as first-line treatment in patients with advanced non-small-cell lung cancer. Ann Oncol 2013;24:20-30.
63. Yu SR, Shi MQ, Xia GH. Combination of bevacizumab and platinum-based chemotherapy as first-line therapy for advanced non-squamous NSCLC. Journal of Practical Oncology 2015;30:418-22.
64. Spratlin JL, Mulder KE, Mackey JR. Ramucirumab (IMC-1121B): a novel attack on angiogenesis. Future Oncol 2010;6:1085-94.
65. Mcintyre JA, Martin L. Panitumumab: oncolytic prop inn anti-EGFR human monoclonal antibody. Drugs of the Future 2004. Available from: https://xueshu.baidu.com/usercenter/paper/show?paperid=af47c87a085e47a6dd730f339a94aa3e&site=xueshu_se&hitarticle=1. [Last accessed on 26 Apr 2022].
66. Pueyo G, Mesia R, Figueras A, et al. Cetuximab may inhibit tumor growth and angiogenesis induced by ionizing radiation: a preclinical rationale for maintenance treatment after radiotherapy. Oncologist 2010;15:976-86.
67. Bugyik E, Dezso K, Reiniger L, et al. Lack of angiogenesis in experimental brain metastases. J Neuropathol Exp Neurol 2011;70:979-91.
68. Valiente M, Obenauf AC, Jin X, et al. Serpins promote cancer cell survival and vascular co-option in brain metastasis. Cell 2014;156:1002-16.
69. Bridgeman VL, Vermeulen PB, Foo S, et al. Vessel co-option is common in human lung metastases and mediates resistance to anti-angiogenic therapy in preclinical lung metastasis models. J Pathol 2017;241:362-74.
70. Stessels F, Van den Eynden G, Van der Auwera I, et al. Breast adenocarcinoma liver metastases, in contrast to colorectal cancer liver metastases, display a non-angiogenic growth pattern that preserves the stroma and lacks hypoxia. Br J Cancer 2004;90:1429-36.
71. Vermeulen PB, Colpaert C, Salgado R, et al. Liver metastases from colorectal adenocarcinomas grow in three patterns with different angiogenesis and desmoplasia. J Pathol 2001;195:336-42.
72. Winkler F. Hostile takeover: how tumours hijack pre-existing vascular environments to thrive. J Pathol 2017;242:267-72.
73. Kuczynski EA, Yin M, Bar-Zion A, et al. Co-option of liver vessels and not sprouting angiogenesis drives acquired sorafenib resistance in hepatocellular carcinoma. J Natl Cancer Inst 2016;108:djw030.
74. Leenders WP, Küsters B, Verrijp K, et al. Antiangiogenic therapy of cerebral melanoma metastases results in sustained tumor progression via vessel co-option. Clin Cancer Res 2004;10:6222-30.
75. Frentzas S, Simoneau E, Bridgeman VL, et al. Vessel co-option mediates resistance to anti-angiogenic therapy in liver metastases. Nat Med 2016;22:1294-302.
76. Kuczynski EA, Reynolds AR. Vessel co-option and resistance to anti-angiogenic therapy. Angiogenesis 2020;23:55-74.
77. Casanovas O, Hicklin DJ, Bergers G, Hanahan D. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 2005;8:299-309.
78. Ebos JM, Lee CR, Christensen JG, Mutsaers AJ, Kerbel RS. Multiple circulating proangiogenic factors induced by sunitinib malate are tumor-independent and correlate with antitumor efficacy. Proc Natl Acad Sci U S A 2007;104:17069-74.
79. Croci DO, Cerliani JP, Dalotto-Moreno T, et al. Glycosylation-dependent lectin-receptor interactions preserve angiogenesis in anti-VEGF refractory tumors. Cell 2014;156:744-58.
80. Pisarsky L, Bill R, Fagiani E, et al. Targeting metabolic symbiosis to overcome resistance to anti-angiogenic therapy. Cell Rep 2016;15:1161-74.
81. Zhai B, Hu F, Jiang X, et al. Inhibition of Akt reverses the acquired resistance to sorafenib by switching protective autophagy to autophagic cell death in hepatocellular carcinoma. Mol Cancer Ther 2014;13:1589-98.
82. Chandra A, Rick J, Yagnik G, Aghi MK. Autophagy as a mechanism for anti-angiogenic therapy resistance. Semin Cancer Biol 2020;66:75-88.
83. Giuliano S, Cormerais Y, Dufies M, et al. Resistance to sunitinib in renal clear cell carcinoma results from sequestration in lysosomes and inhibition of the autophagic flux. Autophagy 2015;11:1891-904.
84. Wu S, Huang L, Shen R, et al. Drug resistance-related sunitinib sequestration in autophagolysosomes of endothelial cells. Int J Oncol 2020;56:113-22.
85. Wen Y, Chelariu-Raicu A, Umamaheswaran S, et al. Endothelial p130cas confers resistance to anti-angiogenesis therapy. Cell Rep 2022;38:110301.
86. Tang D, Gao J, Wang S, et al. Cancer-associated fibroblasts promote angiogenesis in gastric cancer through galectin-1 expression. Tumour Biol 2016;37:1889-99.
87. Hosaka K, Yang Y, Seki T, et al. Pericyte-fibroblast transition promotes tumor growth and metastasis. Proc Natl Acad Sci U S A 2016;113:E5618-27.
88. Orlidge A, D’Amore PA. Inhibition of capillary endothelial cell growth by pericytes and smooth muscle cells. J Cell Biol 1987;105:1455-62.
89. Bussolati B, Deregibus MC, Camussi G. Characterization of molecular and functional alterations of tumor endothelial cells to design anti-angiogenic strategies. Curr Vasc Pharmacol 2010;8:220-32.
90. Bussolati B, Deambrosis I, Russo S, Deregibus MC, Camussi G. Altered angiogenesis and survival in human tumor-derived endothelial cells. FASEB J 2003;17:1159-61.
91. Hida K, Hida Y, Amin DN, et al. Tumor-associated endothelial cells with cytogenetic abnormalities. Cancer Res 2004;64:8249-55.
92. Maishi N, Annan DA, Kikuchi H, Hida Y, Hida K. Tumor endothelial heterogeneity in cancer progression. Cancers (Basel) 2019;11:1511.
93. Holmgren L, O’Reilly MS, Folkman J. Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med 1995;1:149-53.
94. Conley SJ, Gheordunescu E, Kakarala P, et al. Antiangiogenic agents increase breast cancer stem cells via the generation of tumor hypoxia. Proc Natl Acad Sci U S A 2012;109:2784-9.
95. Cheng L, Huang Z, Zhou W, et al. Glioblastoma stem cells generate vascular pericytes to support vessel function and tumor growth. Cell 2013;153:139-52.
96. Mohammed RA, Green A, El-Shikh S, Paish EC, Ellis IO, Martin SG. Prognostic significance of vascular endothelial cell growth factors -A, -C and -D in breast cancer and their relationship with angio- and lymphangiogenesis. Br J Cancer 2007;96:1092-100.
97. Zarrin B, Zarifi F, Vaseghi G, Javanmard SH. Acquired tumor resistance to antiangiogenic therapy: mechanisms at a glance. J Res Med Sci 2017;22:117.
98. Montemagno C, Pagès G. Resistance to anti-angiogenic therapies: a mechanism depending on the time of exposure to the drugs. Front Cell Dev Biol 2020;8:584.
99. Lopes-Coelho F, Martins F, Pereira SA, Serpa J. Anti-angiogenic therapy: current challenges and future perspectives. Int J Mol Sci 2021;22:3765.
100. Kienast Y, Klein C, Scheuer W, et al. Ang-2-VEGF-A CrossMab, a novel bispecific human IgG1 antibody blocking VEGF-A and Ang-2 functions simultaneously, mediates potent antitumor, antiangiogenic, and antimetastatic efficacy. Clin Cancer Res 2013;19:6730-40.
101. Brown JL, Cao ZA, Pinzon-Ortiz M, et al. A human monoclonal anti-ANG2 antibody leads to broad antitumor activity in combination with VEGF inhibitors and chemotherapy agents in preclinical models. Mol Cancer Ther 2010;9:145-56.
102. Gyanchandani R, Ortega Alves MV, Myers JN, Kim S. A proangiogenic signature is revealed in FGF-mediated bevacizumab-resistant head and neck squamous cell carcinoma. Mol Cancer Res 2013;11:1585-96.
103. Powell MA, Sill MW, Goodfellow PJ, et al. A phase II trial of brivanib in recurrent or persistent endometrial cancer: an NRG Oncology/Gynecologic Oncology Group Study. Gynecol Oncol 2014;135:38-43.
104. Shimizu S, Takehara T, Hikita H, et al. Inhibition of autophagy potentiates the antitumor effect of the multikinase inhibitor sorafenib in hepatocellular carcinoma. Int J Cancer 2012;131:548-57.
105. Motzer RJ, Alyasova A, Ye D, et al. Phase II trial of second-line everolimus in patients with metastatic renal cell carcinoma (RECORD-4). Ann Oncol 2016;27:441-8.
106. Bamias A, Karavasilis V, Gavalas N, et al. The combination of bevacizumab/temsirolimus after first-line anti-VEGF therapy in advanced renal-cell carcinoma: a clinical and biomarker study. Int J Clin Oncol 2019;24:411-9.
107. Pietras K, Hanahan D. A multitargeted, metronomic, and maximum-tolerated dose “chemo-switch” regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cancer. J Clin Oncol 2005;23:939-52.
108. Ramadan WS, Zaher DM, Altaie AM, Talaat IM, Elmoselhi A. Potential therapeutic strategies for lung and breast cancers through understanding the anti-angiogenesis resistance mechanisms. Int J Mol Sci 2020;21:565.
109. Song Y, Fu Y, Xie Q, Zhu B, Wang J, Zhang B. Anti-angiogenic agents in combination with immune checkpoint inhibitors: a promising strategy for cancer treatment. Front Immunol 2020;11:1956.
110. Ogasawara S, Mihara Y, Kondo R, Kusano H, Akiba J, Yano H. Antiproliferative effect of lenvatinib on human liver cancer cell lines in vitro and in vivo. Anticancer Res 2019;39:5973-82.
111. Suyama K, Iwase H. Lenvatinib: a promising molecular targeted agent for multiple cancers. Cancer Control 2018;25:1073274818789361.
112. Kudo M, Finn RS, Qin S, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. The Lancet 2018;391:1163-73.
113. Vogel A, Qin S, Kudo M, et al. Lenvatinib versus sorafenib for first-line treatment of unresectable hepatocellular carcinoma: patient-reported outcomes from a randomised, open-label, non-inferiority, phase 3 trial. The Lancet Gastroenterology & Hepatology 2021;6:649-58.
114. Jin H, Shi Y, Lv Y, et al. EGFR activation limits the response of liver cancer to lenvatinib. Nature 2021;595:730-4.
115. Yano S, Yamaguchi M, Dong RP. EGFR tyrosine kinase inhibitor “gefitinib (Iressa)” for cancer therapy. Nihon Yakurigaku Zasshi 2003;122:491-7.
116. Han B, Li K, Wang Q, et al. Effect of anlotinib as a third-line or further treatment on overall survival of patients with advanced non-small cell lung cancer: the ALTER 0303 phase 3 randomized clinical trial. JAMA Oncol 2018;4:1569-75.
117. Chi Y, Fang Z, Hong X, et al. Safety and efficacy of anlotinib, a multikinase angiogenesis inhibitor, in patients with refractory metastatic soft-tissue sarcoma. Clin Cancer Res 2018;24:5233-8.
118. Sherman SI. Advances in chemotherapy of differentiated epithelial and medullary thyroid cancers. J Clin Endocrinol Metab 2009;94:1493-9.
119. Ruan X, Shi X, Dong Q, et al. Antitumor effects of anlotinib in thyroid cancer. Endocr Relat Cancer 2019;26:153-64.
120. Gao Q, Tang S, Chen H, et al. Intratumoral injection of anlotinib hydrogel enhances antitumor effects and reduces toxicity in mouse model of lung cancer. Drug Deliv 2020;27:1524-34.
121. Goff D, Zhang J, Heckrodt T, et al. Discovery of dual Axl/VEGF-R2 inhibitors as potential anti-angiogenic and anti-metastatic drugs for cancer chemotherapy. Bioorg Med Chem Lett 2017;27:3766-71.
122. Holland SJ, Pan A, Franci C, et al. R428, a selective small molecule inhibitor of Axl kinase, blocks tumor spread and prolongs survival in models of metastatic breast cancer. Cancer Res 2010;70:1544-54.
123. Holland SJ, Powell MJ, Franci C, et al. Multiple roles for the receptor tyrosine kinase axl in tumor formation. Cancer Res 2005;65:9294-303.
125. Yoshiji H, Kuriyama S, Yoshii J, et al. Synergistic effect of basic fibroblast growth factor and vascular endothelial growth factor in murine hepatocellular carcinoma. Hepatology 2002;35:834-42.
126. Harimoto N, Taguchi K, Shirabe K, et al. The significance of fibroblast growth factor receptor 2 expression in differentiation of hepatocellular carcinoma. Oncology 2010;78:361-8.
127. Kano MR, Morishita Y, Iwata C, et al. VEGF-A and FGF-2 synergistically promote neoangiogenesis through enhancement of endogenous PDGF-B-PDGFRbeta signaling. J Cell Sci 2005;118:3759-68.
128. Le TBU, Vu TC, Ho RZW, et al. Bevacizumab augments the antitumor efficacy of infigratinib in hepatocellular carcinoma. Int J Mol Sci 2020;21:9405.
129. Prawira A, Le TBU, Vu TC, Huynh H. Ribociclib enhances infigratinib-induced cancer cell differentiation and delays resistance in FGFR-driven hepatocellular carcinoma. Liver Int 2021;41:608-20.
130. Lu RM, Chiu CY, Liu IJ, Chang YL, Liu YJ, Wu HC. Novel human Ab against vascular endothelial growth factor receptor 2 shows therapeutic potential for leukemia and prostate cancer. Cancer Sci 2019;110:3773-87.
131. Zhang J, Liu C, Shi W, et al. The novel VEGF receptor 2 inhibitor YLL545 inhibits angiogenesis and growth in breast cancer. Oncotarget 2016;7:41067-80.
133. West XZ, Malinin NL, Merkulova AA, et al. Oxidative stress induces angiogenesis by activating TLR2 with novel endogenous ligands. Nature 2010;467:972-6.
134. Wu C, Wang X, Tomko N, et al. 2-(ω-carboxyethyl)pyrrole antibody as a new inhibitor of tumor angiogenesis and growth. Anticancer Agents Med Chem 2017;17:813-20.
135. Li QW, Zhang GL, Hao CX, et al. SANT, a novel Chinese herbal monomer combination, decreasing tumor growth and angiogenesis via modulating autophagy in heparanase overexpressed triple-negative breast cancer. J Ethnopharmacol 2021;266:113430.
136. Nadir Y, Vlodavsky I, Brenner B. Heparanase, tissue factor, and cancer. Semin Thromb Hemost 2008;34:187-94.
137. Chen L, Qiu XS. Relationship between expressions of heparanase and basic fibroblast growth factor and clinicopathology of breast cancer tissues. Chinese Journal of Cancer Prevention and Treatment 2010;17:743-745+748. Available from: https://xueshu.baidu.com/usercenter/paper/show?paperid=22fd6776dcd06aea5a4e3876c132fdf9&site=xueshu_se&hitarticle=1. [Last accessed on 26 Apr 2022].
138. Gawthorpe S, Brown JE, Arif M, Nightingale P, Nevill A, Carmichael AR. Heparanase and COX-2 expression as predictors of lymph node metastasis in large, high-grade breast tumors. Anticancer Res 2014;34:2797-800.
139. Zhang PC, Liu X, Li MM, et al. AT-533, a novel Hsp90 inhibitor, inhibits breast cancer growth and HIF-1α/VEGF/VEGFR-2-mediated angiogenesis in vitro and in vivo. Biochem Pharmacol 2020;172:113771.
140. Miyata Y, Nakamoto H, Neckers L. The therapeutic target Hsp90 and cancer hallmarks. Curr Pharm Des 2013;19:347-65.
141. Calderwood SK, Gong J. Heat shock proteins promote cancer: it’s a protection racket. Trends Biochem Sci 2016;41:311-23.
142. Han B, Li K, Zhao Y, et al. Anlotinib as a third-line therapy in patients with refractory advanced non-small-cell lung cancer: a multicentre, randomised phase II trial (ALTER0302). Br J Cancer 2018;118:654-61.
143. Koumarianou A, Kaltsas G. Surufatinib-a novel oral agent for neuroendocrine tumours. Nat Rev Endocrinol 2021;17:9-10.
144. Xu J, Shen L, Zhou Z, et al. Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET-ep): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol 2020;21:1500-12.
145. Xu J, Shen L, Bai C, et al. Surufatinib in advanced pancreatic neuroendocrine tumours (SANET-p): a randomised, double-blind, placebo-controlled, phase 3 study. The Lancet Oncology 2020;21:1489-99.
147. Taylor MH, Lee CH, Makker V, et al. Phase IB/II trial of lenvatinib plus pembrolizumab in patients with advanced renal cell carcinoma, endometrial cancer, and other selected advanced solid tumors. J Clin Oncol 2020;38:1154-63.
148. Yasuda S, Sho M, Yamato I, et al. Simultaneous blockade of programmed death 1 and vascular endothelial growth factor receptor 2 (VEGFR2) induces synergistic anti-tumour effect in vivo. Clin Exp Immunol 2013;172:500-6.
149. Atkins MB, Plimack ER, Puzanov I, et al. Axitinib in combination with pembrolizumab in patients with advanced renal cell cancer: a non-randomised, open-label, dose-finding, and dose-expansion phase 1b trial. The Lancet Oncology 2018;19:405-15.
150. Powles T, Plimack ER, Soulières D, et al. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): extended follow-up from a randomised, open-label, phase 3 trial. The Lancet Oncology 2020;21:1563-73.
151. Riechelmann RP, Leite LS, Bariani GM, et al. Regorafenib in patients with antiangiogenic-naïve and chemotherapy-refractory advanced colorectal cancer: results from a phase IIb trial. Oncologist 2019;24:1180-7.
152. Ettrich TJ, Seufferlein T. Regorafenib. In: Martens UM, editor. Small molecules in oncology. Cham: Springer International Publishing; 2018. p. 45-56.
153. la Fouchardière C. Regorafenib in the treatment of metastatic colorectal cancer. Future Oncol 2018;14:2239-46.
154. Nakazawa T, Hidaka H, Takada J, et al. Early increase in α-fetoprotein for predicting unfavorable clinical outcomes in patients with advanced hepatocellular carcinoma treated with sorafenib. Eur J Gastroenterol Hepatol 2013;25:683-9.
155. Silva JP, Gorman RA, Berger NG, et al. The prognostic utility of baseline alpha-fetoprotein for hepatocellular carcinoma patients. J Surg Oncol 2017;116:831-40.
156. Tangkijvanich P, Anukulkarnkusol N, Suwangool P, et al. Clinical characteristics and prognosis of hepatocellular carcinoma: analysis based on serum alpha-fetoprotein levels. J Clin Gastroenterol 2000;31:302-8.
157. Zhu AX, Kang Y, Yen C, et al. Ramucirumab after sorafenib in patients with advanced hepatocellular carcinoma and increased α-fetoprotein concentrations (REACH-2): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet Oncology 2019;20:282-96.
158. Mayer RJ, Van Cutsem E, Falcone A, et al. RECOURSE Study Group. Randomized trial of TAS-102 for refractory metastatic colorectal cancer. N Engl J Med 2015;372:1909-19.
159. Tsukihara H, Nakagawa F, Sakamoto K, et al. Efficacy of combination chemotherapy using a novel oral chemotherapeutic agent, TAS-102, together with bevacizumab, cetuximab, or panitumumab on human colorectal cancer xenografts. Oncol Rep 2015;33:2135-42.
160. Kuboki Y, Nishina T, Shinozaki E, et al. TAS-102 plus bevacizumab for patients with metastatic colorectal cancer refractory to standard therapies (C-TASK FORCE): an investigator-initiated, open-label, single-arm, multicentre, phase 1/2 study. The Lancet Oncology 2017;18:1172-81.
161. Palazzo A, Dellapasqua S, Munzone E, et al. Phase II trial of bevacizumab plus weekly paclitaxel, carboplatin, and metronomic cyclophosphamide with or without trastuzumab and endocrine therapy as preoperative treatment of inflammatory breast cancer. Clin Breast Cancer 2018;18:328-35.
162. Lan C, Wang Y, Xiong Y, et al. Apatinib combined with oral etoposide in patients with platinum-resistant or platinum-refractory ovarian cancer (AEROC): a phase 2, single-arm, prospective study. The Lancet Oncology 2018;19:1239-46.
163. Marrero AD, Quesada AR, Martínez-Poveda B, Medina MÁ. Antiangiogenic phytochemicals constituent of diet as promising candidates for chemoprevention of cancer. Antioxidants (Basel) 2022;11:302.
164. Hussen BM, Salihi A, Abdullah ST, et al. Signaling pathways modulated by miRNAs in breast cancer angiogenesis and new therapeutics. Pathol Res Pract 2022;230:153764.