Preclinical characterization of anlotinib, a highly potent and selective vascular endothelial growth factor receptor-2 inhibitor

Chengying Xie, Xiaozhe Wan, Haitian Quan, Mingyue Zheng, Li Fu, Yun Li, Liguang Lou, Chengying Xie, Xiaozhe Wan, Haitian Quan, Mingyue Zheng, Li Fu, Yun Li, Liguang Lou

Abstract

Abrogating tumor angiogenesis by inhibiting vascular endothelial growth factor receptor-2 (VEGFR2) has been established as a therapeutic strategy for treating cancer. However, because of their low selectivity, most small molecule inhibitors of VEGFR2 tyrosine kinase show unexpected adverse effects and limited anticancer efficacy. In the present study, we detailed the pharmacological properties of anlotinib, a highly potent and selective VEGFR2 inhibitor, in preclinical models. Anlotinib occupied the ATP-binding pocket of VEGFR2 tyrosine kinase and showed high selectivity and inhibitory potency (IC50 <1 nmol/L) for VEGFR2 relative to other tyrosine kinases. Concordant with this activity, anlotinib inhibited VEGF-induced signaling and cell proliferation in HUVEC with picomolar IC50 values. However, micromolar concentrations of anlotinib were required to inhibit tumor cell proliferation directly in vitro. Anlotinib significantly inhibited HUVEC migration and tube formation; it also inhibited microvessel growth from explants of rat aorta in vitro and decreased vascular density in tumor tissue in vivo. Compared with the well-known tyrosine kinase inhibitor sunitinib, once-daily oral dose of anlotinib showed broader and stronger in vivo antitumor efficacy and, in some models, caused tumor regression in nude mice. Collectively, these results indicate that anlotinib is a well-tolerated, orally active VEGFR2 inhibitor that targets angiogenesis in tumor growth, and support ongoing clinical evaluation of anlotinib for a variety of malignancies.

Keywords: VEGF; VEGFR2; angiogenesis; anlotinib; tyrosine kinase inhibitor.

© 2018 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.

Figures

Figure 1
Figure 1
Characterization of anlotinib as a vascular endothelial growth factor receptor‐2 (VEGFR2) inhibitor. A, Chemical structure of anlotinib. B, Molecular modeling of the VEGFR2–anlotinib/sunitinib complex. Hydrogen bonds are presented as yellow dashed lines, and critical residues are presented as maroon sticks. Anlotinib (cyan, docked pose with structure PDB code 4ASD) and sunitinib (orange, from crystallographic structure PDB code 4AGD). C, Molecular modeling of the c‐Kit–anlotinib complex (orange) and VEGFR2–anlotinib complex (cyan). Hydrogen bonds are presented as yellow dashed lines, and critical residues are presented as maroon sticks (VEGFR2) and dark green sticks (c‐Kit)
Figure 2
Figure 2
Effects of anlotinib on growth factor‐stimulated receptor phosphorylation. Serum‐starved (A) HUVEC, (B) Mo7e, (C) U‐87MG and (D) A431 cells were treated with different concentrations of test agents for 1.5 h and then stimulated with vascular endothelial growth factor (VEGF; 20 ng/mL), stem cell factor‐1 (SCF‐1; 2.5 ng/mL), platelet‐derived growth factor‐BB (PDGF‐BB; 10 ng/mL), or epidermal growth factor (EGF; 10 ng/mL) for 10 min, respectively. E, BT‐474 cells which have constitutive HER2 autophosphorylation and downstream signaling activation were treated with test agents for 1.5 h. Cell lysates were probed with the indicated antibodies
Figure 3
Figure 3
Inhibitory effects of anlotinib on cell proliferation. A, Inhibitory effect of anlotinib on vascular endothelial growth factor (VEGF)‐ or FBS‐stimulated HUVEC proliferation. HUVEC were incubated with different concentrations of drugs together with FBS (20%) or VEGF (20 ng/mL). B, Effects of anlotinib on tumor cell proliferation. Tumor cells were cultured with 10% FBS and then treated with anlotinib. Cell viability was determined by sulforhodamine B (SRB) assay. IC 50 values are presented as means ± SD of 3 independent experiments
Figure 4
Figure 4
Inhibitory effects of anlotinib on angiogenesis in vitro. A, Effect of anlotinib on HUVEC migration induced by vascular endothelial growth factor (VEGF)‐A. B, Effect of anlotinib on FBS‐stimulated HUVEC tube formation. C, Effect of anlotinib on VEGF‐stimulated microvessel sprouting from rat aortic rings. Representative images are shown and data are presented as the mean ± SD of 3 independent experiments. *P < .05, **P < .01 compared with VEGF‐ or FBS‐treated groups
Figure 5
Figure 5
In vivo antitumor efficacy of anlotinib in SW620 tumor xenografts. (A,B) SW620 tumor‐bearing mice were orally given vehicle (n = 12) or the indicated doses of anlotinib or sunitinib (n = 6) daily for 18 d. A, Tumor volumes and (B) mouse bodyweights were determined twice weekly during the course of the experiment. C, Photographs of tumors on the final day. D, Immunohistochemical detection of the endothelial cell‐specific marker, CD31, in tumor tissue sections of SW620 xenografts. Data are presented as means ± SEM. *P < .05, **P < .01 vs vehicle
Figure 6
Figure 6
In vivo antitumor activity of anlotinib against a panel of tumor xenografts. A, U‐87MG, (B) Caki‐1, (C) SK‐OV‐3, and (D) Calu‐3 tumor‐bearing mice were orally given vehicle (n = 12) or the indicated doses of anlotinib or sunitinib (n = 6) daily. In SK‐OV‐3 and Calu‐3 xenograft models, anlotinib at 6 mg/kg was orally given daily for only 9 d. Data are presented as means ± SEM. **P < .05, **P < .01 vs vehicle

References

    1. Bussolino F, Mantovani A, Persico G. Molecular mechanisms of blood vessel formation. Trends Biochem Sci. 1997;22:251‐256.
    1. Harper J, Moses MA. Molecular regulation of tumor angiogenesis: mechanisms and therapeutic implications In: Bignold LP, ed. Cancer: Cell Structures, Carcinogens and Genomic Instability. Experientia Supplementum, vol. 96, Basel, Switzerland: Birkhäuser; 2006:223‐268.
    1. Carmeliet P. Angiogenesis in health and disease. Nat Med. 2003;9:653‐660.
    1. Folkman J. Seminars in Medicine of the Beth Israel Hospital, Boston. Clinical applications of research on angiogenesis. N Engl J Med. 1995;333:1757‐1763.
    1. Folkman J. Angiogenesis. Annu Rev Med. 2006;57:1‐18.
    1. Boehm T, Folkman J, Browder T, O'Reilly MS. Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance. Nature. 1997;390:404‐407.
    1. Al‐Husein B, Abdalla M, Trepte M, Deremer DL, Somanath PR. Antiangiogenic therapy for cancer: an update. Pharmacotherapy. 2012;32:1095‐1111.
    1. Kerbel RS. A decade of experience in developing preclinical models of advanced‐ or early‐stage spontaneous metastasis to study antiangiogenic drugs, metronomic chemotherapy, and the tumor microenvironment. Cancer J. 2015;21:274‐283.
    1. Wang Z, Dabrosin C, Yin X, et al. Broad targeting of angiogenesis for cancer prevention and therapy. Semin Cancer Biol. 2015;35(suppl):S224‐S243.
    1. Shweiki D, Itin A, Soffer D, Keshet E. Vascular endothelial growth factor induced by hypoxia may mediate hypoxia‐initiated angiogenesis. Nature. 1992;359:843‐845.
    1. Kim KJ, Li B, Winer J, et al. Inhibition of vascular endothelial growth factor‐induced angiogenesis suppresses tumour growth in vivo. Nature. 1993;362:841‐844.
    1. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9:669‐676.
    1. Shibuya M. Vascular endothelial growth factor and its receptor system: physiological functions in angiogenesis and pathological roles in various diseases. J Biochem. 2013;153:13‐19.
    1. Takahashi S. Vascular endothelial growth factor (VEGF), VEGF receptors and their inhibitors for antiangiogenic tumor therapy. Biol Pharm Bull. 2011;34:1785‐1788.
    1. Keating GM. Bevacizumab: a review of its use in advanced cancer. Drugs. 2014;74:1891‐1925.
    1. Wilke H, Muro K, Van Cutsem E, et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro‐oesophageal junction adenocarcinoma (RAINBOW): a double‐blind, randomised phase 3 trial. Lancet Oncol. 2014;15:1224‐1235.
    1. Cooper MR, Binkowski C, Hartung J, Towle J. Profile of ramucirumab in the treatment of metastatic non‐small‐cell lung cancer. Onco Targets Ther. 2016;9:1953‐1960.
    1. Fontanella C, Ongaro E, Bolzonello S, Guardascione M, Fasola G, Aprile G. Clinical advances in the development of novel VEGFR2 inhibitors. Ann Transl Med. 2014;2:123.
    1. Elice F, Rodeghiero F. Bleeding complications of antiangiogenic therapy: pathogenetic mechanisms and clinical impact. Thromb Res. 2010;125(suppl 2):S55‐S57.
    1. Sun Y, Niu W, Du F, et al. Safety, pharmacokinetics, and antitumor properties of anlotinib, an oral multi‐target tyrosine kinase inhibitor, in patients with advanced refractory solid tumors. J Hematol Oncol. 2016;9:105.
    1. Jaffe EA, Nachman RL, Becker CG, Minick CR. Culture of human endothelial cells derived from umbilical veins. Identification by morphologic and immunologic criteria. J Clin Invest. 1973;52:2745‐2756.
    1. Lazaro I, Gonzalez M, Roy G, Villar LM, Gonzalez‐Porque P. Description of an enzyme‐linked immunosorbent assay for the detection of protein tyrosine kinase. Anal Biochem. 1991;192:257‐261.
    1. Maestro Schrödinger. New York, NY: LLC; 2015.
    1. LigPrep Schrödinger. New York, NY: LLC; 2015.
    1. Glide Schrödinger. New York, NY: LLC; 2015.
    1. Shelley JC, Cholleti A, Frye LL, Greenwood JR, Timlin MR, Uchimaya M. Epik: a software program for pK a prediction and protonation state generation for drug‐like molecules. J Comput Aided Mol Des. 2007;21:681‐691.
    1. Schrodinger LLC. The PyMOL Molecular Graphics System, Version 1.8. 2015.
    1. Min JK, Han KY, Kim EC, et al. Capsaicin inhibits in vitro and in vivo angiogenesis. Cancer Res. 2004;64:644‐651.
    1. Lee CC, Liu KJ, Wu YC, Lin SJ, Chang CC, Huang TS. Sesamin inhibits macrophage‐induced vascular endothelial growth factor and matrix metalloproteinase‐9 expression and proangiogenic activity in breast cancer cells. Inflammation. 2011;34:209‐221.
    1. Xie CY, Xu YP, Jin W, Lou LG. Antitumor activity of lobaplatin alone or in combination with antitubulin agents in non‐small‐cell lung cancer. Anticancer Drugs. 2012;23:698‐705.
    1. Li J, Zhou N, Luo K, et al. In silico discovery of potential VEGFR‐2 inhibitors from natural derivatives for anti‐angiogenesis therapy. Int J Mol Sci. 2014;15:15994‐16011.
    1. Sanphanya K, Wattanapitayakul SK, Phowichit S, Fokin VV, Vajragupta O. Novel VEGFR‐2 kinase inhibitors identified by the back‐to‐front approach. Bioorg Med Chem Lett. 2013;23:2962‐2967.
    1. Yang TH, Lee CI, Huang WH, Lee AR. Synthesis and evaluation of Novel 2‐pyrrolidone‐fused (2‐oxoindolin‐3‐ylidene)methylpyrrole derivatives as potential multi‐target tyrosine kinase receptor inhibitors. Molecules. 2017;22:913‐932.
    1. Tian S, Quan H, Xie C, et al. YN968D1 is a novel and selective inhibitor of vascular endothelial growth factor receptor‐2 tyrosine kinase with potent activity in vitro and in vivo. Cancer Sci. 2011;102:1374‐1380.
    1. Wedge SR, Kendrew J, Hennequin LF, et al. AZD2171: a highly potent, orally bioavailable, vascular endothelial growth factor receptor‐2 tyrosine kinase inhibitor for the treatment of cancer. Cancer Res. 2005;65:4389‐4400.
    1. Amino N, Ideyama Y, Yamano M, et al. YM‐359445, an orally bioavailable vascular endothelial growth factor receptor‐2 tyrosine kinase inhibitor, has highly potent antitumor activity against established tumors. Clin Cancer Res. 2006;12:1630‐1638.
    1. Shahneh FZ, Baradaran B, Zamani F, Aghebati‐Maleki L. Tumor angiogenesis and anti‐angiogenic therapies. Hum Antibodies. 2013;22:15‐19.
    1. Petrova TV, Makinen T, Alitalo K. Signaling via vascular endothelial growth factor receptors. Exp Cell Res. 1999;253:117‐130.
    1. Ilan N, Mahooti S, Madri JA. Distinct signal transduction pathways are utilized during the tube formation and survival phases of in vitro angiogenesis. J Cell Sci. 1998;111:3621‐3631.
    1. Mendel DB, Laird AD, Xin X, et al. In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet‐derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res. 2003;9:327‐337.
    1. Heinrich EL, Walser TC, Krysan K, et al. The inflammatory tumor microenvironment, epithelial mesenchymal transition and lung carcinogenesis. Cancer Microenviron. 2012;5:5‐18.
    1. Kerbel RS. Antiangiogenic therapy: a universal chemosensitization strategy for cancer? Science. 2006;312:1171‐1175.
    1. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science. 2005;307:58‐62.
    1. De Falco S. Antiangiogenesis therapy: an update after the first decade. Korean J Intern Med. 2014;29:1‐11.
    1. Karkkainen MJ, Haiko P, Sainio K, et al. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol. 2004;5:74‐80.
    1. Arinaga M, Noguchi T, Takeno S, Chujo M, Miura T, Uchida Y. Clinical significance of vascular endothelial growth factor C and vascular endothelial growth factor receptor 3 in patients with nonsmall cell lung carcinoma. Cancer. 2003;97:457‐464.
    1. Onogawa S, Kitadai Y, Tanaka S, Kuwai T, Kuroda T, Chayama K. Regulation of vascular endothelial growth factor (VEGF)‐C and VEGF‐D expression by the organ microenvironment in human colon carcinoma. Eur J Cancer. 2004;40:1604‐1609.
    1. Onogawa S, Kitadai Y, Tanaka S, Kuwai T, Kimura S, Chayama K. Expression of VEGF‐C and VEGF‐D at the invasive edge correlates with lymph node metastasis and prognosis of patients with colorectal carcinoma. Cancer Sci. 2004;95:32‐39.
    1. Xu Y, Feng L, Wang S, et al. Calycosin protects HUVECs from advanced glycation end products‐induced macrophage infiltration. J Ethnopharmacol. 2011;137:359‐370.
    1. Han JY, Lee KH, Kim SW, et al. A phase II study of poziotinib in patients with epidermal growth factor receptor (EGFR)‐mutant lung adenocarcinoma who have acquired resistance to EGFR‐tyrosine kinase inhibitors. Cancer Res Treat. 2017;49:10‐19.
    1. Casaluce F, Sgambato A, Sacco PC, et al. Resistance to crizotinib in advanced non‐small cell lung cancer (NSCLC) with ALK rearrangement: mechanisms, treatment strategies and new targeted therapies. Curr Clin Pharmacol. 2016;11:77‐87.
    1. Han B, Li K, Wang Q, et al. Efficacy and safety of third‐line treatment with anlotinib in patients with refractory advanced non‐small‐cell lung cancer (ALTER‐0303): a randomised, double‐blind, placebo‐controlled phase 3 study. Lancet Oncol. 2017;18:S3.
    1. Wood JM, Bold G, Buchdunger E, et al. PTK787/ZK 222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, impairs vascular endothelial growth factor‐induced responses and tumor growth after oral administration. Cancer Res. 2000;60:2178‐2189.
    1. Beebe JS, Jani JP, Knauth E, et al. Pharmacological characterization of CP‐547,632, a novel vascular endothelial growth factor receptor‐2 tyrosine kinase inhibitor for cancer therapy. Cancer Res. 2003;63:7301‐7309.
    1. Mizumoto A, Yamamoto K, Nakayama Y, et al. Induction of epithelial‐mesenchymal transition via activation of epidermal growth factor receptor contributes to sunitinib resistance in human renal cell carcinoma cell lines. J Pharmacol Exp Ther. 2015;355:152‐158.
    1. Marijon H, Dokmak S, Paradis V, et al. Epithelial‐to‐mesenchymal transition and acquired resistance to sunitinib in a patient with hepatocellular carcinoma. J Hepatol. 2011;54:1073‐1078.
    1. Martinelli E, Troiani T, Morgillo F, et al. Synergistic antitumor activity of sorafenib in combination with epidermal growth factor receptor inhibitors in colorectal and lung cancer cells. Clin Cancer Res. 2010;16:4990‐5001.
    1. Li Y, Yang X, Su LJ, Flaig TW. VEGFR and EGFR inhibition increases epithelial cellular characteristics and chemotherapy sensitivity in mesenchymal bladder cancer cells. Oncol Rep. 2010;24:1019‐1028.
    1. Sosman JA, Puzanov I, Atkins MB. Opportunities and obstacles to combination targeted therapy in renal cell cancer. Clin Cancer Res. 2007;13:764s‐769s.

Source: PubMed

3
Suscribir