2,310
Views
9
CrossRef citations to date
0
Altmetric
Review

Future applications of FGF/FGFR inhibitors in cancer

, , &
Pages 861-872 | Received 22 Dec 2017, Accepted 19 Jun 2018, Published online: 02 Jul 2018

References

  • Ornitz DM, Itoh N. The fibroblast growth factor signaling pathway. Wiley Interdiscip Rev Dev Biol. 2015;4:215–266.
  • Holzmann K, Grunt T, Heinzle C, et al. Alternative splicing of fibroblast growth factor receptor IgIII loops in cancer. J Nucleic Acids. 2012;2012:1–12.
  • Eswarakumar VP, Lax I, Schlessinger J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev. 2005;16:139–149.
  • Ishiwata T. Role of fibroblast growth factor receptor-2 splicing in normal and cancer cells. Front Biosci (Landmark Ed). 2018;23:626–639.
  • Trueb B. Biology of FGFRL1, the fifth fibroblast growth factor receptor. Cell Mol Life Sci. 2011;68:951–964.
  • Silva PN, Altamentova SM, Kilkenny DM, et al. Fibroblast growth factor receptor like-1 (FGFRL1) interacts with SHP-1 phosphatase at insulin secretory granules and induces beta-cell ERK1/2 protein activation. J Biol Chem. 2013;288:17859–17870.
  • Itoh N, Ornitz DM. Fibroblast growth factors: from molecular evolution to roles in development, metabolism and disease. J Biochem. 2011;149:121–130.
  • Richard C, Liuzzo JP, Moscatelli D. Fibroblast growth factor-2 can mediate cell attachment by linking receptors and heparan sulfate proteoglycans on neighboring cells. J Biol Chem. 1995;270:24188–24196.
  • Wan X, Corn PG, Yang J, et al. Prostate cancer cell-stromal cell crosstalk via FGFR1 mediates antitumor activity of dovitinib in bone metastases. Sci Transl Med. 2014;6:252ra122.
  • Giacomini A, Chiodelli P, Matarazzo S, et al. Blocking the FGF/FGFR system as a “two-compartment” antiangiogenic/antitumor approach in cancer therapy. Pharmacol Res. 2016;107:172–185.
  • Helsten T, Elkin S, Arthur E, et al. The FGFR landscape in cancer: analysis of 4,853 tumors by next-generation sequencing. Clin Cancer Res. 2016;22:259–267.
  • Helsten T, Schwaederle M, Kurzrock R. Fibroblast growth factor receptor signaling in hereditary and neoplastic disease: biologic and clinical implications. Cancer Metastasis Rev. 2015;34:479–496.
  • Babina IS, Turner NC. Advances and challenges in targeting FGFR signalling in cancer. Nat Rev Cancer. 2017;17:318–332.
  • Weiss J, Sos ML, Seidel D, et al. Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci Transl Med. 2010;2:62ra93.
  • Peifer M, Fernández-Cuesta L, Sos ML, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet. 2012;44:1104–1110.
  • Turner N, Pearson A, Sharpe R, et al. FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer. Cancer Res. 2010;70:2085–2094.
  • Fischbach A, Rogler A, Erber R, et al. Fibroblast growth factor receptor (FGFR) gene amplifications are rare events in bladder cancer. Histopathology. 2015;66:639–649.
  • Hart KC, Robertson SC, Kanemitsu MY, et al. Transformation and Stat activation by derivatives of FGFR1, FGFR3, and FGFR4. Oncogene. 2000;19:3309–3320.
  • Lew ED, Furdui CM, Anderson KS, et al. The precise sequence of FGF receptor autophosphorylation is kinetically driven and is disrupted by oncogenic mutations. Sci Signal. 2009;2:ra6.
  • Agelopoulos K, Richter GH, Schmidt E, et al. Deep sequencing in conjunction with expression and functional analyses reveals activation of FGFR1 in Ewing Sarcoma. Clin Cancer Res. 2015;21:4935–4946.
  • Gallo LH, Nelson KN, Meyer AN, et al. Functions of fibroblast growth factor receptors in cancer defined by novel translocations and mutations. Cytokine Growth Factor Rev. 2015;26:425–449.
  • Schram AM, Chang MT, Jonsson P, et al. Fusions in solid tumours: diagnostic strategies, targeted therapy, and acquired resistance. Nat Rev Clin Oncol. 2017;14:735–748.
  • Landberg N, Dreimane A, Rissler M, et al. Primary cells in BCR/FGFR1-positive 8p11 myeloproliferative syndrome are sensitive to dovitinib, ponatinib, and dasatinib. Eur J Haematol. 2017;99:442–448.
  • André F, Cortés J. Rationale for targeting fibroblast growth factor receptor signaling in breast cancer. Breast Cancer Res Treat. 2015;150:1–8.
  • Burstein MD, Tsimelzon A, Poage GM, et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res. 2015;21:1688–1698.
  • Su X, Zhan P, Gavine PR, et al. FGFR2 amplification has prognostic significance in gastric cancer: results from a large international multicentre study. Br J Cancer. 2014;110:967–975.
  • Ahn S, Lee J, Hong M, et al. FGFR2 in gastric cancer: protein overexpression predicts gene amplification and high H-index predicts poor survival. Mod Pathol. 2016;29:1095–1103.
  • Cancer Genome Atlas Research N. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–209.
  • Dutt A, Salvesen HB, Chen T-H, et al. Drug-sensitive FGFR2 mutations in endometrial carcinoma. Proc Natl Acad Sci U S A. 2008;105:8713–8717.
  • Tchaicha JH, Akbay EA, Altabef A, et al. Kinase domain activation of FGFR2 yields high-grade lung adenocarcinoma sensitive to a Pan-FGFR inhibitor in a mouse model of NSCLC. Cancer Res. 2014;74:4676–4684.
  • Tanizaki J, Ercan D, Capelletti M, et al. Identification of oncogenic and drug-sensitizing mutations in the extracellular domain of FGFR2. Cancer Res. 2015;75:3139–3146.
  • Jung E-J, Jung E-J, Min SY, et al. Fibroblast growth factor receptor 2 gene amplification status and its clinicopathologic significance in gastric carcinoma. Hum Pathol. 2012;43:1559–1566.
  • Baldia PH, Maurer A, Heide T, et al. Fibroblast growth factor receptor (FGFR) alterations in squamous differentiated bladder cancer: a putative therapeutic target for a small subgroup. Oncotarget. 2016;7(44):71429–71439.
  • Wu Y-M, Su F, Kalyana-Sundaram S, et al. Identification of targetable FGFR gene fusions in diverse cancers. Cancer Discov. 2013;3:636–647.
  • Arai Y, Totoki Y, Hosoda F, et al. Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma. Hepatology. 2014;59:1427–1434.
  • Sia D, Losic B, Moeini A, et al. Massive parallel sequencing uncovers actionable FGFR2-PPHLN1 fusion and ARAF mutations in intrahepatic cholangiocarcinoma. Nat Commun. 2015;6:6087.
  • Vékony H, Ylstra B, Wilting SM, et al. DNA copy number gains at loci of growth factors and their receptors in salivary gland adenoid cystic carcinoma. Clin Cancer Res. 2007;13:3133–3139.
  • Bernard-Pierrot I, Brams A, Dunois-Lardé C, et al. Oncogenic properties of the mutated forms of fibroblast growth factor receptor 3b. Carcinogenesis. 2006;27:740–747.
  • Liu X, Zhang W, Geng D, et al. Clinical significance of fibroblast growth factor receptor-3 mutations in bladder cancer: a systematic review and meta-analysis. Genet Mol Res. 2014;13:1109–1120.
  • Solomon JP, Hansel DE. The emerging molecular landscape of urothelial carcinoma. Surg Pathol Clin. 2016;9:391–394.
  • Dodurga Y, Tataroglu C, Kesen Z, et al. Incidence of fibroblast growth factor receptor 3 gene (FGFR3) A248C, S249C, G372C, and T375C mutations in bladder cancer. Genet Mol Res. 2011;10:86–95.
  • Rosty C, Aubriot M-H, Cappellen D, et al. Clinical and biological characteristics of cervical neoplasias with FGFR3 mutation. Mol Cancer. 2005;4:15.
  • Chesi M, Nardini E, Brents LA, et al. Frequent translocation t(4;14)(p16.3;q32.3) in multiple myeloma is associated with increased expression and activating mutations of fibroblast growth factor receptor 3. Nat Genet. 1997;16:260–264.
  • Soverini S, Terragna C, Testoni N, et al. Novel mutation and RNA splice variant of fibroblast growth factor receptor 3 in multiple myeloma patients at diagnosis. Haematologica. 2002;87:1036–1040.
  • Goriely A, Hansen RM, Taylor IB, et al. Activating mutations in FGFR3 and HRAS reveal a shared genetic origin for congenital disorders and testicular tumors. Nat Genet. 2009;41:1247–1252.
  • Feldman DR, Iyer G, Van Alstine L, et al. Presence of somatic mutations within PIK3CA, AKT, RAS, and FGFR3 but not BRAF in cisplatin-resistant germ cell tumors. Clin Cancer Res. 2014;20:3712–3720.
  • Zhang Y, Hiraishi Y, Wang H, et al. Constitutive activating mutation of the FGFR3b in oral squamous cell carcinomas. Int J Cancer. 2005;117:166–168.
  • Singh D, Chan JM, Zoppoli P, et al. Transforming fusions of FGFR and TACC genes in human glioblastoma. Science. 2012;337:1231–1235.
  • Williams SV, Hurst CD, Knowles MA. Oncogenic FGFR3 gene fusions in bladder cancer. Hum Mol Genet. 2013;22:795–803.
  • Shaver TM, Lehmann BD, Beeler JS, et al. Diverse, biologically relevant, and targetable gene rearrangements in triple-negative breast cancer and other malignancies. Cancer Res. 2016;76:4850–4860.
  • Daly C, Castanaro C, Zhang W, et al. FGFR3-TACC3 fusion proteins act as naturally occurring drivers of tumor resistance by functionally substituting for EGFR/ERK signaling. Oncogene. 2017;36:471–481.
  • Kalff A, Spencer A. The t(4;14) translocation and FGFR3 overexpression in multiple myeloma: prognostic implications and current clinical strategies. Blood Cancer J. 2012;2:e89.
  • Chesi M, Brents LA, Ely SA, et al. Activated fibroblast growth factor receptor 3 is an oncogene that contributes to tumor progression in multiple myeloma. Blood. 2001;97:729–736.
  • Sun X, Guo W, Shen JK, et al. Rhabdomyosarcoma: advances in molecular and cellular biology. Sarcoma. 2015;2015:232010.
  • Seki M, Nishimura R, Yoshida K, et al. Integrated genetic and epigenetic analysis defines novel molecular subgroups in rhabdomyosarcoma. Nat Commun. 2015;6:7557.
  • Tanner Y, Grose RP. Dysregulated FGF signalling in neoplastic disorders. Semin Cell Dev Biol. 2016;53:126–135.
  • Ronca R, Giacomini A, Rusnati M, et al. The potential of fibroblast growth factor/fibroblast growth factor receptor signaling as a therapeutic target in tumor angiogenesis. Expert Opin Ther Targets. 2015;19:1361–1377.
  • Jing Q, Wang Y, Liu H, et al. FGFs: crucial factors that regulate tumour initiation and progression. Cell Prolif. 2016;49:438–447.
  • Ghassemi S, Vejdovszky K, Sahin E, et al. FGF5 is expressed in melanoma and enhances malignancy in vitro and in vivo. Oncotarget. 2017;8:87750–87762.
  • Ronca R, Di Salle E, Giacomini A, et al. Long pentraxin-3 inhibits epithelial-mesenchymal transition in melanoma cells. Mol Cancer Ther. 2013;12:2760–2771.
  • Giacomini A, Matarazzo S, Pagano K, et al. A long pentraxin-3-derived pentapeptide for the therapy of FGF8b-driven steroid hormone-regulated cancers. Oncotarget. 2015;6:13790–13792.
  • Tuomela J, Härkönen P. Tumor models for prostate cancer exemplified by fibroblast growth factor 8-induced tumorigenesis and tumor progression. Reprod Biol. 2014;14:16–24.
  • Nagamatsu H, Teishima J, Goto K, et al. FGF19 promotes progression of prostate cancer. Prostate. 2015;75:1092–1101.
  • Feng S, Wang J, Zhang Y, et al. FGF23 promotes prostate cancer progression. Oncotarget. 2015;6:17291–17301.
  • Lee EK, Martinez MC, Blakely K, et al. FGF23: mediator of poor prognosis in a sizeable subgroup of patients with castration-resistant prostate cancer presenting with severe hypophosphatemia? Med Hypotheses. 2014;83:482–487.
  • Ronca R, Tamma R, Coltrini D, et al. Fibroblast growth factor modulates mast cell recruitment in a murine model of prostate cancer. Oncotarget. 2017;8:82583–82592.
  • Bluemn EG, Coleman IM, Lucas JM, et al. Androgen receptor pathway-independent prostate cancer is sustained through FGF signaling. Cancer Cell. 2017;32:474–489, e476.
  • Sharpe R, Pearson A, Herrera-Abreu MT, et al. FGFR signaling promotes the growth of triple-negative and basal-like breast cancer cell lines both in vitro and in vivo. Clin Cancer Res. 2011;17:5275–5286.
  • Aukes K, Forsman C, Brady NJ, et al. Breast cancer cell-derived fibroblast growth factors enhance osteoclast activity and contribute to the formation of metastatic lesions. PLoS One. 2017;12:e0185736.
  • Marek L, Ware KE, Fritzsche A, et al. Fibroblast growth factor (FGF) and FGF receptor-mediated autocrine signaling in non-small-cell lung cancer cells. Mol Pharmacol. 2009;75:196–197.
  • Qi L, Song W, Li L, et al. FGF4 induces epithelial-mesenchymal transition by inducing store-operated calcium entry in lung adenocarcinoma. Oncotarget. 2016;7:74015–74030.
  • Birrer MJ, Johnson ME, Hao K, et al. Whole genome oligonucleotide-based array comparative genomic hybridization analysis identified fibroblast growth factor 1 as a prognostic marker for advanced-stage serous ovarian adenocarcinomas. J Clin Oncol. 2007;25:2281–2287.
  • Sun Y, Fan X, Zhang Q, et al. Cancer-associated fibroblasts secrete FGF-1 to promote ovarian proliferation, migration, and invasion through the activation of FGF-1/FGFR4 signaling. Tumour Biol. 2017;39:1010428317712592.
  • Marshall ME, Hinz TK, Kono SA, et al. Fibroblast growth factor receptors are components of autocrine signaling networks in head and neck squamous cell carcinoma cells. Clin Cancer Res. 2011;17:5016–5025.
  • Maehara O, Suda G, Natsuizaka M, et al. Fibroblast growth factor-2-mediated FGFR/Erk signaling supports maintenance of cancer stem-like cells in esophageal squamous cell carcinoma. Carcinogenesis. 2017;38:1073–1083.
  • Soria J-C, DeBraud F, Bahleda R, et al. Phase I/IIa study evaluating the safety, efficacy, pharmacokinetics, and pharmacodynamics of lucitanib in advanced solid tumors. Ann Oncol. 2014;25:2244–2251.
  • Guffanti F, Chilà R, Bello E, et al. In vitro and in vivo activity of lucitanib in FGFR1/2 amplified or mutated cancer models. Neoplasia. 2017;19:35–42.
  • Formisano L, Stauffer KM, Young CD, et al. Association of FGFR1 with ERalpha maintains ligand-independent ER transcription and mediates resistance to estrogen deprivation in ER(+) breast cancer. Clin Cancer Res. 2017;23:6138–6150.
  • Sohl CD, Ryan MR, Luo B, et al. Illuminating the molecular mechanisms of tyrosine kinase inhibitor resistance for the FGFR1 gatekeeper mutation: the Achilles’ heel of targeted therapy. ACS Chem Biol. 2015;10:1319–1329.
  • Awasthi N, Schwarz RE. Profile of nintedanib in the treatment of solid tumors: the evidence to date. Onco Targets Ther. 2015;8:3691–3701.
  • Reck M. Nintedanib: examining the development and mechanism of action of a novel triple angiokinase inhibitor. Expert Rev Anticancer Ther. 2015;15:579–594.
  • Hibi M, Kaneda H, Tanizaki J, et al. FGFR gene alterations in lung squamous cell carcinoma are potential targets for the multikinase inhibitor nintedanib. Cancer Sci. 2016;107:1667–1676.
  • Porta C, Giglione P, Liguigli W, et al. Dovitinib (CHIR258, TKI258): structure, development and preclinical and clinical activity. Future Oncol. 2015;11:39–50.
  • Konecny GE, Kolarova T, O’Brien NA, et al. Activity of the fibroblast growth factor receptor inhibitors dovitinib (TKI258) and NVP-BGJ398 in human endometrial cancer cells. Mol Cancer Ther. 2013;12:632–642.
  • Kim HR, Kang HN, Shim HS, et al. Co-clinical trials demonstrate predictive biomarkers for dovitinib, an FGFR inhibitor, in lung squamous cell carcinoma. Ann Oncol. 2017;28:1250–1259.
  • André F, Bachelot T, Campone M, et al. Targeting FGFR with dovitinib (TKI258): preclinical and clinical data in breast cancer. Clin Cancer Res. 2013;19:3693–3702.
  • Musolino A, Campone M, Neven P, et al. Phase II, randomized, placebo-controlled study of dovitinib in combination with fulvestrant in postmenopausal patients with HR(+), HER2(-) breast cancer that had progressed during or after prior endocrine therapy. Breast Cancer Res. 2017;19:18.
  • Byron SA, Chen H, Wortmann A, et al. The N550K/H mutations in FGFR2 confer differential resistance to PD173074, dovitinib, and ponatinib ATP-competitive inhibitors. Neoplasia. 2013;15:975–988.
  • Patani H, Bunney TD, Thiyagarajan N, et al. Landscape of activating cancer mutations in FGFR kinases and their differential responses to inhibitors in clinical use. Oncotarget. 2016;7:24252–24268.
  • Trudel S, Li ZH, Wei E, et al. CHIR-258, a novel, multitargeted tyrosine kinase inhibitor for the potential treatment of t(4;14) multiple myeloma. Blood. 2005;105:2941–2948.
  • Scheid C, Reece D, Beksac M, et al. Phase 2 study of dovitinib in patients with relapsed or refractory multiple myeloma with or without t(4;14) translocation. Eur J Haematol. 2015;95:316–324.
  • Lee C-K, Lee ME, Lee WS, et al. Dovitinib (TKI258), a multi-target angiokinase inhibitor, is effective regardless of KRAS or BRAF mutation status in colorectal cancer. Am J Cancer Res. 2015;5:72–86.
  • Semrad TJ, Kim EJ, Tanaka MS, et al. Phase II study of dovitinib in patients progressing on anti-vascular endothelial growth factor therapy. Cancer Treat Res Commun. 2017;10:21–26.
  • Angevin E, Lopez-Martin JA, Lin CC, et al. Phase I study of dovitinib (TKI258), an oral FGFR, VEGFR, and PDGFR inhibitor, in advanced or metastatic renal cell carcinoma. Clin Cancer Res. 2013;19:1257–1268.
  • Motzer RJ, Hutson TE, Glen H, et al. Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, open-label, multicentre trial. Lancet Oncol. 2015;16:1473–1482.
  • Yadav SS, Li J, Stockert JA, et al. Induction of neuroendocrine differentiation in prostate cancer cells by dovitinib (TKI-258) and its therapeutic implications. Transl Oncol. 2017;10:357–366.
  • Porta R, Borea R, Coelho A, et al. FGFR a promising druggable target in cancer: molecular biology and new drugs. Crit Rev Oncol Hematol. 2017;113:256–267.
  • Chae YK, Ranganath K, Hammerman PS, et al. Inhibition of the fibroblast growth factor receptor (FGFR) pathway: the current landscape and barriers to clinical application. Oncotarget. 2017;8:16052–16074.
  • Nogova L, Sequist LV, Perez Garcia JM, et al. Evaluation of BGJ398, a fibroblast growth factor receptor 1-3 kinase inhibitor, in patients with advanced solid tumors harboring genetic alterations in fibroblast growth factor receptors: results of a global phase I, dose-escalation and dose-expansion study. J Clin Oncol. 2017;35:157–165.
  • Tabernero J, Bahleda R, Dienstmann R, et al. Phase I dose-escalation study of JNJ-42756493, an oral pan-fibroblast growth factor receptor inhibitor, in patients with advanced solid tumors. J Clin Oncol. 2015;33:3401–3408.
  • Gavine PR, Mooney L, Kilgour E, et al. AZD4547: an orally bioavailable, potent, and selective inhibitor of the fibroblast growth factor receptor tyrosine kinase family. Cancer Res. 2012;72:2045–2056.
  • Jang J, Kim HK, Bang H, et al. Antitumor effect of AZD4547 in a fibroblast growth factor receptor 2-amplified gastric cancer patient-derived cell model. Transl Oncol. 2017;10:469–475.
  • Siegfried JM, Farooqui M, Rothenberger NJ, et al. Interaction between the estrogen receptor and fibroblast growth factor receptor pathways in non-small cell lung cancer. Oncotarget. 2017;8:24063–24076.
  • Zhang J, Zhang L, Su X, et al. Translating the therapeutic potential of AZD4547 in FGFR1-amplified non-small cell lung cancer through the use of patient-derived tumor xenograft models. Clin Cancer Res. 2012;18:6658–6667.
  • Kwak Y, Cho H, Hur W, et al. Antitumor effects and mechanisms of AZD4547 on FGFR2-deregulated endometrial cancer cells. Mol Cancer Ther. 2015;14:2292–2302.
  • Yao T-J, Zhu J-H, Peng D-F, et al. AZD-4547 exerts potent cytostatic and cytotoxic activities against fibroblast growth factor receptor (FGFR)-expressing colorectal cancer cells. Tumour Biol. 2015;36:5641–5648.
  • Zhao Q, Parris AB, Howard EW, et al. FGFR inhibitor, AZD4547, impedes the stemness of mammary epithelial cells in the premalignant tissues of MMTV-ErbB2 transgenic mice. Sci Rep. 2017;7:11306.
  • Zhang H-R, Wang X-D, Yang X, et al. An FGFR inhibitor converts the tumor promoting effect of TGF-beta by the induction of fibroblast-associated genes of hepatoma cells. Oncogene. 2017;36:3831–3841.
  • Packer LM, Geng X, Bonazzi VF, et al. PI3K inhibitors synergize with FGFR inhibitors to enhance antitumor responses in FGFR2(mutant) endometrial cancers. Mol Cancer Ther. 2017;16:637–648.
  • Lehnen NC, von Mässenhausen A, Kalthoff H, et al. Fibroblast growth factor receptor 1 gene amplification in pancreatic ductal adenocarcinoma. Histopathology. 2013;63:157–166.
  • Göke F, Göke A, von Mässenhausen A, et al. Fibroblast growth factor receptor 1 as a putative therapy target in colorectal cancer. Digestion. 2013;88:172–181.
  • Schmidt K, Moser C, Hellerbrand C, et al. Targeting fibroblast growth factor receptor (FGFR) with BGJ398 in a gastric cancer model. Anticancer Res. 2015;35:6655–6665.
  • Wang Y, Ding X, Wang S, et al. Antitumor effect of FGFR inhibitors on a novel cholangiocarcinoma patient derived xenograft mouse model endogenously expressing an FGFR2-CCDC6 fusion protein. Cancer Lett. 2016;380:163–173.
  • Goyal L, Saha SK, Liu LY, et al. Polyclonal secondary FGFR2 mutations drive acquired resistance to FGFR inhibition in patients with FGFR2 fusion-positive cholangiocarcinoma. Cancer Discov. 2017;7:252–263.
  • Javle M, Lowery M, Shroff RT, et al. Phase II study of BGJ398 in patients with FGFR-altered advanced cholangiocarcinoma. J Clin Oncol. 2018;36:276–282.
  • Perera TPS, Jovcheva E, Mevellec L, et al. Discovery and pharmacological characterization of JNJ-42756493 (Erdafitinib), a functionally selective small-molecule FGFR family inhibitor. Mol Cancer Ther. 2017;16:1010–1020.
  • Di Stefano AL, Fucci A, Frattini V, et al. Detection, characterization, and inhibition of FGFR-TACC fusions in IDH wild-type glioma. Clin Cancer Res. 2015;21:3307–3317.
  • Katoh M. FGFR inhibitors: effects on cancer cells, tumor microenvironment and whole-body homeostasis (review). Int J Mol Med. 2016;38:3–15.
  • Hierro C, Rodon J, Tabernero J. Fibroblast Growth Factor (FGF) receptor/FGF inhibitors: novel targets and strategies for optimization of response of solid tumors. Semin Oncol. 2015;42:801–819.
  • Dieci MV, Arnedos M, Andre F, et al. Fibroblast growth factor receptor inhibitors as a cancer treatment: from a biologic rationale to medical perspectives. Cancer Discov. 2013;3:264–279.
  • Carter EP, Fearon AE, Grose RP. Careless talk costs lives: fibroblast growth factor receptor signalling and the consequences of pathway malfunction. Trends Cell Biol. 2015;25:221–233.
  • Lee J, Bang YJ, Rha SY, et al. Antitumor activity and safety of FPA144, an ADCC-enhanced, FGFR2b isoform-selective monoclonal antibody, in patients with FGFR2b+ gastric cancer and advanced solid tumors. J Clin Oncol. 2016;24(15_suppl):2502.
  • Catenacci DVT, Rha SY, Bang H, et al. Updated antitumor activity and safety of FPA144, an ADCC-enhanced, FGFR2b isoform-specific monoclonal antibody, in patients with FGFR2b+ gastric cancer. J Clin Oncol. 2017;35(15_suppl):4067.
  • Presta M, Chiodelli P, Giacomini A, et al. Fibroblast growth factors (FGFs) in cancer: FGF traps as a new therapeutic approach. Pharmacol Ther. 2017;179:171–187.
  • Tao J, Xiang J-J, Li D, et al. Selection and characterization of a human neutralizing antibody to human fibroblast growth factor-2. Biochem Biophys Res Commun. 2010;394:767–773.
  • Wang L, Park H, Chhim S, et al. A novel monoclonal antibody to fibroblast growth factor 2 effectively inhibits growth of hepatocellular carcinoma xenografts. Mol Cancer Ther. 2012;11:864–872.
  • de Aguiar RB, Parise CB, Souza CR, et al. Blocking FGF2 with a new specific monoclonal antibody impairs angiogenesis and experimental metastatic melanoma, suggesting a potential role in adjuvant settings. Cancer Lett. 2016;371:151–160.
  • Tanaka A, Furuya A, Yamasaki M, et al. High frequency of fibroblast growth factor (FGF) 8 expression in clinical prostate cancers and breast tissues, immunohistochemically demonstrated by a newly established neutralizing monoclonal antibody against FGF 8. Cancer Res. 1998;58:2053–2056.
  • Shimada N, Ishii T, Imada T, et al. A neutralizing anti-fibroblast growth factor 8 monoclonal antibody shows potent antitumor activity against androgen-dependent mouse mammary tumors in vivo. Clin Cancer Res. 2005;11:3897–3904.
  • Maruyama-Takahashi K, Shimada N, Imada T, et al. A neutralizing anti-fibroblast growth factor (FGF) 8 monoclonal antibody shows anti-tumor activity against FGF8b-expressing LNCaP xenografts in androgen-dependent and -independent conditions. Prostate. 2008;68:640–650.
  • Harding TC, Long L, Palencia S, et al. Blockade of nonhormonal fibroblast growth factors by FP-1039 inhibits growth of multiple types of cancer. Sci Transl Med. 2013;5:178ra139.
  • Blackwell C, Sherk C, Fricko M, et al. Inhibition of FGF/FGFR autocrine signaling in mesothelioma with the FGF ligand trap, FP-1039/GSK3052230. Oncotarget. 2016;7:39861–39871.
  • Tolcher AW, Papadopoulos KP, Patnaik A, et al. A phase I, first in human study of FP-1039 (GSK3052230), a novel FGF ligand trap, in patients with advanced solid tumors. Ann Oncol. 2016;27:526–532.
  • Li D, Wei X, Xie K, et al. A novel decoy receptor fusion protein for FGF-2 potently inhibits tumour growth. Br J Cancer. 2014;111:68–77.
  • Giorgio C, Incerti M, Corrado M, et al. Pharmacological evaluation of new bioavailable small molecules targeting Eph/ephrin interaction. Biochem Pharmacol. 2018;147:21–29.
  • Foglieni C, Pagano K, Lessi M, et al. Integrating computational and chemical biology tools in the discovery of antiangiogenic small molecule ligands of FGF2 derived from endogenous inhibitors. Sci Rep. 2016;6:23432.
  • Leali D, Bianchi R, Bugatti A, et al. Fibroblast growth factor 2-antagonist activity of a long-pentraxin 3-derived anti-angiogenic pentapeptide. J Cell Mol Med. 2010;14:2109–2121.
  • Ronca R, Giacomini A, Di Salle E, et al. Long-pentraxin 3 derivative as a small-molecule FGF trap for cancer therapy. Cancer Cell. 2015;28:225–239.
  • Castelli R, Giacomini A, Anselmi M, et al. Synthesis, structural elucidation, and biological evaluation of NSC12, an orally available Fibroblast Growth Factor (FGF) ligand trap for the treatment of FGF-dependent lung tumors. J Med Chem. 2016;59:4651–4663.
  • Chiodelli P, Bugatti A, Urbinati C, et al. Heparin/Heparan sulfate proteoglycans glycomic interactome in angiogenesis: biological implications and therapeutical use. Molecules. 2015;20:6342–6363.
  • Castelli R, Porro F, Tarsia P. The heparins and cancer: review of clinical trials and biological properties. Vasc Med. 2004;9:205–213.
  • Daigo K, Inforzato A, Barajon I, et al. Pentraxins in the activation and regulation of innate immunity. Immunol Rev. 2016;274:202–217.
  • Giacomini A, Ghedini GC, Presta M, et al. Long pentraxin 3: a novel multifaceted player in cancer. Biochim Biophys Acta. 2017;1869:53–63.
  • Ronca R, Alessi P, Coltrini D, et al. Long pentraxin-3 as an epithelial-stromal fibroblast growth factor-targeting inhibitor in prostate cancer. J Pathol. 2013;230:228–238.
  • Camozzi M, Rusnati M, Bugatti A, et al. Identification of an antiangiogenic FGF2-binding site in the N terminus of the soluble pattern recognition receptor PTX3. J Biol Chem. 2006;281:22605–22613.
  • Bono F, De Smet F, Herbert C, et al. Inhibition of tumor angiogenesis and growth by a small-molecule multi-FGF receptor blocker with allosteric properties. Cancer Cell. 2013;23:477–488.
  • Herbert C, Schieborr U, Saxena K, et al. Molecular mechanism of SSR128129E, an extracellularly acting, small-molecule, allosteric inhibitor of FGF receptor signaling. Cancer Cell. 2013;23:489–501.
  • Ader I, Delmas C, Skuli N, et al. Preclinical evidence that SSR128129E – a novel small-molecule multi-fibroblast growth factor receptor blocker–radiosensitises human glioblastoma. Eur J Cancer. 2014;50:2351–2359.
  • Conn PJ, Christopoulos A, Lindsley CW. Allosteric modulators of GPCRs: a novel approach for the treatment of CNS disorders. Nat Rev Drug Discov. 2009;8:41–54.
  • Christopoulos A. Allosteric binding sites on cell-surface receptors: novel targets for drug discovery. Nat Rev Drug Discov. 2002;1:198–210.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.