Rogaratinib: A potent and selective pan-FGFR inhibitor with broad antitumor activity in FGFR-overexpressing preclinical cancer models

Sylvia Grünewald, Oliver Politz, Sebastian Bender, Mélanie Héroult, Klemens Lustig, Uwe Thuss, Christoph Kneip, Charlotte Kopitz, Dieter Zopf, Marie-Pierre Collin, Ulf Boemer, Stuart Ince, Peter Ellinghaus, Dominik Mumberg, Holger Hess-Stumpp, Karl Ziegelbauer, Sylvia Grünewald, Oliver Politz, Sebastian Bender, Mélanie Héroult, Klemens Lustig, Uwe Thuss, Christoph Kneip, Charlotte Kopitz, Dieter Zopf, Marie-Pierre Collin, Ulf Boemer, Stuart Ince, Peter Ellinghaus, Dominik Mumberg, Holger Hess-Stumpp, Karl Ziegelbauer

Abstract

Aberrant activation in fibroblast growth factor signaling has been implicated in the development of various cancers, including squamous cell lung cancer, squamous cell head and neck carcinoma, colorectal and bladder cancer. Thus, fibroblast growth factor receptors (FGFRs) present promising targets for novel cancer therapeutics. Here, we evaluated the activity of a novel pan-FGFR inhibitor, rogaratinib, in biochemical, cellular and in vivo efficacy studies in a variety of preclinical cancer models. In vitro kinase activity assays demonstrate that rogaratinib potently and selectively inhibits the activity of FGFRs 1, 2, 3 and 4. In line with this, rogaratinib reduced proliferation in FGFR-addicted cancer cell lines of various cancer types including lung, breast, colon and bladder cancer. FGFR and ERK phosphorylation interruption by rogaratinib treatment in several FGFR-amplified cell lines suggests that the anti-proliferative effects are mediated by FGFR/ERK pathway inhibition. Furthermore, rogaratinib exhibited strong in vivo efficacy in several cell line- and patient-derived xenograft models characterized by FGFR overexpression. The observed efficacy of rogaratinib strongly correlated with FGFR mRNA expression levels. These promising results warrant further development of rogaratinib and clinical trials are currently ongoing (ClinicalTrials.gov Identifiers: NCT01976741, NCT03410693, NCT03473756).

Keywords: cancer; colorectal cancer; fibroblast growth factor receptor; preclinical models; rogaratinib.

© 2019 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.

Figures

Figure 1
Figure 1
Structure and in vitro activity of rogaratinib. (a) Chemical structure of rogaratinib (BAY 1163877). (b) TREEspot™ interaction map of KINOMEscan™ assay results with 100 nM rogaratinib. (c) Viability of HUVEC cells stimulated with growth factors FGF2 (5 ng/mL) or VEGF‐A (20 ng/mL) in minimal medium after a 72‐h treatment with increasing concentrations of rogaratinib. (d) Correlation of FGFR mRNA levels (sum of Z‐scores for subtypes with Z‐score > 1, derived from CCLE database) and sensitivity (IC50 of inhibition of cell proliferation expressed as LogIC50) to rogaratinib in a panel of cancer cell lines covering various cancer types. Dotted lines indicate IC50 levels.
Figure 2
Figure 2
Effects of rogaratinib treatment on phosphorylation of FGFR and ERK in various cell lines as determined by Western blotting. (a) p‐FGFR4, total FGFR, p(T202/Y204,T185/Y187)‐ERK1/2, and total ERK1/2 expression in MDA‐MB‐453 cells after treatment with 0–1,000 nM rogaratinib. (b) p‐FGFR2, α‐tubulin, p(T202/Y204,T185/Y187)‐ERK1/2, and total ERK1/2 expression in NCI‐H716 cells after treatment with 0 or 100 nM rogaratinib. (c) p(T202/Y204,T185/Y187)‐ERK1/2 and total ERK1/2 expression in UM‐UC‐3, RT‐112, NCI‐H520, NCI‐H1581 and C51 cells after treatment with 0–10,000 nM rogaratinib.
Figure 3
Figure 3
Antitumor activity of rogaratinib in the C51 syngeneic colon cancer model in immunocompetent BALB/c mice and the C51 xenogeneic model in nude rats. (a) Growth curves of C51 colon tumors treated p.o. with vehicle or rogaratinib (25, 50 or 75 mg/kg, QD; or 25 mg/kg BID) in BALB/c mice as measured by tumor volumes (mm3, mean +/− SD) over time. (b) Weights of C51 tumors in mice described in (a) at the end of the study (day 18). (c) Relative volumes (100% equals volume at start of treatment) of C51 tumors described in (a) at the end of the study (day 18) analyzed using RECIST criteria. PD, progressive disease; SD, stable disease; PR, partial response; CR, complete response. (d) Pharmacokinetic profile of rogaratinib in plasma of C51 tumor‐bearing BALB/c mice, shown as unbound concentration over 24 h after the last dose of rogaratinib and simulated exposure levels for the twice daily application of 25 mg/kg. The dotted lines denote the IC50 values of rogaratinib (280 and 430 nM) in C51 cells for inhibition of p‐ERK and proliferation, respectively. (e) Growth curves of C51 tumors in nude rats treated p.o. with vehicle or rogaratinib (10 or 50 mg/kg, QD) as measured by tumor volumes (mm3, mean ± SD) over time. (f) Weight of C51 tumors in rats described in (e) at the end of the study. (g) Relative volumes of C51 tumors described in (e) at the end of the study analyzed using RECIST criteria. PD, progressive disease; SD, stable disease; PR, partial response. Stars in a, b, e and f denote statistical difference compared to vehicle group. *, p < 0.05; **, p < 0.01; ***, p < 0.001; NS, non‐significant. [Color figure can be viewed at wileyonlinelibrary.com]
Figure 4
Figure 4
In vivo antitumor efficacy and mechanism of action of rogaratinib in the human NCI‐H716 colorectal xenograft model in immunocompromised NMRInu/nu mice. (a) Growth curves of human NCI‐H716 colorectal tumors treated p.o. with vehicle or rogaratinib (35, 50 or 65 mg/kg, BID) from days 22 to 38 after tumor inoculation as measured by tumor volumes (mm3, mean ± SD) over time. Stars denote statistical difference compared to vehicle group. **, p < 0.01; ***, p < 0.001. (b) Relative volumes of NCI‐H716 tumors described in (a) on the last treatment day (day 38). PD, progressive disease; SD, stable disease; PR, partial response. (c) Expression of p‐FGFR2, p‐ERK, total ERK and actin in NCI‐H716 tumor tissue after rogaratinib treatment. After a drug‐free observation period of 10 days, mice of the three rogaratinib‐dose groups (35, 50 or 65 mg/kg) in (a) received a single respective dose of rogaratinib or vehicle. Plasma and tumors were collected 1, 2, 5 or 24 h after treatment for PK/PD analysis and phosphorylation of FGFR2 and ERK1/2 was determined by Western blotting. [Color figure can be viewed at wileyonlinelibrary.com]
Figure 5
Figure 5
In vivo antitumor efficacy of rogaratinib in the DMS‐114 lung cancer xenograft model. (a) Growth curves of DMS‐114 lung tumors treated with vehicle, rogaratinib (50 mg/kg, p.o., BID), docetaxel (30 mg/kg, i.v., Q7D), combination of rogaratinib (50 mg/kg, BID)/docetaxel (30 mg/kg, Q7D), combination of carboplatin (80 mg/kg, i.p., Q7D)/paclitaxel (24 mg/kg, Q7D) or combination of carboplatin (80 mg/kg, Q7D)/paclitaxel (24 mg/kg, i.p., Q7D)/rogaratinib (50 mg/kg, BID), as measured by tumor volumes (mm3, mean ± SD) over time. (b) Weights of DMS‐114 tumors described in (a) at the end of the study. (c) Relative volumes of DMS‐114 tumors described in (a) at the end of the study. PD, progressive disease; SD, stable disease; PR, partial response; CR, complete response. (d) Body weights of mice described in (a) relative to body weights at treatment start. Stars denote statistical difference compared to vehicle group. **, p < 0.01; ***, p < 0.001.
Figure 6
Figure 6
In vivo antitumor efficacy of rogaratinib in the patient‐derived LU299 lung cancer xenograft model. (a) Growth curves of LU299 lung tumors treated with vehicle, rogaratinib (50 mg/kg, BID), docetaxel (15 mg/kg, i.v., Q7D), or combination of rogaratinib and docetaxel for 33 days followed by a drug‐free observation period of 53 days, as measured by tumor volumes (mm3, mean ± SD) over time. Stars denote statistical difference compared to vehicle group. *, p < 0.05; ***, p < 0.01. (b) Relative volumes of tumors described in (a) at the last treatment day (day 54 after tumor inoculation). PD, progressive disease; SD, stable disease; PR, partial response; CR, complete response. (c) Relative body weights of LU299 mice described in (a). Body weight loss in docetaxel groups recovered after dosing break at day 37. (d) Growth curves of single mice of LU299 lung tumors treated as noted above (a).

References

    1. Katoh M. Therapeutics targeting FGF signaling network in human diseases. Trends Pharmacol Sci 2016;37:1081–96.
    1. Turner N, Grose R. Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer 2010;10:116–29.
    1. 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;22:259–67.
    1. Cihoric N, Savic S, Schneider S, et al. Prognostic role of FGFR1 amplification in early‐stage non‐small cell lung cancer. Br J Cancer 2014;110:2914–22.
    1. Heist RS, Mino‐Kenudson M, Sequist LV, et al. FGFR1 amplification in squamous cell carcinoma of the lung. J Thorac Oncol 2012;7:1775–80.
    1. Preusser M, Berghoff AS, Berger W, et al. High rate of FGFR1 amplifications in brain metastases of squamous and non‐squamous lung cancer. Lung Cancer 2014;83:83–9.
    1. 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.
    1. Goke F, Bode M, Franzen A, et al. Fibroblast growth factor receptor 1 amplification is a common event in squamous cell carcinoma of the head and neck. Mod Pathol 2013;26:1298–306.
    1. Koole K, Brunen D, van Kempen PM, et al. FGFR1 is a potential prognostic biomarker and therapeutic target in head and neck squamous cell carcinoma. Clin Cancer Res 2016;22:3884–93.
    1. Cappellen D, De Oliveira C, Ricol D, et al. Frequent activating mutations of FGFR3 in human bladder and cervix carcinomas. Nat Genet 1999;23:18–20.
    1. Gust KM, McConkey DJ, Awrey S, et al. Fibroblast growth factor receptor 3 is a rational therapeutic target in bladder cancer. Mol Cancer Ther 2013;12:1245–54.
    1. Morales‐Barrera R, Suarez C, de Castro AM, et al. Targeting fibroblast growth factor receptors and immune checkpoint inhibitors for the treatment of advanced bladder cancer: new direction and new Hope. Cancer Treat Rev 2016;50:208–16.
    1. Tomlinson DC, Baldo O, Harnden P, et al. FGFR3 protein expression and its relationship to mutation status and prognostic variables in bladder cancer. J Pathol 2007;213:91–8.
    1. Dienstmann R, Rodon J, Prat A, et al. Genomic aberrations in the FGFR pathway: opportunities for targeted therapies in solid tumors. Ann Oncol 2014;25:552–63.
    1. Katoh M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat Rev Clin Oncol 2018;16:105–22.
    1. Goke F, Franzen A, Hinz TK, et al. FGFR1 expression levels predict BGJ398 sensitivity of FGFR1‐dependent head and neck squamous cell cancers. Clin Cancer Res 2015;21:4356–64.
    1. Wynes MW, Hinz TK, Gao D, et al. FGFR1 mRNA and protein expression, not gene copy number, predict FGFR TKI sensitivity across all lung cancer histologies. Clin Cancer Res 2014;20:3299–309.
    1. Katoh M. FGFR inhibitors: effects on cancer cells, tumor microenvironment and whole‐body homeostasis (review). Int J Mol Med 2016;38:3–15.
    1. Collin M‐P, Lobell M, Hübsch W, et al. Discovery of Rogaratinib (BAY 1163877): a pan‐FGFR inhibitor. ChemMedChem 2018;13:1–10.
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228–47.
    1. Uitdehaag JCM, de Roos JADM, Prinsen MBW, et al. Cell panel profiling reveals conserved therapeutic clusters and differentiates the mechanism of action of different PI3K/mTOR, Aurora kinase and EZH2 inhibitors. Mol Cancer Ther 2016;15:3097–3109.
    1. Brower M, Carney DN, Oie HK, et al. Growth of cell lines and clinical specimens of human non‐small cell lung cancer in a serum‐free defined medium. Cancer Res 1986;46:798–806.
    1. Grossman HB, Wedemeyer G, Ren L, et al. Improved growth of human Urothelial carcinoma cell cultures. J Urol 1986;136:953–9.
    1. Goke A, Goke R, Ofner A, et al. The FGFR inhibitor NVP‐BGJ398 induces NSCLC cell death by activating caspase‐dependent pathways as well as Caspase‐independent apoptosis. Anticancer Res 2015;35:5873–9.
    1. Mathur A, Ware C, Davis L, et al. FGFR2 is amplified in the NCI‐H716 colorectal cancer cell line and is required for growth and survival. PLoS One 2014;9:e98515.
    1. Pettengill OS, Sorenson GD, Wurster‐Hill DH, et al. Isolation and growth characteristics of continuous cell lines from small‐cell carcinoma of the lung. Cancer 1980;45:906–18.
    1. Hackenberg R, Lüttchens S, Hofmann J, et al. Androgen sensitivity of the new human breast cancer cell line MFM‐223. Cancer Res 1991;51:5722–7.
    1. Tomlinson DC, Lamont FR, Shnyder SD, et al. Fibroblast growth factor receptor 1 promotes proliferation and survival via activation of the mitogen‐activated protein kinase pathway in bladder cancer. Cancer Res 2009;69:4613–20.
    1. Guagnano V, Furet P, Spanka C, et al. Discovery of 3‐(2,6‐dichloro‐3,5‐dimethoxy‐phenyl)‐1‐{6‐[4‐(4‐ethyl‐piperazin‐1‐yl)‐phenylamin o]‐pyrimidin‐4‐yl}‐1‐methyl‐urea (NVP‐BGJ398), a potent and selective inhibitor of the fibroblast growth factor receptor family of receptor tyrosine kinase. J Med Chem 2011;54:7066–83.
    1. Barretina J, Caponigro G, Stransky N, et al. The cancer cell line encyclopedia enables predictive modeling of anticancer drug sensitivity. Nature 2012;483:603–7.
    1. 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;35:157–65.
    1. Kotani H, Ebi H, Kitai H, et al. Co‐active receptor tyrosine kinases mitigate the effect of FGFR inhibitors in FGFR1‐amplified lung cancers with low FGFR1 protein expression. Oncogene 2016;35:3587–97.
    1. Beenken A, Mohammadi M. The FGF family: biology, pathophysiology and therapy. Nat Rev Drug Discov 2009;8:235–53.
    1. Zhao X, Xu F, Dominguez NP, et al. FGFR4 provides the conduit to facilitate FGF19 signaling in breast cancer progression. Mol Carcinog 2018;0;57:1616–25.
    1. Perera TP, 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;16:1010–20.
    1. Benvenuti S, Sartore‐Bianchi A, Di Nicolantonio F, et al. Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti‐epidermal growth factor receptor antibody therapies. Cancer Res 2007;67:2643–8.
    1. Lievre A, Bachet JB, Boige V, et al. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J Clin Oncol 2008;26:374–9.
    1. Haugsten EM, Wiedlocha A, Olsnes S, et al. Roles of fibroblast growth factor receptors in carcinogenesis. Mol Cancer Res 2010;8:1439–52.
    1. Ong SH, Hadari YR, Gotoh N, et al. Stimulation of phosphatidylinositol 3‐kinase by fibroblast growth factor receptors is mediated by coordinated recruitment of multiple docking proteins. Proc Natl Acad Sci USA 2001;98:6074–9.
    1. Ornitz DM, Itoh N. The fibroblast growth factor signaling pathway. Wiley Interdiscip Rev Dev Biol 2015;4:215–66.
    1. Hu Y, Lu H, Zhang J, et al. Essential role of AKT in tumor cells addicted to FGFR. Anticancer Drugs 2014;25:183–8.
    1. Starska K, Forma E, Lewy‐Trenda I, et al. Fibroblast growth factor receptor 1 and 3 expression is associated with regulatory PI3K/AKT kinase activity, as well as invasion and prognosis, in human laryngeal cancer. Cell Oncol 2018;41:253–68.

Source: PubMed

Подписаться