Preclinical Activity of ARQ 087, a Novel Inhibitor Targeting FGFR Dysregulation

Terence G Hall, Yi Yu, Sudharshan Eathiraj, Yunxia Wang, Ronald E Savage, Jean-Marc Lapierre, Brian Schwartz, Giovanni Abbadessa, Terence G Hall, Yi Yu, Sudharshan Eathiraj, Yunxia Wang, Ronald E Savage, Jean-Marc Lapierre, Brian Schwartz, Giovanni Abbadessa

Abstract

Dysregulation of Fibroblast Growth Factor Receptor (FGFR) signaling through amplifications, mutations, and gene fusions has been implicated in a broad array of cancers (e.g. liver, gastric, ovarian, endometrial, and bladder). ARQ 087 is a novel, ATP competitive, small molecule, multi-kinase inhibitor with potent in vitro and in vivo activity against FGFR addicted cell lines and tumors. Biochemically, ARQ 087 exhibited IC50 values of 1.8 nM for FGFR2, and 4.5 nM for FGFR1 and 3. In cells, inhibition of FGFR2 auto-phosphorylation and other proteins downstream in the FGFR pathway (FRS2α, AKT, ERK) was evident by the response to ARQ 087 treatment. Cell proliferation studies demonstrated ARQ 087 has anti-proliferative activity in cell lines driven by FGFR dysregulation, including amplifications, fusions, and mutations. Cell cycle studies in cell lines with high levels of FGFR2 protein showed a positive relationship between ARQ 087 induced G1 cell cycle arrest and subsequent induction of apoptosis. In addition, ARQ 087 was effective at inhibiting tumor growth in vivo in FGFR2 altered, SNU-16 and NCI-H716, xenograft tumor models with gene amplifications and fusions. ARQ 087 is currently being studied in a phase 1/2 clinical trial that includes a sub cohort for intrahepatic cholangiocarcinoma patients with confirmed FGFR2 gene fusions (NCT01752920).

Conflict of interest statement

TGH, RES, YY, SE, TH, GA, JS, and BS are employees of ArQule, Inc. KP and WS have no competing financial interests. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Mode of FGFR inhibition for…
Fig 1. Mode of FGFR inhibition for ARQ 087.
(A) ARQ 069. (B) ARQ 087. (C) Enzyme kinetic analysis was performed to determine the mode of inhibition of ARQ 087 with FGFR1 and FGFR2. Concentrations of ATP [ATP] and ARQ 087 [I] are indicated on the graphs. The rate plotted is the AlphaScreen™ signal obtained from the plate reader with background correction. The experiments were conducted in triplicates and the means and the standard deviations were plotted. The number of parameter (np), the sum of squares rel (SSrel), Akaike information criterion (AIC), weight of AIC (w(AIC)), Bayesian information criterion (BIC) and the weight of BIC (w(BIC)) for each binding mode were determined by the DynaFit software and are summarized below the plots. The dissociation constant (Ki) of ARQ 087 for FGFR1 and FGFR2 are shown. (D) The effect of ARQ 087 on the activation of FGFR1 and FGFR2 was examined in a continuous autophosphorylation assay.
Fig 2. ARQ 087 inhibits FGFR phosphorylation.
Fig 2. ARQ 087 inhibits FGFR phosphorylation.
COS-1 cells ectopically expressing FGFR1, FGFR2, FGFR3 or FGFR4 were treated with the indicated concentrations of ARQ 087 for 2 hours followed by stimulation with 100 pM of FGF1/2/7 for 15 minutes. Total and phospho-FGFR was assessed by Western blot analyses. β-Actin was used as a loading control. The EC50 values of individual FGFR family members are shown.
Fig 3. ARQ 087 inhibits the FGFR…
Fig 3. ARQ 087 inhibits the FGFR pathway in cancer cell lines.
(A) NCI-H716, SNU-16, and KATO-III cells were treated with the indicated concentrations of ARQ 087 for 2 hours, and phospho-FGFR was assessed by Western blot analyses. β-Actin was used as a loading control. (B) NCI-H716 and SNU-16 cells were treated with increasing concentrations of ARQ 087 from 0 μM to 3 μM for 2 hours. The quantity of indicated protein was assessed by Western blot analyses. β-Actin was used as a loading control. (C) KG-1 cells were treated with indicated concentrations of ARQ 087 for 2 hours followed by stimulation with a mixture of FGF1/FGF2/FGF7 for 15 minutes. Cell lysates were analyzed by Western blot to determine the expression of phospho-FGFR, phospho-ERK, and β-actin.
Fig 4. ARQ 087 arrests cells in…
Fig 4. ARQ 087 arrests cells in the G1 cell cycle phase and induces apoptosis.
(A) NCI-H716 and SNU-16 cells were treated with 1 μM of ARQ 087 or vehicle for 72 hours. Cell cycle profiles were measured by flow cytometric analyses. (B) SNU-16 cells were treated with ARQ 087 (1 mM) for 0, 24, 48 and 72 hrs. Western blot analysis was performed for XIAP, cleaved-PARP (c-PARP), activated-caspase 3 (a-Caspase 3), and phospho-p53 were assessed by Western blot analyses. β-Actin was used as the loading control.
Fig 5. ARQ 087 inhibits the FGFR…
Fig 5. ARQ 087 inhibits the FGFR pathway in xenograft tumors.
Mice were dosed with 75 mg/kg of ARQ 087, and sacrificed at day 9 after administration Tumor samples were collected 4 hr after last dose. IHC staining of pFGFR, FGFR2, pFRS2-a and pERK were performed from tumor tissues. The intensity of staining was scored by a veterinary pathologist and presented as mean±SEM. Representative photomicrographs are shown.
Fig 6. ARQ 087 activity in tumor…
Fig 6. ARQ 087 activity in tumor growth models.
(A, C, D), Growth is inhibited in BaF3/FGFR2, SNU-16, and NCI-H716, xenograft models, but not inhibited in (B) BaF3/INSR. Results are represented as the mean of tumor volume in mm3 ± SEM of each group (n = 8–10) in function of the treatment period. The tumor growth inhibition (TGI) is indicated on the plots.
Fig 7. Animal weights in BaF3/FGFR2 animals…
Fig 7. Animal weights in BaF3/FGFR2 animals dosed with ARQ 087.
Mice were dosed with 0, 50, 100, and 150 mg/kg of ARQ 087, and sacrificed at 10 days after administration after the start of dosing. Mean weights +/- SEM.

References

    1. Wesche J, Haglund K, Haugsten EM. Fibroblast growth factors and their receptors in cancer. Biochem J. 2011;437(2):199–213. 10.1042/BJ20101603 .
    1. Beenken A, Mohammadi M. The FGF family: biology, pathophysiology and therapy. Nature reviews Drug discovery. 2009;8(3):235–53. 10.1038/nrd2792
    1. Turner N, Grose R. Fibroblast growth factor signalling: from development to cancer. Nature Reviews Cancer. 2010;10(2):116–29. 10.1038/nrc2780
    1. Kelleher FC, O'Sullivan H, Smyth E, McDermott R, Viterbo A. Fibroblast growth factor receptors, developmental corruption and malignant disease. Carcinogenesis. 2013;34(10):2198–205. 10.1093/carcin/bgt254 .
    1. Graham RP, Barr Fritcher EG, Pestova E, Schulz J, Sitailo LA, Vasmatzis G, et al. Fibroblast growth factor receptor 2 translocations in intrahepatic cholangiocarcinoma. Human pathology. 2014;45(8):1630–8. 10.1016/j.humpath.2014.03.014 .
    1. Finch PW, Rubin JS. Keratinocyte growth factor expression and activity in cancer: implications for use in patients with solid tumors. J Natl Cancer Inst. 2006;98(12):812–24. 10.1093/jnci/djj228 .
    1. Katoh Y, Katoh M. FGFR2-related pathogenesis and FGFR2-targeted therapeutics (Review). Int J Mol Med. 2009;23(3):307–11. .
    1. Weiss J, Sos ML, Seidel D, Peifer M, Zander T, Heuckmann JM, et al. Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Science translational medicine. 2010;2(62):62ra93 10.1126/scitranslmed.3001451
    1. Dutt A, Ramos AH, Hammerman PS, Mermel C, Cho J, Sharifnia T, et al. Inhibitor-sensitive FGFR1 amplification in human non-small cell lung cancer. PloS one. 2011;6(6):e20351 10.1371/journal.pone.0020351
    1. Xie L, Su X, Zhang L, Yin X, Tang L, Zhang X, et al. FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547. Clinical cancer research: an official journal of the American Association for Cancer Research. 2013;19(9):2572–83. 10.1158/1078-0432.CCR-12-3898 .
    1. Bai A, Meetze K, Vo NY, Kollipara S, Mazsa EK, Winston WM, et al. GP369, an FGFR2-IIIb-specific antibody, exhibits potent antitumor activity against human cancers driven by activated FGFR2 signaling. Cancer research. 2010;70(19):7630–9. 10.1158/0008-5472.CAN-10-1489 .
    1. Nakanishi Y, Akiyama N, Tsukaguchi T, Fujii T, Sakata K, Sase H, et al. The fibroblast growth factor receptor genetic status as a potential predictor of the sensitivity to CH5183284/Debio 1347, a novel selective FGFR inhibitor. Molecular cancer therapeutics. 2014;13(11):2547–58. 10.1158/1535-7163.MCT-14-0248 .
    1. Liang G, Chen G, Wei X, Zhao Y, Li X. Small molecule inhibition of fibroblast growth factor receptors in cancer. Cytokine & growth factor reviews. 2013;24(5):467–75. 10.1016/j.cytogfr.2013.05.002 .
    1. Lemieux SM, Hadden MK. Targeting the fibroblast growth factor receptors for the treatment of cancer. Anticancer Agents Med Chem. 2013;13(5):748–61. .
    1. Kumar SB, Narasu L, Gundla R, Dayam R, ARPS J. Fibroblast growth factor receptor inhibitors. Curr Pharm Des. 2013;19(4):687–701. .
    1. Chase A, Bryant C, Score J, Cross NC. Ponatinib as targeted therapy for FGFR1 fusions associated with the 8p11 myeloproliferative syndrome. Haematologica. 2013;98(1):103–6. 10.3324/haematol.2012.066407
    1. Kim ST, Jang HL, Lee SJ, Lee J, Choi YL, Kim KM, et al. Pazopanib, a novel multitargeted kinase inhibitor, shows potent in vitro antitumor activity in gastric cancer cell lines with FGFR2 amplification. Molecular cancer therapeutics. 2014;13(11):2527–36. 10.1158/1535-7163.MCT-14-0255 .
    1. Guagnano V, Furet P, Spanka C, Bordas V, Le Douget M, Stamm 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. Journal of medicinal chemistry. 2011;54(20):7066–83. 10.1021/jm2006222 .
    1. Williams SV, Hurst CD, Knowles MA. Oncogenic FGFR3 gene fusions in bladder cancer. Human molecular genetics. 2013;22(4):795–803. 10.1093/hmg/dds486
    1. Borad MJ, Gores GJ, Roberts LR. Fibroblast growth factor receptor 2 fusions as a target for treating cholangiocarcinoma. Curr Opin Gastroenterol. 2015;31(3):264–8. 10.1097/MOG.0000000000000171 .
    1. Borad MJ, Champion MD, Egan JB, Liang WS, Fonseca R, Bryce AH, et al. Integrated genomic characterization reveals novel, therapeutically relevant drug targets in FGFR and EGFR pathways in sporadic intrahepatic cholangiocarcinoma. PLoS genetics. 2014;10(2):e1004135 10.1371/journal.pgen.1004135
    1. Capelletti M, Dodge ME, Ercan D, Hammerman PS, Park SI, Kim J, et al. Identification of recurrent FGFR3-TACC3 fusion oncogenes from lung adenocarcinoma. Clinical cancer research: an official journal of the American Association for Cancer Research. 2014;20(24):6551–8. 10.1158/1078-0432.CCR-14-1337 .
    1. Parker BC, Engels M, Annala M, Zhang W. Emergence of FGFR family gene fusions as therapeutic targets in a wide spectrum of solid tumours. J Pathol. 2014;232(1):4–15. .
    1. Parker BC, Zhang W. Fusion genes in solid tumors: an emerging target for cancer diagnosis and treatment. Chinese journal of cancer. 2013;32(11):594–603. 10.5732/cjc.013.10178
    1. Stransky N, Cerami E, Schalm S, Kim JL, Lengauer C. The landscape of kinase fusions in cancer. Nature communications. 2014;5:4846 10.1038/ncomms5846
    1. Dienstmann R, Rodon J, Prat A, Perez-Garcia J, Adamo B, Felip E, et al. Genomic aberrations in the FGFR pathway: opportunities for targeted therapies in solid tumors. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. 2014;25(3):552–63. 10.1093/annonc/mdt419 .
    1. Sia D, Losic B, Moeini A, Cabellos L, Hao K, Revill K, et al. Massive parallel sequencing uncovers actionable FGFR2-PPHLN1 fusion and ARAF mutations in intrahepatic cholangiocarcinoma. Nature communications. 2015;6:6087 10.1038/ncomms7087 .
    1. Nakanishi Y, Akiyama N, Tsukaguchi T, Fujii T, Satoh Y, Ishii N, et al. Mechanism of Oncogenic Signal Activation by the Novel Fusion Kinase FGFR3-BAIAP2L1. Molecular cancer therapeutics. 2015;14(3):704–12. 10.1158/1535-7163.MCT-14-0927-T .
    1. Churi CR, Shroff R, Wang Y, Rashid A, Kang HC, Weatherly J, et al. Mutation profiling in cholangiocarcinoma: prognostic and therapeutic implications. PloS one. 2014;9(12):e115383 10.1371/journal.pone.0115383
    1. Ross JS, Wang K, Gay L, Al-Rohil R, Rand JV, Jones DM, et al. New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing. Oncologist. 2014;19(3):235–42. 10.1634/theoncologist.2013-0352
    1. Zheng Z, Liebers M, Zhelyazkova B, Cao Y, Panditi D, Lynch KD, et al. Anchored multiplex PCR for targeted next-generation sequencing. Nat Med. 2014;20(12):1479–84. 10.1038/nm.3729 .
    1. Nakamura H, Arai Y, Totoki Y, Shirota T, Elzawahry A, Kato M, et al. Genomic spectra of biliary tract cancer. Nat Genet. 2015. 10.1038/ng.3375 .
    1. Blechacz B, Komuta M, Roskams T, Gores GJ. Clinical diagnosis and staging of cholangiocarcinoma. Nat Rev Gastroenterol Hepatol. 2011;8(9):512–22. 10.1038/nrgastro.2011.131
    1. Arai Y, Totoki Y, Hosoda F, Shirota T, Hama N, Nakamura H, et al. Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma. Hepatology. 2014;59(4):1427–34. 10.1002/hep.26890 .
    1. Eathiraj S, Palma R, Hirschi M, Volckova E, Nakuci E, Castro J, et al. A novel mode of protein kinase inhibition exploiting hydrophobic motifs of autoinhibited kinases: discovery of ATP-independent inhibitors of fibroblast growth factor receptor. The Journal of biological chemistry. 2011;286(23):20677–87. 10.1074/jbc.M110.213736
    1. Cha S. Tight-binding inhibitors-I. Kinetic behavior. Biochemical pharmacology. 1975;24(23):2177–85. .
    1. Morrison JF. Kinetics of the reversible inhibition of enzyme-catalysed reactions by tight-binding inhibitors. Biochimica et biophysica acta. 1969;185(2):269–86. .
    1. Syed M. Ali, Mark A. Ashwell, Sudharshan Eathiraj, Eugene Kelleher, Jean-Marc Lapierre, Yanbin Liu, Nivedita Namdev, Rocio Palma, Manish Tandon, David Vensel, Neil Westlund, Hui Wu, Rui-Yang Yang; Substituted 5,6-dihydro-6-phenylbenzo[F]isoquinolin-2-amine compounds; US Patent Application 12/649,573, 2010.
    1. Lew ED, Furdui CM, Anderson KS, Schlessinger J. The precise sequence of FGF receptor autophosphorylation is kinetically driven and is disrupted by oncogenic mutations. Sci Signal. 2009;2(58):ra6 10.1126/scisignal.2000021
    1. Furdui CM, Lew ED, Schlessinger J, Anderson KS. Autophosphorylation of FGFR1 kinase is mediated by a sequential and precisely ordered reaction. Mol Cell. 2006;21(5):711–7. 10.1016/j.molcel.2006.01.022 .
    1. Liu Y, Gray NS. Rational design of inhibitors that bind to inactive kinase conformations. Nat Chem Biol. 2006;2(7):358–64. 10.1038/nchembio799 .
    1. Turner N, Lambros MB, Horlings HM, Pearson A, Sharpe R, Natrajan R, et al. Integrative molecular profiling of triple negative breast cancers identifies amplicon drivers and potential therapeutic targets. Oncogene. 2010;29(14):2013–23. 10.1038/onc.2009.489
    1. Guagnano V, Kauffmann A, Wohrle S, Stamm C, Ito M, Barys L, et al. FGFR genetic alterations predict for sensitivity to NVP-BGJ398, a selective pan-FGFR inhibitor. Cancer discovery. 2012;2(12):1118–33. 10.1158/-12-0210 .
    1. Mathur A, Ware C, Davis L, Gazdar A, Pan BS, Lutterbach B. FGFR2 is amplified in the NCI-H716 colorectal cancer cell line and is required for growth and survival. PloS one. 2014;9(6):e98515 10.1371/journal.pone.0098515
    1. Klijn C, Durinck S, Stawiski EW, Haverty PM, Jiang Z, Liu H, et al. A comprehensive transcriptional portrait of human cancer cell lines. Nat Biotechnol. 2015;33(3):306–12. 10.1038/nbt.3080 .
    1. Chase A, Grand FH, Cross NC. Activity of TKI258 against primary cells and cell lines with FGFR1 fusion genes associated with the 8p11 myeloproliferative syndrome. Blood. 2007;110(10):3729–34. 10.1182/blood-2007-02-074286 .
    1. Zaid TM, Yeung TL, Thompson MS, Leung CS, Harding T, Co NN, et al. Identification of FGFR4 as a potential therapeutic target for advanced-stage, high-grade serous ovarian cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2013;19(4):809–20. 10.1158/1078-0432.CCR-12-2736
    1. Wu YM, Su F, Kalyana-Sundaram S, Khazanov N, Ateeq B, Cao X, et al. Identification of targetable FGFR gene fusions in diverse cancers. Cancer discovery. 2013;3(6):636–47. 10.1158/-13-0050
    1. Armstrong E, Vainikka S, Partanen J, Korhonen J, Alitalo R. Expression of fibroblast growth factor receptors in human leukemia cells. Cancer research. 1992;52(7):2004–7. .
    1. Dutt A, Salvesen HB, Chen TH, Ramos AH, Onofrio RC, Hatton C, et al. Drug-sensitive FGFR2 mutations in endometrial carcinoma. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(25):8713–7. 10.1073/pnas.0803379105
    1. Gavine PR, Mooney L, Kilgour E, Thomas AP, Al-Kadhimi K, Beck S, et al. AZD4547: an orally bioavailable, potent, and selective inhibitor of the fibroblast growth factor receptor tyrosine kinase family. Cancer research. 2012;72(8):2045–56. 10.1158/0008-5472.CAN-11-3034 .
    1. Konecny GE, Finkler N, Garcia AA, Lorusso D, Lee PS, Rocconi RP, et al. Second-line dovitinib (TKI258) in patients with FGFR2-mutated or FGFR2-non-mutated advanced or metastatic endometrial cancer: a non-randomised, open-label, two-group, two-stage, phase 2 study. Lancet Oncol. 2015;16(6):686–94. 10.1016/S1470-2045(15)70159-2 .
    1. Lewin J, Siu LL. Development of Fibroblast Growth Factor Receptor Inhibitors: Kissing Frogs to Find a Prince? Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2015. 10.1200/JCO.2015.62.7380 .
    1. Motzer RJ, Porta C, Vogelzang NJ, Sternberg CN, Szczylik C, Zolnierek J, et al. Dovitinib versus sorafenib for third-line targeted treatment of patients with metastatic renal cell carcinoma: an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15(3):286–96. 10.1016/S1470-2045(14)70030-0 .
    1. Tabernero J, Bahleda R, Dienstmann R, Infante JR, Mita A, Italiano A, et al. Phase I Dose-Escalation Study of JNJ-42756493, an Oral Pan-Fibroblast Growth Factor Receptor Inhibitor, in Patients With Advanced Solid Tumors. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2015. 10.1200/JCO.2014.60.7341 .
    1. Rizvi S, Borad MJ, Patel T, Gores GJ. Cholangiocarcinoma: molecular pathways and therapeutic opportunities. Semin Liver Dis. 2014;34(4):456–64. 10.1055/s-0034-1394144
    1. Papadopoulos KP, Tolcher AW, Patnaik A, Rasco DW, Chambers G, Beeram M, et al. Phase 1, first-in-human study of ARQ 087, an oral pan-Fibroblast Growth Factor Receptor (FGFR) inhibitor, in patients (pts) with advanced solid tumors. ASCO Meeting Abstracts. 2015;33(15_suppl):2545.

Source: PubMed

3
Sottoscrivi