Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Cholangiocarcinoma | Review

Fibroblast growth factor receptor fusions in cancer: opportunities and challenges

Authors: Lingfeng Chen, Yanmei Zhang, Lina Yin, Binhao Cai, Ping Huang, Xiaokun Li, Guang Liang

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Fibroblast growth factors (FGFs) and their receptors (FGFRs) play critical roles in many biological processes and developmental functions. Chromosomal translocation of FGFRs result in the formation of chimeric FGFR fusion proteins, which often cause aberrant signaling leading to the development and progression of human cancer. Due to the high recurrence rate and carcinogenicity, oncogenic FGFR gene fusions have been identified as promising therapeutic targets. Erdafitinib and pemigatinib, two FGFR selective inhibitors targeting FGFR fusions, have been approved by the U.S. Food and Drug Administration (FDA) to treat patients with urothelial cancer and cholangiocarcinoma, respectively. Futibatinib, a third-generation FGFR inhibitor, is under phase III clinical trials in patients with FGFR gene rearrangements. Herein, we review the current understanding of the FGF/FGFRs system and the oncogenic effect of FGFR fusions, summarize promising inhibitors under clinical development for patients with FGFR fusions, and highlight the challenges in this field.
Literature
1.
go back to reference Mertens F, Johansson B, Fioretos T, Mitelman F. The emerging complexity of gene fusions in cancer. Nat Rev Cancer. 2015;15:371–81.PubMedCrossRef Mertens F, Johansson B, Fioretos T, Mitelman F. The emerging complexity of gene fusions in cancer. Nat Rev Cancer. 2015;15:371–81.PubMedCrossRef
2.
3.
go back to reference De Luca A, Esposito Abate R, Rachiglio AM, Maiello MR, Esposito C, Schettino C, et al. FGFR fusions in cancer: from diagnostic approaches to therapeutic intervention. Int J Mol Sci. 2020;21(18):6856.PubMedCentralCrossRef De Luca A, Esposito Abate R, Rachiglio AM, Maiello MR, Esposito C, Schettino C, et al. FGFR fusions in cancer: from diagnostic approaches to therapeutic intervention. Int J Mol Sci. 2020;21(18):6856.PubMedCentralCrossRef
4.
go back to reference 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 Discov. 2013;3:636–47.PubMedPubMedCentralCrossRef 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 Discov. 2013;3:636–47.PubMedPubMedCentralCrossRef
5.
go back to reference Hochhaus A, Larson RA, Guilhot F, Radich JP, Branford S, Hughes TP, et al. Long-term outcomes of Imatinib treatment for chronic myeloid leukemia. N Engl J Med. 2017;376:917–27.PubMedPubMedCentralCrossRef Hochhaus A, Larson RA, Guilhot F, Radich JP, Branford S, Hughes TP, et al. Long-term outcomes of Imatinib treatment for chronic myeloid leukemia. N Engl J Med. 2017;376:917–27.PubMedPubMedCentralCrossRef
7.
go back to reference Drilon A, Siena S, Dziadziuszko R, Barlesi F, Krebs MG, Shaw AT, et al. Entrectinib in ROS1 fusion-positive non-small-cell lung cancer: integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020;21:261–70.PubMedCrossRef Drilon A, Siena S, Dziadziuszko R, Barlesi F, Krebs MG, Shaw AT, et al. Entrectinib in ROS1 fusion-positive non-small-cell lung cancer: integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020;21:261–70.PubMedCrossRef
8.
go back to reference Subbiah V, Velcheti V, Tuch BB, Ebata K, Busaidy NL, Cabanillas ME, et al. Selective RET kinase inhibition for patients with RET-altered cancers. Ann Oncol. 2018;29:1869–76.PubMedPubMedCentralCrossRef Subbiah V, Velcheti V, Tuch BB, Ebata K, Busaidy NL, Cabanillas ME, et al. Selective RET kinase inhibition for patients with RET-altered cancers. Ann Oncol. 2018;29:1869–76.PubMedPubMedCentralCrossRef
9.
go back to reference Guo R, Luo J, Chang J, Rekhtman N, Arcila M, Drilon A. MET-dependent solid tumours - molecular diagnosis and targeted therapy. Nat Rev Clin Oncol. 2020;17:569–87.PubMedPubMedCentralCrossRef Guo R, Luo J, Chang J, Rekhtman N, Arcila M, Drilon A. MET-dependent solid tumours - molecular diagnosis and targeted therapy. Nat Rev Clin Oncol. 2020;17:569–87.PubMedPubMedCentralCrossRef
10.
go back to reference Marcus L, Donoghue M, Aungst S, Myers CE, Helms WS, Shen G, et al. FDA approval summary: entrectinib for the treatment of NTRK gene fusion solid tumors. Clin Cancer Res. 2021;27:928–32.PubMedCrossRef Marcus L, Donoghue M, Aungst S, Myers CE, Helms WS, Shen G, et al. FDA approval summary: entrectinib for the treatment of NTRK gene fusion solid tumors. Clin Cancer Res. 2021;27:928–32.PubMedCrossRef
12.
go back to reference Katoh M, Nakagama H. FGF receptors: cancer biology and therapeutics. Med Res Rev. 2014;34:280–300.PubMedCrossRef Katoh M, Nakagama H. FGF receptors: cancer biology and therapeutics. Med Res Rev. 2014;34:280–300.PubMedCrossRef
13.
go back to reference Babina IS, Turner NC. Advances and challenges in targeting FGFR signalling in cancer. Nat Rev Cancer. 2017;17:318–32.PubMedCrossRef Babina IS, Turner NC. Advances and challenges in targeting FGFR signalling in cancer. Nat Rev Cancer. 2017;17:318–32.PubMedCrossRef
14.
go back to reference Katoh M. Therapeutics targeting FGF signaling network in human diseases. Trends Pharmacol Sci. 2016;37:1081–96.PubMedCrossRef Katoh M. Therapeutics targeting FGF signaling network in human diseases. Trends Pharmacol Sci. 2016;37:1081–96.PubMedCrossRef
15.
go back to reference Guasch G, Delaval B, Arnoulet C, Xie MJ, Xerri L, Sainty D, et al. FOP-FGFR1 tyrosine kinase, the product of a t(6;8) translocation, induces a fatal myeloproliferative disease in mice. Blood. 2004;103:309–12.PubMedCrossRef Guasch G, Delaval B, Arnoulet C, Xie MJ, Xerri L, Sainty D, et al. FOP-FGFR1 tyrosine kinase, the product of a t(6;8) translocation, induces a fatal myeloproliferative disease in mice. Blood. 2004;103:309–12.PubMedCrossRef
16.
go back to reference Nelson KN, Meyer AN, Siari A, Campos AR, Motamedchaboki K, Donoghue DJ. Oncogenic gene fusion FGFR3-TACC3 is regulated by tyrosine phosphorylation. Mol Cancer Res. 2016;14:458–69.PubMedCrossRef Nelson KN, Meyer AN, Siari A, Campos AR, Motamedchaboki K, Donoghue DJ. Oncogenic gene fusion FGFR3-TACC3 is regulated by tyrosine phosphorylation. Mol Cancer Res. 2016;14:458–69.PubMedCrossRef
17.
18.
go back to reference Helsten T, Elkin S, Arthur E, Tomson BN, Carter J, Kurzrock R. The FGFR landscape in cancer: analysis of 4,853 tumors by next-generation sequencing. Clin Cancer Res. 2016;22:259–67.PubMedCrossRef Helsten T, Elkin S, Arthur E, Tomson BN, Carter J, Kurzrock R. The FGFR landscape in cancer: analysis of 4,853 tumors by next-generation sequencing. Clin Cancer Res. 2016;22:259–67.PubMedCrossRef
19.
go back to reference Xie Y, Zinkle A, Chen L, Mohammadi M. Fibroblast growth factor signalling in osteoarthritis and cartilage repair. Nat Rev Rheumatol. 2020;16:547–64.PubMedCrossRef Xie Y, Zinkle A, Chen L, Mohammadi M. Fibroblast growth factor signalling in osteoarthritis and cartilage repair. Nat Rev Rheumatol. 2020;16:547–64.PubMedCrossRef
20.
go back to reference Belov AA, Mohammadi M. Molecular mechanisms of fibroblast growth factor signaling in physiology and pathology. Cold Spring Harb Perspect Biol. 2013;5(6):a015958.PubMedPubMedCentralCrossRef Belov AA, Mohammadi M. Molecular mechanisms of fibroblast growth factor signaling in physiology and pathology. Cold Spring Harb Perspect Biol. 2013;5(6):a015958.PubMedPubMedCentralCrossRef
21.
go back to reference Kalinina J, Dutta K, Ilghari D, Beenken A, Goetz R, Eliseenkova AV, et al. The alternatively spliced acid box region plays a key role in FGF receptor autoinhibition. Structure. 2012;20:77–88.PubMedPubMedCentralCrossRef Kalinina J, Dutta K, Ilghari D, Beenken A, Goetz R, Eliseenkova AV, et al. The alternatively spliced acid box region plays a key role in FGF receptor autoinhibition. Structure. 2012;20:77–88.PubMedPubMedCentralCrossRef
22.
go back to reference Yeh BK, Igarashi M, Eliseenkova AV, Plotnikov AN, Sher I, Ron D, et al. Structural basis by which alternative splicing confers specificity in fibroblast growth factor receptors. Proc Natl Acad Sci U S A. 2003;100:2266–71.PubMedPubMedCentralCrossRef Yeh BK, Igarashi M, Eliseenkova AV, Plotnikov AN, Sher I, Ron D, et al. Structural basis by which alternative splicing confers specificity in fibroblast growth factor receptors. Proc Natl Acad Sci U S A. 2003;100:2266–71.PubMedPubMedCentralCrossRef
23.
go back to reference Zhang X, Ibrahimi OA, Olsen SK, Umemori H, Mohammadi M, Ornitz DM. Receptor specificity of the fibroblast growth factor family. The complete mammalian FGF family. J Biol Chem. 2006;281:15694–700.PubMedCrossRef Zhang X, Ibrahimi OA, Olsen SK, Umemori H, Mohammadi M, Ornitz DM. Receptor specificity of the fibroblast growth factor family. The complete mammalian FGF family. J Biol Chem. 2006;281:15694–700.PubMedCrossRef
24.
go back to reference Schlessinger J, Plotnikov AN, Ibrahimi OA, Eliseenkova AV, Yeh BK, Yayon A, et al. Crystal structure of a ternary FGF-FGFR-heparin complex reveals a dual role for heparin in FGFR binding and dimerization. Mol Cell. 2000;6:743–50.PubMedCrossRef Schlessinger J, Plotnikov AN, Ibrahimi OA, Eliseenkova AV, Yeh BK, Yayon A, et al. Crystal structure of a ternary FGF-FGFR-heparin complex reveals a dual role for heparin in FGFR binding and dimerization. Mol Cell. 2000;6:743–50.PubMedCrossRef
25.
go back to reference Chen G, Liu Y, Goetz R, Fu L, Jayaraman S, Hu MC, et al. alpha-Klotho is a non-enzymatic molecular scaffold for FGF23 hormone signalling. Nature. 2018;553:461–6.PubMedPubMedCentralCrossRef Chen G, Liu Y, Goetz R, Fu L, Jayaraman S, Hu MC, et al. alpha-Klotho is a non-enzymatic molecular scaffold for FGF23 hormone signalling. Nature. 2018;553:461–6.PubMedPubMedCentralCrossRef
26.
go back to reference Lee S, Choi J, Mohanty J, Sousa LP, Tome F, Pardon E, et al. Structures of beta-klotho reveal a ‘zip code’-like mechanism for endocrine FGF signalling. Nature. 2018;553:501–5.PubMedPubMedCentralCrossRef Lee S, Choi J, Mohanty J, Sousa LP, Tome F, Pardon E, et al. Structures of beta-klotho reveal a ‘zip code’-like mechanism for endocrine FGF signalling. Nature. 2018;553:501–5.PubMedPubMedCentralCrossRef
27.
go back to reference Eswarakumar VP, Lax I, Schlessinger J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev. 2005;16:139–49.PubMedCrossRef Eswarakumar VP, Lax I, Schlessinger J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev. 2005;16:139–49.PubMedCrossRef
28.
go back to reference Dhalluin C, Yan KS, Plotnikova O, Lee KW, Zeng L, Kuti M, et al. Structural basis of SNT PTB domain interactions with distinct neurotrophic receptors. Mol Cell. 2000;6:921–9.PubMedPubMedCentralCrossRef Dhalluin C, Yan KS, Plotnikova O, Lee KW, Zeng L, Kuti M, et al. Structural basis of SNT PTB domain interactions with distinct neurotrophic receptors. Mol Cell. 2000;6:921–9.PubMedPubMedCentralCrossRef
29.
go back to reference Ong SH, Guy GR, Hadari YR, Laks S, Gotoh N, Schlessinger J, et al. FRS2 proteins recruit intracellular signaling pathways by binding to diverse targets on fibroblast growth factor and nerve growth factor receptors. Mol Cell Biol. 2000;20:979–89.PubMedPubMedCentralCrossRef Ong SH, Guy GR, Hadari YR, Laks S, Gotoh N, Schlessinger J, et al. FRS2 proteins recruit intracellular signaling pathways by binding to diverse targets on fibroblast growth factor and nerve growth factor receptors. Mol Cell Biol. 2000;20:979–89.PubMedPubMedCentralCrossRef
30.
go back to reference Chardin P, Camonis JH, Gale NW, van Aelst L, Schlessinger J, Wigler MH, et al. Human Sos1: a guanine nucleotide exchange factor for Ras that binds to GRB2. Science. 1993;260:1338–43.PubMedCrossRef Chardin P, Camonis JH, Gale NW, van Aelst L, Schlessinger J, Wigler MH, et al. Human Sos1: a guanine nucleotide exchange factor for Ras that binds to GRB2. Science. 1993;260:1338–43.PubMedCrossRef
31.
go back to reference Touat M, Ileana E, Postel-Vinay S, Andre F, Soria JC. Targeting FGFR signaling in cancer. Clin Cancer Res. 2015;21:2684–94.PubMedCrossRef Touat M, Ileana E, Postel-Vinay S, Andre F, Soria JC. Targeting FGFR signaling in cancer. Clin Cancer Res. 2015;21:2684–94.PubMedCrossRef
32.
go back to reference Hanafusa H, Torii S, Yasunaga T, Matsumoto K, Nishida E. Shp2, an SH2-containing protein-tyrosine phosphatase, positively regulates receptor tyrosine kinase signaling by dephosphorylating and inactivating the inhibitor Sprouty. J Biol Chem. 2004;279:22992–5.PubMedCrossRef Hanafusa H, Torii S, Yasunaga T, Matsumoto K, Nishida E. Shp2, an SH2-containing protein-tyrosine phosphatase, positively regulates receptor tyrosine kinase signaling by dephosphorylating and inactivating the inhibitor Sprouty. J Biol Chem. 2004;279:22992–5.PubMedCrossRef
33.
go back to reference Nichols RJ, Haderk F, Stahlhut C, Schulze CJ, Hemmati G, Wildes D, et al. RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancers. Nat Cell Biol. 2018;20:1064–73.PubMedPubMedCentralCrossRef Nichols RJ, Haderk F, Stahlhut C, Schulze CJ, Hemmati G, Wildes D, et al. RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancers. Nat Cell Biol. 2018;20:1064–73.PubMedPubMedCentralCrossRef
34.
go back to reference Bunney TD, Esposito D, Mas-Droux C, Lamber E, Baxendale RW, Martins M, et al. Structural and functional integration of the PLCgamma interaction domains critical for regulatory mechanisms and signaling deregulation. Structure. 2012;20:2062–75.PubMedPubMedCentralCrossRef Bunney TD, Esposito D, Mas-Droux C, Lamber E, Baxendale RW, Martins M, et al. Structural and functional integration of the PLCgamma interaction domains critical for regulatory mechanisms and signaling deregulation. Structure. 2012;20:2062–75.PubMedPubMedCentralCrossRef
35.
go back to reference Huang Z, Marsiglia WM, Basu Roy U, Rahimi N, Ilghari D, Wang H, et al. Two FGF receptor kinase molecules act in concert to recruit and transphosphorylate phospholipase Cgamma. Mol Cell. 2016;61:98–110.PubMedCrossRef Huang Z, Marsiglia WM, Basu Roy U, Rahimi N, Ilghari D, Wang H, et al. Two FGF receptor kinase molecules act in concert to recruit and transphosphorylate phospholipase Cgamma. Mol Cell. 2016;61:98–110.PubMedCrossRef
36.
go back to reference Chen H, Marsiglia WM, Cho MK, Huang Z, Deng J, Blais SP, et al. Elucidation of a four-site allosteric network in fibroblast growth factor receptor tyrosine kinases. Elife. 2017;6:e21137.PubMedPubMedCentralCrossRef Chen H, Marsiglia WM, Cho MK, Huang Z, Deng J, Blais SP, et al. Elucidation of a four-site allosteric network in fibroblast growth factor receptor tyrosine kinases. Elife. 2017;6:e21137.PubMedPubMedCentralCrossRef
37.
38.
go back to reference Furdui CM, Lew ED, Schlessinger J, Anderson KS. Autophosphorylation of FGFR1 kinase is mediated by a sequential and precisely ordered reaction. Mol Cell. 2006;21:711–7.PubMedCrossRef Furdui CM, Lew ED, Schlessinger J, Anderson KS. Autophosphorylation of FGFR1 kinase is mediated by a sequential and precisely ordered reaction. Mol Cell. 2006;21:711–7.PubMedCrossRef
39.
go back to reference Bae JH, Boggon TJ, Tome F, Mandiyan V, Lax I, Schlessinger J. Asymmetric receptor contact is required for tyrosine autophosphorylation of fibroblast growth factor receptor in living cells. Proc Natl Acad Sci U S A. 2010;107:2866–71.PubMedPubMedCentralCrossRef Bae JH, Boggon TJ, Tome F, Mandiyan V, Lax I, Schlessinger J. Asymmetric receptor contact is required for tyrosine autophosphorylation of fibroblast growth factor receptor in living cells. Proc Natl Acad Sci U S A. 2010;107:2866–71.PubMedPubMedCentralCrossRef
40.
go back to reference Chen L, Marsiglia WM, Chen H, Katigbak J, Erdjument-Bromage H, Kemble DJ, et al. Molecular basis for receptor tyrosine kinase A-loop tyrosine transphosphorylation. Nat Chem Biol. 2020;16:267–77.PubMedPubMedCentralCrossRef Chen L, Marsiglia WM, Chen H, Katigbak J, Erdjument-Bromage H, Kemble DJ, et al. Molecular basis for receptor tyrosine kinase A-loop tyrosine transphosphorylation. Nat Chem Biol. 2020;16:267–77.PubMedPubMedCentralCrossRef
41.
go back to reference Chen H, Ma J, Li W, Eliseenkova AV, Xu C, Neubert TA, et al. A molecular brake in the kinase hinge region regulates the activity of receptor tyrosine kinases. Mol Cell. 2007;27:717–30.PubMedPubMedCentralCrossRef Chen H, Ma J, Li W, Eliseenkova AV, Xu C, Neubert TA, et al. A molecular brake in the kinase hinge region regulates the activity of receptor tyrosine kinases. Mol Cell. 2007;27:717–30.PubMedPubMedCentralCrossRef
42.
go back to reference Marsiglia WM, Katigbak J, Zheng S, Mohammadi M, Zhang Y, Traaseth NJ. A conserved allosteric pathway in tyrosine kinase regulation. Structure. 2019;27:1308–1315. e1303.PubMedPubMedCentralCrossRef Marsiglia WM, Katigbak J, Zheng S, Mohammadi M, Zhang Y, Traaseth NJ. A conserved allosteric pathway in tyrosine kinase regulation. Structure. 2019;27:1308–1315. e1303.PubMedPubMedCentralCrossRef
43.
go back to reference Olsen SK, Ibrahimi OA, Raucci A, Zhang F, Eliseenkova AV, Yayon A, et al. Insights into the molecular basis for fibroblast growth factor receptor autoinhibition and ligand-binding promiscuity. Proc Natl Acad Sci U S A. 2004;101:935–40.PubMedPubMedCentralCrossRef Olsen SK, Ibrahimi OA, Raucci A, Zhang F, Eliseenkova AV, Yayon A, et al. Insights into the molecular basis for fibroblast growth factor receptor autoinhibition and ligand-binding promiscuity. Proc Natl Acad Sci U S A. 2004;101:935–40.PubMedPubMedCentralCrossRef
44.
go back to reference Katoh M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat Rev Clin Oncol. 2019;16:105–22.PubMedCrossRef Katoh M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat Rev Clin Oncol. 2019;16:105–22.PubMedCrossRef
45.
go back to reference 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;232:4–15.PubMedCrossRef 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;232:4–15.PubMedCrossRef
46.
go back to reference Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005;310:644–8.PubMedCrossRef Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005;310:644–8.PubMedCrossRef
47.
go back to reference Tulpule A, Guan J, Neel DS, Allegakoen HR, Lin YP, Brown D, et al. Kinase-mediated RAS signaling via membraneless cytoplasmic protein granules. Cell. 2021;184(10):2649–64.PubMedPubMedCentralCrossRef Tulpule A, Guan J, Neel DS, Allegakoen HR, Lin YP, Brown D, et al. Kinase-mediated RAS signaling via membraneless cytoplasmic protein granules. Cell. 2021;184(10):2649–64.PubMedPubMedCentralCrossRef
48.
go back to reference Qin A, Johnson A, Ross JS, Miller VA, Ali SM, Schrock AB, et al. Detection of known and novel FGFR fusions in non-small cell lung cancer by comprehensive genomic profiling. J Thorac Oncol. 2019;14:54–62.PubMedCrossRef Qin A, Johnson A, Ross JS, Miller VA, Ali SM, Schrock AB, et al. Detection of known and novel FGFR fusions in non-small cell lung cancer by comprehensive genomic profiling. J Thorac Oncol. 2019;14:54–62.PubMedCrossRef
49.
go back to reference 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;19:235–42.PubMedPubMedCentralCrossRef 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;19:235–42.PubMedPubMedCentralCrossRef
50.
go back to reference 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. Nat Commun. 2015;6:6087.PubMedCrossRef 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. Nat Commun. 2015;6:6087.PubMedCrossRef
51.
go back to reference Kumar KR, Chen W, Koduru PR, Luu HS. Myeloid and lymphoid neoplasm with abnormalities of FGFR1 presenting with trilineage blasts and RUNX1 rearrangement: a case report and review of literature. Am J Clin Pathol. 2015;143:738–48.PubMedCrossRef Kumar KR, Chen W, Koduru PR, Luu HS. Myeloid and lymphoid neoplasm with abnormalities of FGFR1 presenting with trilineage blasts and RUNX1 rearrangement: a case report and review of literature. Am J Clin Pathol. 2015;143:738–48.PubMedCrossRef
52.
go back to reference Jackson CC, Medeiros LJ, Miranda RN. 8p11 myeloproliferative syndrome: a review. Hum Pathol. 2010;41:461–76.PubMedCrossRef Jackson CC, Medeiros LJ, Miranda RN. 8p11 myeloproliferative syndrome: a review. Hum Pathol. 2010;41:461–76.PubMedCrossRef
53.
go back to reference Xiao S, Nalabolu SR, Aster JC, Ma J, Abruzzo L, Jaffe ES, et al. FGFR1 is fused with a novel zinc-finger gene, ZNF198, in the t(8;13) leukaemia/lymphoma syndrome. Nat Genet. 1998;18:84–7.PubMedCrossRef Xiao S, Nalabolu SR, Aster JC, Ma J, Abruzzo L, Jaffe ES, et al. FGFR1 is fused with a novel zinc-finger gene, ZNF198, in the t(8;13) leukaemia/lymphoma syndrome. Nat Genet. 1998;18:84–7.PubMedCrossRef
54.
go back to reference Xiao S, McCarthy JG, Aster JC, Fletcher JA. ZNF198-FGFR1 transforming activity depends on a novel proline-rich ZNF198 oligomerization domain. Blood. 2000;96:699–704.PubMedCrossRef Xiao S, McCarthy JG, Aster JC, Fletcher JA. ZNF198-FGFR1 transforming activity depends on a novel proline-rich ZNF198 oligomerization domain. Blood. 2000;96:699–704.PubMedCrossRef
55.
go back to reference Peiris MN, Meyer AN, Nelson KN, Bisom-Rapp EW, Donoghue DJ. Oncogenic fusion protein BCR-FGFR1 requires the breakpoint cluster region-mediated oligomerization and chaperonin Hsp90 for activation. Haematologica. 2020;105:1262–73.PubMedPubMedCentralCrossRef Peiris MN, Meyer AN, Nelson KN, Bisom-Rapp EW, Donoghue DJ. Oncogenic fusion protein BCR-FGFR1 requires the breakpoint cluster region-mediated oligomerization and chaperonin Hsp90 for activation. Haematologica. 2020;105:1262–73.PubMedPubMedCentralCrossRef
56.
go back to reference Popovici C, Zhang B, Gregoire MJ, Jonveaux P, Lafage-Pochitaloff M, Birnbaum D, et al. The t(6;8)(q27;p11) translocation in a stem cell myeloproliferative disorder fuses a novel gene, FOP, to fibroblast growth factor receptor 1. Blood. 1999;93:1381–9.PubMedCrossRef Popovici C, Zhang B, Gregoire MJ, Jonveaux P, Lafage-Pochitaloff M, Birnbaum D, et al. The t(6;8)(q27;p11) translocation in a stem cell myeloproliferative disorder fuses a novel gene, FOP, to fibroblast growth factor receptor 1. Blood. 1999;93:1381–9.PubMedCrossRef
57.
go back to reference Ren M, Qin H, Kitamura E, Cowell JK. Dysregulated signaling pathways in the development of CNTRL-FGFR1-induced myeloid and lymphoid malignancies associated with FGFR1 in human and mouse models. Blood. 2013;122:1007–16.PubMedPubMedCentralCrossRef Ren M, Qin H, Kitamura E, Cowell JK. Dysregulated signaling pathways in the development of CNTRL-FGFR1-induced myeloid and lymphoid malignancies associated with FGFR1 in human and mouse models. Blood. 2013;122:1007–16.PubMedPubMedCentralCrossRef
59.
go back to reference 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;59:1427–34.PubMedCrossRef 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;59:1427–34.PubMedCrossRef
60.
go back to reference Graham RP, Barr Fritcher EG, Pestova E, Schulz J, Sitailo LA, Vasmatzis G, et al. Fibroblast growth factor receptor 2 translocations in intrahepatic cholangiocarcinoma. Hum Pathol. 2014;45:1630–8.PubMedCrossRef Graham RP, Barr Fritcher EG, Pestova E, Schulz J, Sitailo LA, Vasmatzis G, et al. Fibroblast growth factor receptor 2 translocations in intrahepatic cholangiocarcinoma. Hum Pathol. 2014;45:1630–8.PubMedCrossRef
61.
go back to reference Kazerounian S, Aho S. Characterization of periphilin, a widespread, highly insoluble nuclear protein and potential constituent of the keratinocyte cornified envelope. J Biol Chem. 2003;278:36707–17.PubMedCrossRef Kazerounian S, Aho S. Characterization of periphilin, a widespread, highly insoluble nuclear protein and potential constituent of the keratinocyte cornified envelope. J Biol Chem. 2003;278:36707–17.PubMedCrossRef
62.
go back to reference Wang Y, Ding X, Wang S, Moser CD, Shaleh HM, Mohamed EA, 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;380:163–73.PubMedPubMedCentralCrossRef Wang Y, Ding X, Wang S, Moser CD, Shaleh HM, Mohamed EA, 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;380:163–73.PubMedPubMedCentralCrossRef
63.
go back to reference Ying X, Tu J, Wang W, Li X, Xu C, Ji J. FGFR2-BICC1: a subtype of FGFR2 oncogenic fusion variant in cholangiocarcinoma and the response to Sorafenib. Onco Targets Ther. 2019;12:9303–7.PubMedPubMedCentralCrossRef Ying X, Tu J, Wang W, Li X, Xu C, Ji J. FGFR2-BICC1: a subtype of FGFR2 oncogenic fusion variant in cholangiocarcinoma and the response to Sorafenib. Onco Targets Ther. 2019;12:9303–7.PubMedPubMedCentralCrossRef
64.
go back to reference Glaser AP, Fantini D, Shilatifard A, Schaeffer EM, Meeks JJ. The evolving genomic landscape of urothelial carcinoma. Nat Rev Urol. 2017;14:215–29.PubMedCrossRef Glaser AP, Fantini D, Shilatifard A, Schaeffer EM, Meeks JJ. The evolving genomic landscape of urothelial carcinoma. Nat Rev Urol. 2017;14:215–29.PubMedCrossRef
65.
go back to reference Singh D, Chan JM, Zoppoli P, Niola F, Sullivan R, Castano A, et al. Transforming fusions of FGFR and TACC genes in human glioblastoma. Science. 2012;337:1231–5.PubMedPubMedCentralCrossRef Singh D, Chan JM, Zoppoli P, Niola F, Sullivan R, Castano A, et al. Transforming fusions of FGFR and TACC genes in human glioblastoma. Science. 2012;337:1231–5.PubMedPubMedCentralCrossRef
66.
go back to reference Williams SV, Hurst CD, Knowles MA. Oncogenic FGFR3 gene fusions in bladder cancer. Hum Mol Genet. 2013;22:795–803.PubMedCrossRef Williams SV, Hurst CD, Knowles MA. Oncogenic FGFR3 gene fusions in bladder cancer. Hum Mol Genet. 2013;22:795–803.PubMedCrossRef
67.
go back to reference 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. J Biol Chem. 2011;286:20677–87.PubMedPubMedCentralCrossRef 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. J Biol Chem. 2011;286:20677–87.PubMedPubMedCentralCrossRef
68.
go back to reference Hall TG, Yu Y, Eathiraj S, Wang Y, Savage RE, Lapierre JM, et al. Preclinical activity of ARQ 087, a novel inhibitor targeting FGFR dysregulation. PLoS One. 2016;11:e0162594.PubMedPubMedCentralCrossRef Hall TG, Yu Y, Eathiraj S, Wang Y, Savage RE, Lapierre JM, et al. Preclinical activity of ARQ 087, a novel inhibitor targeting FGFR dysregulation. PLoS One. 2016;11:e0162594.PubMedPubMedCentralCrossRef
69.
go back to reference Mazzaferro V, El-Rayes BF, Droz Dit Busset M, Cotsoglou C, Harris WP, Damjanov N, et al. Derazantinib (ARQ 087) in advanced or inoperable FGFR2 gene fusion-positive intrahepatic cholangiocarcinoma. Br J Cancer. 2019;120:165–71.PubMedCrossRef Mazzaferro V, El-Rayes BF, Droz Dit Busset M, Cotsoglou C, Harris WP, Damjanov N, et al. Derazantinib (ARQ 087) in advanced or inoperable FGFR2 gene fusion-positive intrahepatic cholangiocarcinoma. Br J Cancer. 2019;120:165–71.PubMedCrossRef
70.
go back to reference Papadopoulos KP, El-Rayes BF, Tolcher AW, Patnaik A, Rasco DW, Harvey RD, et al. A phase 1 study of ARQ 087, an oral pan-FGFR inhibitor in patients with advanced solid tumours. Br J Cancer. 2017;117:1592–9.PubMedPubMedCentralCrossRef Papadopoulos KP, El-Rayes BF, Tolcher AW, Patnaik A, Rasco DW, Harvey RD, et al. A phase 1 study of ARQ 087, an oral pan-FGFR inhibitor in patients with advanced solid tumours. Br J Cancer. 2017;117:1592–9.PubMedPubMedCentralCrossRef
71.
go back to reference Soria JC, DeBraud F, Bahleda R, Adamo B, Andre F, Dientsmann R, et al. Phase I/IIa study evaluating the safety, efficacy, pharmacokinetics, and pharmacodynamics of lucitanib in advanced solid tumors. Ann Oncol. 2014;25:2244–51.PubMedCrossRef Soria JC, DeBraud F, Bahleda R, Adamo B, Andre F, Dientsmann R, et al. Phase I/IIa study evaluating the safety, efficacy, pharmacokinetics, and pharmacodynamics of lucitanib in advanced solid tumors. Ann Oncol. 2014;25:2244–51.PubMedCrossRef
72.
go back to reference Hahn NM, Bivalacqua TJ, Ross AE, Netto GJ, Baras A, Park JC, et al. A phase II trial of Dovitinib in BCG-unresponsive urothelial carcinoma with FGFR3 mutations or overexpression: Hoosier Cancer Research Network trial HCRN 12-157. Clin Cancer Res. 2017;23:3003–11.PubMedCrossRef Hahn NM, Bivalacqua TJ, Ross AE, Netto GJ, Baras A, Park JC, et al. A phase II trial of Dovitinib in BCG-unresponsive urothelial carcinoma with FGFR3 mutations or overexpression: Hoosier Cancer Research Network trial HCRN 12-157. Clin Cancer Res. 2017;23:3003–11.PubMedCrossRef
73.
go back to reference Loriot Y, Necchi A, Park SH, Garcia-Donas J, Huddart R, Burgess E, et al. Erdafitinib in locally advanced or metastatic urothelial carcinoma. N Engl J Med. 2019;381:338–48.PubMedCrossRef Loriot Y, Necchi A, Park SH, Garcia-Donas J, Huddart R, Burgess E, et al. Erdafitinib in locally advanced or metastatic urothelial carcinoma. N Engl J Med. 2019;381:338–48.PubMedCrossRef
74.
go back to reference Patani H, Bunney TD, Thiyagarajan N, Norman RA, Ogg D, Breed J, et al. Landscape of activating cancer mutations in FGFR kinases and their differential responses to inhibitors in clinical use. Oncotarget. 2016;7:24252–68.PubMedPubMedCentralCrossRef Patani H, Bunney TD, Thiyagarajan N, Norman RA, Ogg D, Breed J, et al. Landscape of activating cancer mutations in FGFR kinases and their differential responses to inhibitors in clinical use. Oncotarget. 2016;7:24252–68.PubMedPubMedCentralCrossRef
75.
go back to reference Abou-Alfa GK, Sahai V, Hollebecque A, Vaccaro G, Melisi D, Al-Rajabi R, et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol. 2020;21:671–84.PubMedPubMedCentralCrossRef Abou-Alfa GK, Sahai V, Hollebecque A, Vaccaro G, Melisi D, Al-Rajabi R, et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol. 2020;21:671–84.PubMedPubMedCentralCrossRef
76.
77.
go back to reference Javle M, Lowery M, Shroff RT, Weiss KH, Springfeld C, Borad MJ, et al. Phase II study of BGJ398 in patients with FGFR-altered advanced cholangiocarcinoma. J Clin Oncol. 2018;36:276–82.PubMedCrossRef Javle M, Lowery M, Shroff RT, Weiss KH, Springfeld C, Borad MJ, et al. Phase II study of BGJ398 in patients with FGFR-altered advanced cholangiocarcinoma. J Clin Oncol. 2018;36:276–82.PubMedCrossRef
78.
go back to reference Van Cutsem E, Bang YJ, Mansoor W, Petty RD, Chao Y, Cunningham D, et al. A randomized, open-label study of the efficacy and safety of AZD4547 monotherapy versus paclitaxel for the treatment of advanced gastric adenocarcinoma with FGFR2 polysomy or gene amplification. Ann Oncol. 2017;28:1316–24.PubMedCrossRef Van Cutsem E, Bang YJ, Mansoor W, Petty RD, Chao Y, Cunningham D, et al. A randomized, open-label study of the efficacy and safety of AZD4547 monotherapy versus paclitaxel for the treatment of advanced gastric adenocarcinoma with FGFR2 polysomy or gene amplification. Ann Oncol. 2017;28:1316–24.PubMedCrossRef
79.
go back to reference 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. Mol Cancer Ther. 2014;13:2547–58.PubMedCrossRef 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. Mol Cancer Ther. 2014;13:2547–58.PubMedCrossRef
80.
go back to reference Goyal L, Saha SK, Liu LY, Siravegna G, Leshchiner I, Ahronian LG, et al. Polyclonal secondary FGFR2 mutations drive acquired resistance to FGFR inhibition in patients with FGFR2 fusion-positive cholangiocarcinoma. Cancer Discov. 2017;7:252–63.PubMedCrossRef Goyal L, Saha SK, Liu LY, Siravegna G, Leshchiner I, Ahronian LG, et al. Polyclonal secondary FGFR2 mutations drive acquired resistance to FGFR inhibition in patients with FGFR2 fusion-positive cholangiocarcinoma. Cancer Discov. 2017;7:252–63.PubMedCrossRef
81.
82.
go back to reference Goyal L, Shi L, Liu LY, Fece de la Cruz F, Lennerz JK, Raghavan S, et al. TAS-120 overcomes resistance to ATP-competitive FGFR inhibitors in patients with FGFR2 fusion-positive intrahepatic cholangiocarcinoma. Cancer Discov. 2019;9:1064–79.PubMedPubMedCentralCrossRef Goyal L, Shi L, Liu LY, Fece de la Cruz F, Lennerz JK, Raghavan S, et al. TAS-120 overcomes resistance to ATP-competitive FGFR inhibitors in patients with FGFR2 fusion-positive intrahepatic cholangiocarcinoma. Cancer Discov. 2019;9:1064–79.PubMedPubMedCentralCrossRef
83.
go back to reference Zhao Z, Bourne PE. Progress with covalent small-molecule kinase inhibitors. Drug Discov Today. 2018;23:727–35.PubMedCrossRef Zhao Z, Bourne PE. Progress with covalent small-molecule kinase inhibitors. Drug Discov Today. 2018;23:727–35.PubMedCrossRef
84.
go back to reference Chen L, Fu W, Zheng L, Liu Z, Liang G. Recent progress of small-molecule epidermal growth factor receptor (EGFR) inhibitors against C797S resistance in non-small-cell lung cancer. J Med Chem. 2018;61:4290–300.PubMedCrossRef Chen L, Fu W, Zheng L, Liu Z, Liang G. Recent progress of small-molecule epidermal growth factor receptor (EGFR) inhibitors against C797S resistance in non-small-cell lung cancer. J Med Chem. 2018;61:4290–300.PubMedCrossRef
85.
go back to reference Woyach JA, Furman RR, Liu TM, Ozer HG, Zapatka M, Ruppert AS, et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014;370:2286–94.PubMedPubMedCentralCrossRef Woyach JA, Furman RR, Liu TM, Ozer HG, Zapatka M, Ruppert AS, et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014;370:2286–94.PubMedPubMedCentralCrossRef
87.
go back to reference Du G, Jiang J, Wu Q, Henning NJ, Donovan KA, Yue H, et al. Discovery of a potent degrader for fibroblast growth factor receptor 1/2. Angew Chem Int Ed Engl. 2021;60:15905–11.PubMedCrossRefPubMedCentral Du G, Jiang J, Wu Q, Henning NJ, Donovan KA, Yue H, et al. Discovery of a potent degrader for fibroblast growth factor receptor 1/2. Angew Chem Int Ed Engl. 2021;60:15905–11.PubMedCrossRefPubMedCentral
88.
go back to reference Krook MA, Reeser JW, Ernst G, Barker H, Wilberding M, Li G, et al. Fibroblast growth factor receptors in cancer: genetic alterations, diagnostics, therapeutic targets and mechanisms of resistance. Br J Cancer. 2021;124:880–92.PubMedCrossRef Krook MA, Reeser JW, Ernst G, Barker H, Wilberding M, Li G, et al. Fibroblast growth factor receptors in cancer: genetic alterations, diagnostics, therapeutic targets and mechanisms of resistance. Br J Cancer. 2021;124:880–92.PubMedCrossRef
89.
90.
go back to reference Campo L, Breuer EK. Inhibition of TACC3 by a small molecule inhibitor in breast cancer. Biochem Biophys Res Commun. 2018;498:1085–92.PubMedCrossRef Campo L, Breuer EK. Inhibition of TACC3 by a small molecule inhibitor in breast cancer. Biochem Biophys Res Commun. 2018;498:1085–92.PubMedCrossRef
91.
go back to reference Akbulut O, Lengerli D, Saatci O, Duman E, Seker UOS, Isik A, et al. A highly potent TACC3 inhibitor as a novel anticancer drug candidate. Mol Cancer Ther. 2020;19:1243–54.PubMedCrossRef Akbulut O, Lengerli D, Saatci O, Duman E, Seker UOS, Isik A, et al. A highly potent TACC3 inhibitor as a novel anticancer drug candidate. Mol Cancer Ther. 2020;19:1243–54.PubMedCrossRef
92.
93.
go back to reference Yanochko GM, Vitsky A, Heyen JR, Hirakawa B, Lam JL, May J, et al. Pan-FGFR inhibition leads to blockade of FGF23 signaling, soft tissue mineralization, and cardiovascular dysfunction. Toxicol Sci. 2013;135:451–64.PubMedCrossRef Yanochko GM, Vitsky A, Heyen JR, Hirakawa B, Lam JL, May J, et al. Pan-FGFR inhibition leads to blockade of FGF23 signaling, soft tissue mineralization, and cardiovascular dysfunction. Toxicol Sci. 2013;135:451–64.PubMedCrossRef
Metadata
Title
Fibroblast growth factor receptor fusions in cancer: opportunities and challenges
Authors
Lingfeng Chen
Yanmei Zhang
Lina Yin
Binhao Cai
Ping Huang
Xiaokun Li
Guang Liang
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-02156-6

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine