Abstract
Purpose
Endocrine therapy resistance is the major challenge of managing patients with estrogen receptor positive (ER+) breast cancer. We previously reported frequent overexpression of FGFR4 in endocrine-resistant cell lines and breast cancers that recurred and metastasized following endocrine therapy, suggesting FGFR4 as a potential driver of endocrine resistance. In this study, we investigated the role of FGFR4 in mediating endocrine resistance and explored the therapeutic potential of targeting FGFR4 in advanced breast cancer.
Methods
A gene expression signature of FGFR4 activity was examined in ER+breast cancer pre- and post-neoadjuvant endocrine therapy and the association between FGFR4 expression and patient survival was examined. A correlation analysis was used to uncover potential regulators of FGFR4 overexpression. To investigate if FGFR4 is necessary to drive endocrine resistance, we tested response to FGFR4 inhibition in long-term estrogen-deprived (LTED) cells and their paired parental cells. Doxycycline inducible FGFR4 overexpression and knockdown cell models were generated to examine if FGFR4 was sufficient to confer endocrine resistance. Finally, we examined response to FGFR4 monotherapy or combination therapy with fulvestrant in breast cancer cell lines to explore the potential of FGFR4 targeted therapy for advanced breast cancer and assessed the importance of PAM50 subtype in response to FGFR4 inhibition.
Results
A FGFR4 activity gene signature was significantly upregulated post-neoadjuvant aromatase inhibitor treatment, and high FGFR4 expression predicted poorer survival in patients with ER+breast cancer. Gene expression association analysis using TCGA, METABRIC, and SCAN-B datasets uncovered ER as the most significant gene negatively correlated with FGFR4 expression. ER negatively regulates FGFR4 expression at both the mRNA and protein level across multiple ER+breast cancer cell lines. Despite robust overexpression of FGFR4, LTED cells did not show enhanced responses to FGFR4 inhibition compared to parental cells. Similarly, FGFR4 overexpression and knockdown did not substantially alter response to endocrine treatment in ER+cell lines, nor did FGFR4 and fulvestrant combination treatment show synergistic effects. The HER2-like subtype of breast cancer showed elevated expression of FGFR4 and an increased response to FGFR4 inhibition relative to other breast cancer subtypes.
Conclusions
Despite ER-mediated upregulation of FGFR4 post-endocrine therapy, our study does not support a general role of FGFR4 in mediating endocrine resistance in ER+breast cancer. The significant upregulation of FGFR4 expression in treatment-resistant clinical samples and models following endocrine therapy does not necessarily establish a causal link between the gene and treatment response. Our data suggest that specific genomic backgrounds such as HER2 expression may be required for FGFR4 function in breast cancer and should be further explored.
Similar content being viewed by others
Data availability
No datasets were generated or analysed during the current study.
References
Johnston SJ, Cheung KL (2018) Endocrine therapy for breast cancer: a model of hormonal manipulation. Oncol Ther 6(2):141–156
Clarke R, Liu MC, Bouker KB, Gu Z, Lee RY, Zhu Y, Skaar TC, Gomez B, O’Brien K, Wang Y et al (2003) Antiestrogen resistance in breast cancer and the role of estrogen receptor signaling. Oncogene 22(47):7316–7339
Clarke R, Tyson JJ, Dixon JM (2015) Endocrine resistance in breast cancer–an overview and update. Mol Cell Endocrinol. https://doi.org/10.1016/j.mce.2015.09.035
Redig AJ, McAllister SS (2013) Breast cancer as a systemic disease: a view of metastasis. J Intern Med 274(2):113–126
Hanker AB, Sudhan DR, Arteaga CL (2020) Overcoming endocrine resistance in breast cancer. Cancer Cell 37(4):496–513
Murphy CG, Dickler MN (2016) Endocrine resistance in hormone-responsive breast cancer: mechanisms and therapeutic strategies. Endocr Relat Cancer 23(8):R337-352
Osborne CK, Schiff R (2011) Mechanisms of endocrine resistance in breast cancer. Annu Rev Med 62:233–247
Levine KM, Priedigkeit N, Basudan A, Tasdemir N, Sikora MJ, Sokol ES, Hartmaier RJ, Ding K, Ahmad NZ, Watters RJ et al (2019) FGFR4 overexpression and hotspot mutations in metastatic ER+ breast cancer are enriched in the lobular subtype. NPJ Breast Cancer 5:19
Cejalvo JM, Martínez de Dueñas E, Galván P, García-Recio S, Burgués Gasión O, Paré L, Antolín S, Martinello R, Blancas I, Adamo B et al (2017) Intrinsic subtypes and gene expression profiles in primary and metastatic breast cancer. Cancer Res 77(9):2213–2221
Nagle AM, Levine KM, Tasdemir N, Scott JA, Burlbaugh K, Kehm J, Katz TA, Boone DN, Jacobsen BM, Atkinson JM et al (2018) Loss of E-cadherin enhances IGF1-IGF1R pathway activation and sensitizes breast cancers to anti-IGF1R/InsR inhibitors. Clin Cancer Res 24(20):5165–5177
Elangovan A, Hooda J, Savariau L, Puthanmadhomnarayanan S, Yates ME, Chen J, Brown DD, McAuliffe PF, Oesterreich S, Atkinson JM et al (2022) Loss of E-cadherin Induces IGF1R activation and reveals a targetable pathway in invasive lobular breast carcinoma. Mol Cancer Res 27:81
Meijer D, Sieuwerts AM, Look MP, van Agthoven T, Foekens JA, Dorssers LC (2008) Fibroblast growth factor receptor 4 predicts failure on tamoxifen therapy in patients with recurrent breast cancer. Endocr Relat Cancer 15(1):101–111
Levine KM, Ding K, Chen L, Oesterreich S (2020) FGFR4: a promising therapeutic target for breast cancer and other solid tumors. Pharmacol Ther 214:107590
Turner N, Grose R (2010) Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer 10(2):116–129
Kostrzewa M, Müller U (1998) Genomic structure and complete sequence of the human FGFR4 gene. Mamm Genome 9(2):131–135
Hagel M, Miduturu C, Sheets M, Rubin N, Weng W, Stransky N, Bifulco N, Kim JL, Hodous B, Brooijmans N et al (2015) First selective small molecule inhibitor of FGFR4 for the treatment of hepatocellular carcinomas with an activated FGFR4 signaling pathway. Cancer Discov 5(4):424–437
Joshi JJ, Coffey H, Corcoran E, Tsai J, Huang CL, Ichikawa K, Prajapati S, Hao MH, Bailey S, Wu J et al (2017) H3B–6527 Is a potent and selective inhibitor of FGFR4 in FGF19-driven hepatocellular carcinoma. Cancer Res 77(24):6999–7013
Ruggeri B, Stubbs M, Yang Y-o, Juvekar A, Lu L, Condon S, DiMatteo D, Wen X, Collier P, Burn T et al (2017) Abstract 1234: the novel FGFR4-selective inhibitor INCB062079 is efficacious in models of hepatocellular carcinoma harboring FGF19 amplification. Cancer Res. https://doi.org/10.1158/1538-7445.AM2017-1234
Chen X, Huang Y, Chen B, Liu H, Cai Y, Yang Y (2024) Insight into the design of FGFR4 selective inhibitors in cancer therapy: prospects and challenges. Eur J Med Chem 263:115947
Goetz R, Mohammadi M (2013) Exploring mechanisms of FGF signalling through the lens of structural biology. Nat Rev Mol Cell Biol 14(3):166–180
Cavallaro U, Niedermeyer J, Fuxa M, Christofori G (2001) N-CAM modulates tumour-cell adhesion to matrix by inducing FGF-receptor signalling. Nat Cell Biol 3(7):650–657
Christensen C, Berezin V, Bock E (2011) Neural cell adhesion molecule differentially interacts with isoforms of the fibroblast growth factor receptor. NeuroReport 22(15):727–732
Yokote H, Fujita K, Jing X, Sawada T, Liang S, Yao L, Yan X, Zhang Y, Schlessinger J, Sakaguchi K (2005) Trans-activation of EphA4 and FGF receptors mediated by direct interactions between their cytoplasmic domains. Proc Natl Acad Sci U S A 102(52):18866–18871
Sugiyama N, Varjosalo M, Meller P, Lohi J, Chan KM, Zhou Z, Alitalo K, Taipale J, Keski-Oja J, Lehti K (2010) FGF receptor-4 (FGFR4) polymorphism acts as an activity switch of a membrane type 1 matrix metalloproteinase-FGFR4 complex. Proc Natl Acad Sci U S A 107(36):15786–15791
Cheng AL, Shen YC, Zhu AX (2011) Targeting fibroblast growth factor receptor signaling in hepatocellular carcinoma. Oncology 81(5–6):372–380
Ho HK, Pok S, Streit S, Ruhe JE, Hart S, Lim KS, Loo HL, Aung MO, Lim SG, Ullrich A (2009) Fibroblast growth factor receptor 4 regulates proliferation, anti-apoptosis and alpha-fetoprotein secretion during hepatocellular carcinoma progression and represents a potential target for therapeutic intervention. J Hepatol 50(1):118–127
Raja A, Park I, Haq F, Ahn SM (2019) FGF19-FGFR4 signaling in hepatocellular carcinoma. Cells. https://doi.org/10.3390/cells8060536
Crose LE, Etheridge KT, Chen C, Belyea B, Talbot LJ, Bentley RC, Linardic CM (2012) FGFR4 blockade exerts distinct antitumorigenic effects in human embryonal versus alveolar rhabdomyosarcoma. Clin Cancer Res 18(14):3780–3790
Vi JGT, Cheuk AT, Tsang PS, Chung JY, Song YK, Desai K, Yu Y, Chen QR, Shah K, Youngblood V, Fang J (2009) Identification of FGFR4-activating mutations in human rhabdomyosarcomas that promote metastasis in xenotransplanted models. J Clin Invest 119(11):3395–3407
Luo Y, Yang C, Ye M, Jin C, Abbruzzese JL, Lee MH, Yeung SC, McKeehan WL (2013) Deficiency of metabolic regulator FGFR4 delays breast cancer progression through systemic and microenvironmental metabolic alterations. Cancer Metab 1(1):21
Tiong KH, Tan BS, Choo HL, Chung FF, Hii LW, Tan SH, Khor NT, Wong SF, See SJ, Tan YF et al (2016) Fibroblast growth factor receptor 4 (FGFR4) and fibroblast growth factor 19 (FGF19) autocrine enhance breast cancer cells survival. Oncotarget 7(36):57633–57650
Xu M, Chen S, Yang W, Cheng X, Ye Y, Mao J, Wu X, Huang L, Ji J (2018) FGFR4 links glucose metabolism and chemotherapy resistance in breast cancer. Cell Physiol Biochem 47(1):151–160
Li SQ, Cheuk AT, Shern JF, Song YK, Hurd L, Liao H, Wei JS, Khan J (2013) Targeting wild-type and mutationally activated FGFR4 in rhabdomyosarcoma with the inhibitor ponatinib (AP24534). PLoS ONE 8(10):e76551
Garcia-Recio S, Thennavan A, East MP, Parker JS, Cejalvo JM, Garay JP, Hollern DP, He X, Mott KR, Galván P et al (2020) FGFR4 regulates tumor subtype differentiation in luminal breast cancer and metastatic disease. J Clin Invest 130(9):4871–4887
Miller CA, Gindin Y, Lu C, Griffith OL, Griffith M, Shen D, Hoog J, Li T, Larson DE, Watson M et al (2016) Aromatase inhibition remodels the clonal architecture of estrogen-receptor-positive breast cancers. Nat Commun 7:12498
Turnbull AK, Arthur LM, Renshaw L, Larionov AA, Kay C, Dunbier AK, Thomas JS, Dowsett M, Sims AH, Dixon JM (2015) Accurate prediction and validation of response to endocrine therapy in breast cancer. J Clin Oncol 33(20):2270–2278
Sikora MJ, Jacobsen BM, Levine K, Chen J, Davidson NE, Lee AV, Alexander CM, Oesterreich S (2016) WNT4 mediates estrogen receptor signaling and endocrine resistance in invasive lobular carcinoma cell lines. Breast Cancer Res 18(1):92
Fu X, Jeselsohn R, Pereira R, Hollingsworth EF, Creighton CJ, Li F, Shea M, Nardone A, De Angelis C, Heiser LM et al (2016) FOXA1 overexpression mediates endocrine resistance by altering the ER transcriptome and IL-8 expression in ER-positive breast cancer. Proc Natl Acad Sci U S A 113(43):E6600-e6609
Ornitz DM, Xu J, Colvin JS, McEwen DG, MacArthur CA, Coulier F, Gao G, Goldfarb M (1996) Receptor specificity of the fibroblast growth factor family. J Biol Chem 271(25):15292–15297
Zhang X, Ibrahimi OA, Olsen SK, Umemori H, Mohammadi M, Ornitz DM (2006) Receptor specificity of the fibroblast growth factor family: the complete mammalian FGF family. J Biol Chem 281(23):15694–15700
Bange J, Prechtl D, Cheburkin Y, Specht K, Harbeck N, Schmitt M, Knyazeva T, Müller S, Gärtner S, Sures I et al (2002) Cancer progression and tumor cell motility are associated with the FGFR4 Arg(388) allele. Cancer Res 62(3):840–847
Morimoto Y, Ozaki T, Ouchida M, Umehara N, Ohata N, Yoshida A, Shimizu K, Inoue H (2003) Single nucleotide polymorphism in fibroblast growth factor receptor 4 at codon 388 is associated with prognosis in high-grade soft tissue sarcoma. Cancer 98(10):2245–2250
Ghandi M, Huang FW, Jané-Valbuena J, Kryukov GV, Lo CC, McDonald ER 3rd, Barretina J, Gelfand ET, Bielski CM, Li H et al (2019) Next-generation characterization of the cancer cell line encyclopedia. Nature 569(7757):503–508
Helsten T, Elkin S, Arthur E, Tomson BN, Carter J, Kurzrock R (2016) The FGFR landscape in cancer: analysis of 4853 tumors by next-generation sequencing. Clin Cancer Res 22(1):259–267
Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA et al (2000) Molecular portraits of human breast tumours. Nature 406(6797):747–752
Carroll JS (2016) Mechanisms of oestrogen receptor (ER) gene regulation in breast cancer. Eur J Endocrinol 175(1):R41-49
Cheng R, Qi L, Kong X, Wang Z, Fang Y, Wang J (2020) Identification of the significant genes regulated by estrogen receptor in estrogen receptor-positive breast cancer and their expression pattern changes when tamoxifen or fulvestrant resistance occurs. Front Genet 11:538734
Maor S, Mayer D, Yarden RI, Lee AV, Sarfstein R, Werner H, Papa MZ (2006) Estrogen receptor regulates insulin-like growth factor-I receptor gene expression in breast tumor cells: involvement of transcription factor Sp1. J Endocrinol 191(3):605–612
Yarden RI, Wilson MA, Chrysogelos SA (2001) Estrogen suppression of EGFR expression in breast cancer cells: a possible mechanism to modulate growth. J Cell Biochem Suppl Suppl 36:232–246
Li Z, Li T, Yates ME, Wu Y, Ferber A, Chen L, Brown DD, Carroll JS, Sikora MJ, Tseng GC et al (2023) The estrogene database reveals diverse temporal, context-dependent, and bidirectional estrogen receptor regulomes in breast cancer. Cancer Res 83(16):2656–2674
Turner N, Pearson A, Sharpe R, Lambros M, Geyer F, Lopez-Garcia MA, Natrajan R, Marchio C, Iorns E, Mackay A et al (2010) FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer. Cancer Res 70(5):2085–2094
Drago JZ, Formisano L, Juric D, Niemierko A, Servetto A, Wander SA, Spring LM, Vidula N, Younger J, Peppercorn J et al (2019) FGFR1 amplification mediates endocrine resistance but retains TORC sensitivity in metastatic hormone receptor-positive (HR(+)) breast cancer. Clin Cancer Res 25(21):6443–6451
Formisano L, Stauffer KM, Young CD, Bhola NE, Guerrero-Zotano AL, Jansen VM, Estrada MM, Hutchinson KE, Giltnane JM, Schwarz LJ et al (2017) Association of FGFR1 with ERα maintains ligand-independent ER transcription and mediates resistance to estrogen deprivation in ER(+) breast cancer. Clin Cancer Res 23(20):6138–6150
Turczyk L, Kitowska K, Mieszkowska M, Mieczkowski K, Czaplinska D, Piasecka D, Kordek R, Skladanowski AC, Potemski P, Romanska HM et al (2017) FGFR2-driven signaling counteracts tamoxifen effect on ERα-positive breast cancer cells. Neoplasia 19(10):791–804
Mao P, Cohen O, Kowalski KJ, Kusiel JG, Buendia-Buendia JE, Cuoco MS, Exman P, Wander SA, Waks AG, Nayar U et al (2020) Acquired FGFR and FGF alterations confer resistance to estrogen receptor (ER) targeted therapy in ER(+) metastatic breast cancer. Clin Cancer Res 26(22):5974–5989
French DM, Lin BC, Wang M, Adams C, Shek T, Hötzel K, Bolon B, Ferrando R, Blackmore C, Schroeder K et al (2012) Targeting FGFR4 inhibits hepatocellular carcinoma in preclinical mouse models. PLoS ONE 7(5):e36713
Zhao H, Lv F, Liang G, Huang X, Wu G, Zhang W, Yu L, Shi L, Teng Y (2016) FGF19 promotes epithelial-mesenchymal transition in hepatocellular carcinoma cells by modulating the GSK3β/β- catenin signaling cascade via FGFR4 activation. Oncotarget 7(12):13575–13586
Zou Y, Zheng S, Xie X, Ye F, Hu X, Tian Z, Yan SM, Yang L, Kong Y, Tang Y et al (2022) N6-methyladenosine regulated FGFR4 attenuates ferroptotic cell death in recalcitrant HER2-positive breast cancer. Nat Commun 13(1):2672
Hänzelmann S, Castelo R, Guinney J (2013) GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14:7
Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, Speed D, Lynch AG, Samarajiwa S, Yuan Y et al (2012) The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 486(7403):346–352
Robinson MD, McCarthy DJ, Smyth GK (2010) edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26(1):139–140
Jambal P, Badtke MM, Harrell JC, Borges VF, Post MD, Sollender GE, Spillman MA, Horwitz KB, Jacobsen BM (2013) Estrogen switches pure mucinous breast cancer to invasive lobular carcinoma with mucinous features. Breast Cancer Res Treat 137(2):431–448
Du T, Zhu L, Levine KM, Tasdemir N, Lee AV, Vignali DAA, Houten BV, Tseng GC, Oesterreich S (2018) Invasive lobular and ductal breast carcinoma differ in immune response, protein translation efficiency and metabolism. Sci Rep 8(1):7205
Acknowledgements
We would like to thank The Cancer Genome Atlas (TCGA) Research Network (https://www.cancer.gov/tcga), the Sweden Cancerome Analysis Network, and the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) for generating and sharing their data to empower our study. We appreciate technical support from Jian Chen and Loise Mazur, and support from personnel the Department of Pathology at Magee Womens Hospital, and the Pittsburgh Biospecimen Core (PBC). We are grateful to Blueprint Medicines (BPMC) and H3 Biomedicine for providing BLU554 and H3B-6527, respectively.
Funding
This work was supported in part by NIH R01 CA224909 (to SO), Susan G Komen (SAC160073 SO), Shear Family Foundation, Magee Womens Research Institute and Foundation, and by UPMC Hillman Cancer Center and Tissue and Research Pathology/Pitt Biospecimen Core and the Biostatistics Core shared resources, which are supported in part by award P30CA047904.
Author information
Authors and Affiliations
Contributions
All authors contributed to the study design and the preparation of the manuscript. K.D., L.C., K.L., and O.S. conducted the studies under the guidance of J.A., A.V.L. and S.O. M.S., N.T., D.D., R.J. and R.H. provided models and clinical data essential for this study.
Corresponding author
Ethics declarations
Conflict of interest
The authors have no conflicts of interest to disclose.
Ethical approval
All clinical samples were collected with approval from the Institutional Review Board (IRB) at the University of Pittsburgh.
Consent to participate
All clinical samples were collected with informed consent.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Below is the link to the electronic supplementary material.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Ding, K., Chen, L., Levine, K.M. et al. FGFR4 in endocrine resistance: overexpression and estrogen regulation without direct causative role. Breast Cancer Res Treat 211, 501–515 (2025). https://doi.org/10.1007/s10549-025-07666-x
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1007/s10549-025-07666-x