HGNC Family | Immunoglobulin superfamily domain containing, CD molecules (CD), Receptor kinases |
Name | fibroblast growth factor receptor 2 |
Description | The protein encoded by this gene is a member of the fibroblast growth factor receptor family, where amino acid sequence is highly conserved between members and throughout evolution. FGFR family members differ from one another in their ligand affinities and tissue distribution. A full-length representative protein consists of an extracellular region, composed of three immunoglobulin-like domains, a single hydrophobic membrane-spanning segment and a cytoplasmic tyrosine kinase domain. The extracellular portion of the protein interacts with fibroblast growth factors, setting in motion a cascade of downstream signals, ultimately influencing mitogenesis and differentiation. This particular family member is a high-affinity receptor for acidic, basic and/or keratinocyte growth factor, depending on the isoform. Mutations in this gene are associated with Crouzon syndrome, Pfeiffer syndrome, Craniosynostosis, Apert syndrome, Jackson-Weiss syndrome, Beare-Stevenson cutis gyrata syndrome, Saethre-Chotzen syndrome, and syndromic craniosynostosis. Multiple alternatively spliced transcript variants encoding different isoforms have been noted for this gene. [provided by RefSeq, Jan 2009] |
Summary |
{"type": "root", "children": [{"type": "p", "children": [{"type": "t", "text": "\nFibroblast growth factor receptor 2 (FGFR2) is a receptor tyrosine kinase that plays a pivotal role in normal development and tissue homeostasis. Its activity is tightly controlled by mechanisms such as alternative splicing—which generates the epithelial FGFR2b and mesenchymal FGFR2c isoforms to confer distinct ligand‐binding specificities—and by intramolecular regulatory features (for example, an autoinhibitory “molecular brake” and modulatory interactions with adaptor proteins). Structural and functional studies have revealed that mutations affecting key extracellular or kinase domain regions of FGFR2 can disrupt this delicate regulation and lead to developmental disorders such as craniosynostosis and Apert syndrome."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "1", "end_ref": "7"}]}, {"type": "t", "text": "\n"}]}, {"type": "t", "text": "\n\n"}, {"type": "p", "children": [{"type": "t", "text": "\nIn the context of breast cancer, genome‐wide association studies have pinpointed single nucleotide polymorphisms (SNPs) in an intronic region of FGFR2 that associate with a moderately increased risk—particularly for estrogen receptor–positive tumors. Functional analyses support a model in which these noncoding variants alter transcription factor binding (for example, sites for Oct‐1/Runx2 and C/EBPβ) and consequently upregulate FGFR2 expression, thereby enhancing proliferative signaling in breast epithelium. Additional studies demonstrate that FGFR2 signaling integrates with nuclear receptor networks and is further modulated by microRNAs (such as miR‑125b), collectively influencing cell proliferation and differentiation during tumor initiation."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "8", "end_ref": "13"}]}, {"type": "t", "text": "\n"}]}, {"type": "t", "text": "\n\n"}, {"type": "p", "children": [{"type": "t", "text": "\nBeyond its role in normal physiology and breast cancer predisposition, FGFR2 is a recurrent target of oncogenic alterations in a variety of malignancies. Activating mutations, gene amplifications, and fusion events involving FGFR2 have been identified in cancers such as endometrial carcinoma, gastric carcinoma, intrahepatic cholangiocarcinoma, lung, and even colorectal tumors. In these settings, aberrant FGFR2 signaling—often mediated through downstream MAPK, PI3K, and EGFR family cascades—drives tumor cell proliferation, survival, and even angiogenesis via an FGF‑2/FGFR2 axis. Notably, FGFR2 fusions (for example, FGFR2–AHCYL1, FGFR2–BICC1, or FGFR2–PPHLN1) have emerged as actionable targets in cholangiocarcinoma, and preclinical models demonstrate that selective FGFR inhibitors can overcome primary and acquired resistance arising from secondary kinase mutations. Moreover, in some tumors FGFR2 pathway activity interplays with cancer stem cell–like gene expression programs and influences epithelial–mesenchymal transitions, underlining its diverse contributions to oncogenesis and therapy resistance."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "14", "end_ref": "46"}]}, {"type": "t", "text": "\n"}]}, {"type": "rg", "children": [{"type": "r", "ref": 1, "children": [{"type": "t", "text": "Shih-hsin Kan, Navaratnam Elanko, David Johnson, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Genomic screening of fibroblast growth-factor receptor 2 reveals a wide spectrum of mutations in patients with syndromic craniosynostosis."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Am J Hum Genet (2002)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1086/338758"}], "href": "https://doi.org/10.1086/338758"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "11781872"}], "href": "https://pubmed.ncbi.nlm.nih.gov/11781872"}]}, {"type": "r", "ref": 2, "children": [{"type": "t", "text": "Brian K Yeh, Makoto Igarashi, Anna V Eliseenkova, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Structural basis by which alternative splicing confers specificity in fibroblast growth factor receptors."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Proc Natl Acad Sci U S A (2003)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1073/pnas.0436500100"}], "href": "https://doi.org/10.1073/pnas.0436500100"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "12591959"}], "href": "https://pubmed.ncbi.nlm.nih.gov/12591959"}]}, {"type": "r", "ref": 3, "children": [{"type": "t", "text": "Omar A Ibrahimi, Fuming Zhang, Anna V Eliseenkova, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Biochemical analysis of pathogenic ligand-dependent FGFR2 mutations suggests distinct pathophysiological mechanisms for craniofacial and limb abnormalities."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Hum Mol Genet (2004)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1093/hmg/ddh235"}], "href": "https://doi.org/10.1093/hmg/ddh235"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "15282208"}], "href": "https://pubmed.ncbi.nlm.nih.gov/15282208"}]}, {"type": "r", "ref": 4, "children": [{"type": "t", "text": "Masaru Katoh "}, {"type": "b", "children": [{"type": "t", "text": "Cancer genomics and genetics of FGFR2 (Review)."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Int J Oncol (2008)"}]}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18636142"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18636142"}]}, {"type": "r", "ref": 5, "children": [{"type": "t", "text": "Claude C Warzecha, Trey K Sato, Behnam Nabet, et al. "}, {"type": "b", "children": [{"type": "t", "text": "ESRP1 and ESRP2 are epithelial cell-type-specific regulators of FGFR2 splicing."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Cell (2009)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.molcel.2009.01.025"}], "href": "https://doi.org/10.1016/j.molcel.2009.01.025"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "19285943"}], "href": "https://pubmed.ncbi.nlm.nih.gov/19285943"}]}, {"type": "r", "ref": 6, "children": [{"type": "t", "text": "Chi-Chuan Lin, Fernando A Melo, Ragini Ghosh, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Inhibition of basal FGF receptor signaling by dimeric Grb2."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cell (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.cell.2012.04.033"}], "href": "https://doi.org/10.1016/j.cell.2012.04.033"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22726438"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22726438"}]}, {"type": "r", "ref": 7, "children": [{"type": "t", "text": "Lipika Goyal, Funda Meric-Bernstam, Antoine Hollebecque, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Futibatinib for "}, {"type": "a", "children": [{"type": "t", "text": "i"}], "href": "i"}, {"type": "t", "text": "FGFR2"}, {"type": "a", "children": [{"type": "t", "text": "/i"}], "href": "/i"}, {"type": "t", "text": "-Rearranged Intrahepatic Cholangiocarcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "N Engl J Med (2023)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1056/NEJMoa2206834"}], "href": "https://doi.org/10.1056/NEJMoa2206834"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "36652354"}], "href": "https://pubmed.ncbi.nlm.nih.gov/36652354"}]}, {"type": "r", "ref": 8, "children": [{"type": "t", "text": "David J Hunter, Peter Kraft, Kevin B Jacobs, et al. "}, {"type": "b", "children": [{"type": "t", "text": "A genome-wide association study identifies alleles in FGFR2 associated with risk of sporadic postmenopausal breast cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Nat Genet (2007)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/ng2075"}], "href": "https://doi.org/10.1038/ng2075"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "17529973"}], "href": "https://pubmed.ncbi.nlm.nih.gov/17529973"}]}, {"type": "r", "ref": 9, "children": [{"type": "t", "text": "Huaibin Chen, Jinghong Ma, Wanqing Li, et al. "}, {"type": "b", "children": [{"type": "t", "text": "A molecular brake in the kinase hinge region regulates the activity of receptor tyrosine kinases."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Cell (2007)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.molcel.2007.06.028"}], "href": "https://doi.org/10.1016/j.molcel.2007.06.028"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "17803937"}], "href": "https://pubmed.ncbi.nlm.nih.gov/17803937"}]}, {"type": "r", "ref": 10, "children": [{"type": "t", "text": "Montserrat Garcia-Closas, Per Hall, Heli Nevanlinna, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Heterogeneity of breast cancer associations with five susceptibility loci by clinical and pathological characteristics."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "PLoS Genet (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1371/journal.pgen.1000054"}], "href": "https://doi.org/10.1371/journal.pgen.1000054"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18437204"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18437204"}]}, {"type": "r", "ref": 11, "children": [{"type": "t", "text": "Kerstin B Meyer, Ana-Teresa Maia, Martin O'Reilly, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Allele-specific up-regulation of FGFR2 increases susceptibility to breast cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "PLoS Biol (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1371/journal.pbio.0060108"}], "href": "https://doi.org/10.1371/journal.pbio.0060108"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18462018"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18462018"}]}, {"type": "r", "ref": 12, "children": [{"type": "t", "text": "Ning Xu, Petter Brodin, Tianling Wei, et al. "}, {"type": "b", "children": [{"type": "t", "text": "MiR-125b, a microRNA downregulated in psoriasis, modulates keratinocyte proliferation by targeting FGFR2."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Invest Dermatol (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/jid.2011.55"}], "href": "https://doi.org/10.1038/jid.2011.55"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21412257"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21412257"}]}, {"type": "r", "ref": 13, "children": [{"type": "t", "text": "Michael N C Fletcher, Mauro A A Castro, Xin Wang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Master regulators of FGFR2 signalling and breast cancer risk."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Nat Commun (2013)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/ncomms3464"}], "href": "https://doi.org/10.1038/ncomms3464"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24043118"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24043118"}]}, {"type": "r", "ref": 14, "children": [{"type": "t", "text": "Anne Goriely, Gilean A T McVean, Maria Röjmyr, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Evidence for selective advantage of pathogenic FGFR2 mutations in the male germ line."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Science (2003)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1126/science.1085710"}], "href": "https://doi.org/10.1126/science.1085710"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "12893942"}], "href": "https://pubmed.ncbi.nlm.nih.gov/12893942"}]}, {"type": "r", "ref": 15, "children": [{"type": "t", "text": "Anne Goriely, Gilean A T McVean, Ans M M van Pelt, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Gain-of-function amino acid substitutions drive positive selection of FGFR2 mutations in human spermatogonia."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Proc Natl Acad Sci U S A (2005)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1073/pnas.0500267102"}], "href": "https://doi.org/10.1073/pnas.0500267102"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "15840724"}], "href": "https://pubmed.ncbi.nlm.nih.gov/15840724"}]}, {"type": "r", "ref": 16, "children": [{"type": "t", "text": "Gennadi V Glinsky, Olga Berezovska, Anna B Glinskii "}, {"type": "b", "children": [{"type": "t", "text": "Microarray analysis identifies a death-from-cancer signature predicting therapy failure in patients with multiple types of cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Clin Invest (2005)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1172/JCI23412"}], "href": "https://doi.org/10.1172/JCI23412"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "15931389"}], "href": "https://pubmed.ncbi.nlm.nih.gov/15931389"}]}, {"type": "r", "ref": 17, "children": [{"type": "t", "text": "P M Pollock, M G Gartside, L C Dejeza, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Frequent activating FGFR2 mutations in endometrial carcinomas parallel germline mutations associated with craniosynostosis and skeletal dysplasia syndromes."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Oncogene (2007)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/sj.onc.1210529"}], "href": "https://doi.org/10.1038/sj.onc.1210529"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "17525745"}], "href": "https://pubmed.ncbi.nlm.nih.gov/17525745"}]}, {"type": "r", "ref": 18, "children": [{"type": "t", "text": "Antonis C Antoniou, Amanda B Spurdle, Olga M Sinilnikova, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Common breast cancer-predisposition alleles are associated with breast cancer risk in BRCA1 and BRCA2 mutation carriers."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Am J Hum Genet (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.ajhg.2008.02.008"}], "href": "https://doi.org/10.1016/j.ajhg.2008.02.008"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18355772"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18355772"}]}, {"type": "r", "ref": 19, "children": [{"type": "t", "text": "Kaiko Kunii, Lenora Davis, Julie Gorenstein, et al. "}, {"type": "b", "children": [{"type": "t", "text": "FGFR2-amplified gastric cancer cell lines require FGFR2 and Erbb3 signaling for growth and survival."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cancer Res (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1158/0008-5472.CAN-07-5229"}], "href": "https://doi.org/10.1158/0008-5472.CAN-07-5229"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18381441"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18381441"}]}, {"type": "r", "ref": 20, "children": [{"type": "t", "text": "Sara A Byron, Michael G Gartside, Candice L Wellens, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Inhibition of activated fibroblast growth factor receptor 2 in endometrial cancer cells induces cell death despite PTEN abrogation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cancer Res (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1158/0008-5472.CAN-08-0770"}], "href": "https://doi.org/10.1158/0008-5472.CAN-08-0770"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18757403"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18757403"}]}, {"type": "r", "ref": 21, "children": [{"type": "t", "text": "Wei Liu, Sonia Cheng, Sylvia L Asa, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The melanoma-associated antigen A3 mediates fibronectin-controlled cancer progression and metastasis."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cancer Res (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1158/0008-5472.CAN-08-2132"}], "href": "https://doi.org/10.1158/0008-5472.CAN-08-2132"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18829569"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18829569"}]}, {"type": "r", "ref": 22, "children": [{"type": "t", "text": "Michael G Gartside, Huaibin Chen, Omar A Ibrahimi, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Loss-of-function fibroblast growth factor receptor-2 mutations in melanoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Cancer Res (2009)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1158/1541-7786.MCR-08-0021"}], "href": "https://doi.org/10.1158/1541-7786.MCR-08-0021"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "19147536"}], "href": "https://pubmed.ncbi.nlm.nih.gov/19147536"}]}, {"type": "r", "ref": 23, "children": [{"type": "t", "text": "Yuriko Katoh, Masaru Katoh "}, {"type": "b", "children": [{"type": "t", "text": "FGFR2-related pathogenesis and FGFR2-targeted therapeutics (Review)."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Int J Mol Med (2009)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.3892/ijmm_00000132"}], "href": "https://doi.org/10.3892/ijmm_00000132"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "19212647"}], "href": "https://pubmed.ncbi.nlm.nih.gov/19212647"}]}, {"type": "r", "ref": 24, "children": [{"type": "t", "text": "Veronica C Ardi, Philippe E Van den Steen, Ghislain Opdenakker, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Neutrophil MMP-9 proenzyme, unencumbered by TIMP-1, undergoes efficient activation in vivo and catalytically induces angiogenesis via a basic fibroblast growth factor (FGF-2)/FGFR-2 pathway."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2009)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M109.033472"}], "href": "https://doi.org/10.1074/jbc.M109.033472"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "19608737"}], "href": "https://pubmed.ncbi.nlm.nih.gov/19608737"}]}, {"type": "r", "ref": 25, "children": [{"type": "t", "text": "Sholom Wacholder, Patricia Hartge, Ross Prentice, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Performance of common genetic variants in breast-cancer risk models."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "N Engl J Med (2010)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1056/NEJMoa0907727"}], "href": "https://doi.org/10.1056/NEJMoa0907727"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "20237344"}], "href": "https://pubmed.ncbi.nlm.nih.gov/20237344"}]}, {"type": "r", "ref": 26, "children": [{"type": "t", "text": "Ruth C Travis, Gillian K Reeves, Jane Green, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Gene-environment interactions in 7610 women with breast cancer: prospective evidence from the Million Women Study."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Lancet (2010)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/S0140-6736(10)60636-8"}], "href": "https://doi.org/10.1016/S0140-6736(10"}, {"type": "t", "text": "60636-8) PMID: "}, {"type": "a", "children": [{"type": "t", "text": "20605201"}], "href": "https://pubmed.ncbi.nlm.nih.gov/20605201"}]}, {"type": "r", "ref": 27, "children": [{"type": "t", "text": "Kathryn E Ware, Marianne E Marshall, Lydia R Heasley, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Rapidly acquired resistance to EGFR tyrosine kinase inhibitors in NSCLC cell lines through de-repression of FGFR2 and FGFR3 expression."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "PLoS One (2010)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1371/journal.pone.0014117"}], "href": "https://doi.org/10.1371/journal.pone.0014117"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21152424"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21152424"}]}, {"type": "r", "ref": 28, "children": [{"type": "t", "text": "Daniel L Coutu, Moïra François, Jacques Galipeau "}, {"type": "b", "children": [{"type": "t", "text": "Inhibition of cellular senescence by developmentally regulated FGF receptors in mesenchymal stem cells."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Blood (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1182/blood-2010-12-321539"}], "href": "https://doi.org/10.1182/blood-2010-12-321539"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21527526"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21527526"}]}, {"type": "r", "ref": 29, "children": [{"type": "t", "text": "K Matsumoto, T Arao, T Hamaguchi, et al. "}, {"type": "b", "children": [{"type": "t", "text": "FGFR2 gene amplification and clinicopathological features in gastric cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Br J Cancer (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/bjc.2011.603"}], "href": "https://doi.org/10.1038/bjc.2011.603"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22240789"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22240789"}]}, {"type": "r", "ref": 30, "children": [{"type": "t", "text": "Sara A Byron, Michael Gartside, Matthew A Powell, et al. "}, {"type": "b", "children": [{"type": "t", "text": "FGFR2 point mutations in 466 endometrioid endometrial tumors: relationship with MSI, KRAS, PIK3CA, CTNNB1 mutations and clinicopathological features."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "PLoS One (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1371/journal.pone.0030801"}], "href": "https://doi.org/10.1371/journal.pone.0030801"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22383975"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22383975"}]}, {"type": "r", "ref": 31, "children": [{"type": "t", "text": "Liang Xie, Xinying Su, Lin Zhang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Clin Cancer Res (2013)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1158/1078-0432.CCR-12-3898"}], "href": "https://doi.org/10.1158/1078-0432.CCR-12-3898"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "23493349"}], "href": "https://pubmed.ncbi.nlm.nih.gov/23493349"}]}, {"type": "r", "ref": 32, "children": [{"type": "t", "text": "Sara A Byron, Huaibin Chen, Andreas Wortmann, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The N550K/H mutations in FGFR2 confer differential resistance to PD173074, dovitinib, and ponatinib ATP-competitive inhibitors."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Neoplasia (2013)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1593/neo.121106"}], "href": "https://doi.org/10.1593/neo.121106"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "23908597"}], "href": "https://pubmed.ncbi.nlm.nih.gov/23908597"}]}, {"type": "r", "ref": 33, "children": [{"type": "t", "text": "Yasuhito Arai, Yasushi Totoki, Fumie Hosoda, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Hepatology (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1002/hep.26890"}], "href": "https://doi.org/10.1002/hep.26890"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24122810"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24122810"}]}, {"type": "r", "ref": 34, "children": [{"type": "t", "text": "Mitesh J Borad, Mia D Champion, Jan B Egan, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Integrated genomic characterization reveals novel, therapeutically relevant drug targets in FGFR and EGFR pathways in sporadic intrahepatic cholangiocarcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "PLoS Genet (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1371/journal.pgen.1004135"}], "href": "https://doi.org/10.1371/journal.pgen.1004135"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24550739"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24550739"}]}, {"type": "r", "ref": 35, "children": [{"type": "t", "text": "Rondell P Graham, Emily G Barr Fritcher, Ekaterina Pestova, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Fibroblast growth factor receptor 2 translocations in intrahepatic cholangiocarcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Hum Pathol (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.humpath.2014.03.014"}], "href": "https://doi.org/10.1016/j.humpath.2014.03.014"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24837095"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24837095"}]}, {"type": "r", "ref": 36, "children": [{"type": "t", "text": "Rui Wang, Lei Wang, Yuan Li, et al. "}, {"type": "b", "children": [{"type": "t", "text": "FGFR1/3 tyrosine kinase fusions define a unique molecular subtype of non-small cell lung cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Clin Cancer Res (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1158/1078-0432.CCR-14-0284"}], "href": "https://doi.org/10.1158/1078-0432.CCR-14-0284"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24850843"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24850843"}]}, {"type": "r", "ref": 37, "children": [{"type": "t", "text": "Noah A Brown, Delphine Rolland, Jonathan B McHugh, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Activating FGFR2-RAS-BRAF mutations in ameloblastoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Clin Cancer Res (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1158/1078-0432.CCR-14-1069"}], "href": "https://doi.org/10.1158/1078-0432.CCR-14-1069"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24993163"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24993163"}]}, {"type": "r", "ref": 38, "children": [{"type": "t", "text": "Li Tan, Jun Wang, Junko Tanizaki, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Development of covalent inhibitors that can overcome resistance to first-generation FGFR kinase inhibitors."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Proc Natl Acad Sci U S A (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1073/pnas.1403438111"}], "href": "https://doi.org/10.1073/pnas.1403438111"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25349422"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25349422"}]}, {"type": "r", "ref": 39, "children": [{"type": "t", "text": "Daniela Sia, Bojan Losic, Agrin Moeini, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Massive parallel sequencing uncovers actionable FGFR2-PPHLN1 fusion and ARAF mutations in intrahepatic cholangiocarcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Nat Commun (2015)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/ncomms7087"}], "href": "https://doi.org/10.1038/ncomms7087"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25608663"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25608663"}]}, {"type": "r", "ref": 40, "children": [{"type": "t", "text": "Mohamed R Akl, Poonam Nagpal, Nehad M Ayoub, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Molecular and clinical significance of fibroblast growth factor 2 (FGF2 /bFGF) in malignancies of solid and hematological cancers for personalized therapies."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Oncotarget (2016)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.18632/oncotarget.8203"}], "href": "https://doi.org/10.18632/oncotarget.8203"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "27007053"}], "href": "https://pubmed.ncbi.nlm.nih.gov/27007053"}]}, {"type": "r", "ref": 41, "children": [{"type": "t", "text": "William J Gibson, Erling A Hoivik, Mari K Halle, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The genomic landscape and evolution of endometrial carcinoma progression and abdominopelvic metastasis."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Nat Genet (2016)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/ng.3602"}], "href": "https://doi.org/10.1038/ng.3602"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "27348297"}], "href": "https://pubmed.ncbi.nlm.nih.gov/27348297"}]}, {"type": "r", "ref": 42, "children": [{"type": "t", "text": "Lipika Goyal, Supriya K Saha, Leah Y Liu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Polyclonal Secondary "}, {"type": "a", "children": [{"type": "t", "text": "i"}], "href": "i"}, {"type": "t", "text": "FGFR2"}, {"type": "a", "children": [{"type": "t", "text": "/i"}], "href": "/i"}, {"type": "t", "text": " Mutations Drive Acquired Resistance to FGFR Inhibition in Patients with FGFR2 Fusion-Positive Cholangiocarcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cancer Discov (2017)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1158/2159-8290.CD-16-1000"}], "href": "https://doi.org/10.1158/2159-8290.CD-16-1000"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "28034880"}], "href": "https://pubmed.ncbi.nlm.nih.gov/28034880"}]}, {"type": "r", "ref": 43, "children": [{"type": "t", "text": "Vincenzo Mazzaferro, Bassel F El-Rayes, Michele Droz Dit Busset, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Derazantinib (ARQ 087) in advanced or inoperable FGFR2 gene fusion-positive intrahepatic cholangiocarcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Br J Cancer (2019)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/s41416-018-0334-0"}], "href": "https://doi.org/10.1038/s41416-018-0334-0"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "30420614"}], "href": "https://pubmed.ncbi.nlm.nih.gov/30420614"}]}, {"type": "r", "ref": 44, "children": [{"type": "t", "text": "Luigi Formisano, Yao Lu, Alberto Servetto, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Aberrant FGFR signaling mediates resistance to CDK4/6 inhibitors in ER+ breast cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Nat Commun (2019)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/s41467-019-09068-2"}], "href": "https://doi.org/10.1038/s41467-019-09068-2"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "30914635"}], "href": "https://pubmed.ncbi.nlm.nih.gov/30914635"}]}, {"type": "r", "ref": 45, "children": [{"type": "t", "text": "Piao Li, Tingting Huang, Qi Zou, et al. "}, {"type": "b", "children": [{"type": "t", "text": "FGFR2 Promotes Expression of PD-L1 in Colorectal Cancer via the JAK/STAT3 Signaling Pathway."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Immunol (2019)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.4049/jimmunol.1801199"}], "href": "https://doi.org/10.4049/jimmunol.1801199"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "30979816"}], "href": "https://pubmed.ncbi.nlm.nih.gov/30979816"}]}, {"type": "r", "ref": 46, "children": [{"type": "t", "text": "Ghassan K Abou-Alfa, Vaibhav Sahai, Antoine Hollebecque, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Lancet Oncol (2020)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/S1470-2045(20)30109-1"}], "href": "https://doi.org/10.1016/S1470-2045(20"}, {"type": "t", "text": "30109-1) PMID: "}, {"type": "a", "children": [{"type": "t", "text": "32203698"}], "href": "https://pubmed.ncbi.nlm.nih.gov/32203698"}]}]}]}
|
Synonyms | TK25, CD332, KGFR, CEK3, K-SAM, TK14, CFD1, BEK, BBDS, BFR-1, ECT1, JWS |
Proteins | FGFR2_HUMAN |
NCBI Gene ID | 2263 |
API | |
Download Associations | |
Predicted Functions |
![]() |
Co-expressed Genes |
![]() |
Expression in Tissues and Cell Lines |
![]() |
FGFR2 has 16,762 functional associations with biological entities spanning 9 categories (molecular profile, organism, disease, phenotype or trait, chemical, functional term, phrase or reference, structural feature, cell line, cell type or tissue, gene, protein or microRNA, sequence feature) extracted from 141 datasets.
Click the + buttons to view associations for FGFR2 from the datasets below.
If available, associations are ranked by standardized value
Dataset | Summary | |
---|---|---|
Allen Brain Atlas Adult Human Brain Tissue Gene Expression Profiles | tissues with high or low expression of FGFR2 gene relative to other tissues from the Allen Brain Atlas Adult Human Brain Tissue Gene Expression Profiles dataset. | |
Allen Brain Atlas Adult Mouse Brain Tissue Gene Expression Profiles | tissues with high or low expression of FGFR2 gene relative to other tissues from the Allen Brain Atlas Adult Mouse Brain Tissue Gene Expression Profiles dataset. | |
Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by Microarray | tissue samples with high or low expression of FGFR2 gene relative to other tissue samples from the Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by Microarray dataset. | |
Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by RNA-seq | tissue samples with high or low expression of FGFR2 gene relative to other tissue samples from the Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by RNA-seq dataset. | |
Allen Brain Atlas Prenatal Human Brain Tissue Gene Expression Profiles | tissues with high or low expression of FGFR2 gene relative to other tissues from the Allen Brain Atlas Prenatal Human Brain Tissue Gene Expression Profiles dataset. | |
BioGPS Cell Line Gene Expression Profiles | cell lines with high or low expression of FGFR2 gene relative to other cell lines from the BioGPS Cell Line Gene Expression Profiles dataset. | |
BioGPS Human Cell Type and Tissue Gene Expression Profiles | cell types and tissues with high or low expression of FGFR2 gene relative to other cell types and tissues from the BioGPS Human Cell Type and Tissue Gene Expression Profiles dataset. | |
BioGPS Mouse Cell Type and Tissue Gene Expression Profiles | cell types and tissues with high or low expression of FGFR2 gene relative to other cell types and tissues from the BioGPS Mouse Cell Type and Tissue Gene Expression Profiles dataset. | |
CCLE Cell Line Gene CNV Profiles | cell lines with high or low copy number of FGFR2 gene relative to other cell lines from the CCLE Cell Line Gene CNV Profiles dataset. | |
CCLE Cell Line Gene Expression Profiles | cell lines with high or low expression of FGFR2 gene relative to other cell lines from the CCLE Cell Line Gene Expression Profiles dataset. | |
CCLE Cell Line Gene Mutation Profiles | cell lines with FGFR2 gene mutations from the CCLE Cell Line Gene Mutation Profiles dataset. | |
CCLE Cell Line Proteomics | Cell lines associated with FGFR2 protein from the CCLE Cell Line Proteomics dataset. | |
CellMarker Gene-Cell Type Associations | cell types associated with FGFR2 gene from the CellMarker Gene-Cell Type Associations dataset. | |
ChEA Transcription Factor Binding Site Profiles | transcription factor binding site profiles with transcription factor binding evidence at the promoter of FGFR2 gene from the CHEA Transcription Factor Binding Site Profiles dataset. | |
ChEA Transcription Factor Targets | transcription factors binding the promoter of FGFR2 gene in low- or high-throughput transcription factor functional studies from the CHEA Transcription Factor Targets dataset. | |
ChEA Transcription Factor Targets 2022 | transcription factors binding the promoter of FGFR2 gene in low- or high-throughput transcription factor functional studies from the CHEA Transcription Factor Targets 2022 dataset. | |
ClinVar Gene-Phenotype Associations | phenotypes associated with FGFR2 gene from the curated ClinVar Gene-Phenotype Associations dataset. | |
CMAP Signatures of Differentially Expressed Genes for Small Molecules | small molecule perturbations changing expression of FGFR2 gene from the CMAP Signatures of Differentially Expressed Genes for Small Molecules dataset. | |
COMPARTMENTS Curated Protein Localization Evidence Scores | cellular components containing FGFR2 protein from the COMPARTMENTS Curated Protein Localization Evidence Scores dataset. | |
COMPARTMENTS Curated Protein Localization Evidence Scores 2025 | cellular components containing FGFR2 protein from the COMPARTMENTS Curated Protein Localization Evidence Scores 2025 dataset. | |
COMPARTMENTS Experimental Protein Localization Evidence Scores | cellular components containing FGFR2 protein in low- or high-throughput protein localization assays from the COMPARTMENTS Experimental Protein Localization Evidence Scores dataset. | |
COMPARTMENTS Experimental Protein Localization Evidence Scores 2025 | cellular components containing FGFR2 protein in low- or high-throughput protein localization assays from the COMPARTMENTS Experimental Protein Localization Evidence Scores 2025 dataset. | |
COMPARTMENTS Text-mining Protein Localization Evidence Scores | cellular components co-occuring with FGFR2 protein in abstracts of biomedical publications from the COMPARTMENTS Text-mining Protein Localization Evidence Scores dataset. | |
COMPARTMENTS Text-mining Protein Localization Evidence Scores 2025 | cellular components co-occuring with FGFR2 protein in abstracts of biomedical publications from the COMPARTMENTS Text-mining Protein Localization Evidence Scores 2025 dataset. | |
CORUM Protein Complexes | protein complexs containing FGFR2 protein from the CORUM Protein Complexes dataset. | |
COSMIC Cell Line Gene CNV Profiles | cell lines with high or low copy number of FGFR2 gene relative to other cell lines from the COSMIC Cell Line Gene CNV Profiles dataset. | |
COSMIC Cell Line Gene Mutation Profiles | cell lines with FGFR2 gene mutations from the COSMIC Cell Line Gene Mutation Profiles dataset. | |
CTD Gene-Chemical Interactions | chemicals interacting with FGFR2 gene/protein from the curated CTD Gene-Chemical Interactions dataset. | |
CTD Gene-Disease Associations | diseases associated with FGFR2 gene/protein from the curated CTD Gene-Disease Associations dataset. | |
dbGAP Gene-Trait Associations | traits associated with FGFR2 gene in GWAS and other genetic association datasets from the dbGAP Gene-Trait Associations dataset. | |
DeepCoverMOA Drug Mechanisms of Action | small molecule perturbations with high or low expression of FGFR2 protein relative to other small molecule perturbations from the DeepCoverMOA Drug Mechanisms of Action dataset. | |
DepMap CRISPR Gene Dependency | cell lines with fitness changed by FGFR2 gene knockdown relative to other cell lines from the DepMap CRISPR Gene Dependency dataset. | |
DEPOD Substrates of Phosphatases | phosphatases that dephosphorylate FGFR2 protein from the curated DEPOD Substrates of Phosphatases dataset. | |
DISEASES Curated Gene-Disease Association Evidence Scores | diseases involving FGFR2 gene from the DISEASES Curated Gene-Disease Assocation Evidence Scores dataset. | |
DISEASES Curated Gene-Disease Association Evidence Scores 2025 | diseases involving FGFR2 gene from the DISEASES Curated Gene-Disease Association Evidence Scores 2025 dataset. | |
DISEASES Experimental Gene-Disease Association Evidence Scores | diseases associated with FGFR2 gene in GWAS datasets from the DISEASES Experimental Gene-Disease Assocation Evidence Scores dataset. | |
DISEASES Experimental Gene-Disease Association Evidence Scores 2025 | diseases associated with FGFR2 gene in GWAS datasets from the DISEASES Experimental Gene-Disease Assocation Evidence Scores 2025 dataset. | |
DISEASES Text-mining Gene-Disease Association Evidence Scores | diseases co-occuring with FGFR2 gene in abstracts of biomedical publications from the DISEASES Text-mining Gene-Disease Assocation Evidence Scores dataset. | |
DISEASES Text-mining Gene-Disease Association Evidence Scores 2025 | diseases co-occuring with FGFR2 gene in abstracts of biomedical publications from the DISEASES Text-mining Gene-Disease Assocation Evidence Scores 2025 dataset. | |
DisGeNET Gene-Disease Associations | diseases associated with FGFR2 gene in GWAS and other genetic association datasets from the DisGeNET Gene-Disease Associations dataset. | |
DisGeNET Gene-Phenotype Associations | phenotypes associated with FGFR2 gene in GWAS and other genetic association datasets from the DisGeNET Gene-Phenoptype Associations dataset. | |
DrugBank Drug Targets | interacting drugs for FGFR2 protein from the curated DrugBank Drug Targets dataset. | |
ENCODE Histone Modification Site Profiles | histone modification site profiles with high histone modification abundance at FGFR2 gene from the ENCODE Histone Modification Site Profiles dataset. | |
ENCODE Transcription Factor Binding Site Profiles | transcription factor binding site profiles with transcription factor binding evidence at the promoter of FGFR2 gene from the ENCODE Transcription Factor Binding Site Profiles dataset. | |
ENCODE Transcription Factor Targets | transcription factors binding the promoter of FGFR2 gene in ChIP-seq datasets from the ENCODE Transcription Factor Targets dataset. | |
ESCAPE Omics Signatures of Genes and Proteins for Stem Cells | PubMedIDs of publications reporting gene signatures containing FGFR2 from the ESCAPE Omics Signatures of Genes and Proteins for Stem Cells dataset. | |
GAD Gene-Disease Associations | diseases associated with FGFR2 gene in GWAS and other genetic association datasets from the GAD Gene-Disease Associations dataset. | |
GAD High Level Gene-Disease Associations | diseases associated with FGFR2 gene in GWAS and other genetic association datasets from the GAD High Level Gene-Disease Associations dataset. | |
GDSC Cell Line Gene Expression Profiles | cell lines with high or low expression of FGFR2 gene relative to other cell lines from the GDSC Cell Line Gene Expression Profiles dataset. | |
GeneRIF Biological Term Annotations | biological terms co-occuring with FGFR2 gene in literature-supported statements describing functions of genes from the GeneRIF Biological Term Annotations dataset. | |
GeneSigDB Published Gene Signatures | PubMedIDs of publications reporting gene signatures containing FGFR2 from the GeneSigDB Published Gene Signatures dataset. | |
GEO Signatures of Differentially Expressed Genes for Diseases | disease perturbations changing expression of FGFR2 gene from the GEO Signatures of Differentially Expressed Genes for Diseases dataset. | |
GEO Signatures of Differentially Expressed Genes for Gene Perturbations | gene perturbations changing expression of FGFR2 gene from the GEO Signatures of Differentially Expressed Genes for Gene Perturbations dataset. | |
GEO Signatures of Differentially Expressed Genes for Kinase Perturbations | kinase perturbations changing expression of FGFR2 gene from the GEO Signatures of Differentially Expressed Genes for Kinase Perturbations dataset. | |
GEO Signatures of Differentially Expressed Genes for Small Molecules | small molecule perturbations changing expression of FGFR2 gene from the GEO Signatures of Differentially Expressed Genes for Small Molecules dataset. | |
GEO Signatures of Differentially Expressed Genes for Transcription Factor Perturbations | transcription factor perturbations changing expression of FGFR2 gene from the GEO Signatures of Differentially Expressed Genes for Transcription Factor Perturbations dataset. | |
GEO Signatures of Differentially Expressed Genes for Viral Infections | virus perturbations changing expression of FGFR2 gene from the GEO Signatures of Differentially Expressed Genes for Viral Infections dataset. | |
GlyGen Glycosylated Proteins | ligands (chemical) binding FGFR2 protein from the GlyGen Glycosylated Proteins dataset. | |
GO Biological Process Annotations 2015 | biological processes involving FGFR2 gene from the curated GO Biological Process Annotations 2015 dataset. | |
GO Biological Process Annotations 2023 | biological processes involving FGFR2 gene from the curated GO Biological Process Annotations 2023 dataset. | |
GO Biological Process Annotations 2025 | biological processes involving FGFR2 gene from the curated GO Biological Process Annotations2025 dataset. | |
GO Cellular Component Annotations 2015 | cellular components containing FGFR2 protein from the curated GO Cellular Component Annotations 2015 dataset. | |
GO Cellular Component Annotations 2023 | cellular components containing FGFR2 protein from the curated GO Cellular Component Annotations 2023 dataset. | |
GO Cellular Component Annotations 2025 | cellular components containing FGFR2 protein from the curated GO Cellular Component Annotations 2025 dataset. | |
GO Molecular Function Annotations 2015 | molecular functions performed by FGFR2 gene from the curated GO Molecular Function Annotations 2015 dataset. | |
GO Molecular Function Annotations 2023 | molecular functions performed by FGFR2 gene from the curated GO Molecular Function Annotations 2023 dataset. | |
GO Molecular Function Annotations 2025 | molecular functions performed by FGFR2 gene from the curated GO Molecular Function Annotations 2025 dataset. | |
GTEx eQTL 2025 | SNPs regulating expression of FGFR2 gene from the GTEx eQTL 2025 dataset. | |
GTEx Tissue Gene Expression Profiles | tissues with high or low expression of FGFR2 gene relative to other tissues from the GTEx Tissue Gene Expression Profiles dataset. | |
GTEx Tissue Gene Expression Profiles 2023 | tissues with high or low expression of FGFR2 gene relative to other tissues from the GTEx Tissue Gene Expression Profiles 2023 dataset. | |
GTEx Tissue Sample Gene Expression Profiles | tissue samples with high or low expression of FGFR2 gene relative to other tissue samples from the GTEx Tissue Sample Gene Expression Profiles dataset. | |
GTEx Tissue-Specific Aging Signatures | tissue samples with high or low expression of FGFR2 gene relative to other tissue samples from the GTEx Tissue-Specific Aging Signatures dataset. | |
Guide to Pharmacology Chemical Ligands of Receptors | ligands (chemical) binding FGFR2 receptor from the curated Guide to Pharmacology Chemical Ligands of Receptors dataset. | |
GWAS Catalog SNP-Phenotype Associations | phenotypes associated with FGFR2 gene in GWAS datasets from the GWAS Catalog SNP-Phenotype Associations dataset. | |
GWASdb SNP-Disease Associations | diseases associated with FGFR2 gene in GWAS and other genetic association datasets from the GWASdb SNP-Disease Associations dataset. | |
GWASdb SNP-Phenotype Associations | phenotypes associated with FGFR2 gene in GWAS datasets from the GWASdb SNP-Phenotype Associations dataset. | |
Heiser et al., PNAS, 2011 Cell Line Gene Expression Profiles | cell lines with high or low expression of FGFR2 gene relative to other cell lines from the Heiser et al., PNAS, 2011 Cell Line Gene Expression Profiles dataset. | |
HMDB Metabolites of Enzymes | interacting metabolites for FGFR2 protein from the curated HMDB Metabolites of Enzymes dataset. | |
HPA Cell Line Gene Expression Profiles | cell lines with high or low expression of FGFR2 gene relative to other cell lines from the HPA Cell Line Gene Expression Profiles dataset. | |
HPA Tissue Gene Expression Profiles | tissues with high or low expression of FGFR2 gene relative to other tissues from the HPA Tissue Gene Expression Profiles dataset. | |
HPA Tissue Protein Expression Profiles | tissues with high or low expression of FGFR2 protein relative to other tissues from the HPA Tissue Protein Expression Profiles dataset. | |
HPA Tissue Sample Gene Expression Profiles | tissue samples with high or low expression of FGFR2 gene relative to other tissue samples from the HPA Tissue Sample Gene Expression Profiles dataset. | |
HPM Cell Type and Tissue Protein Expression Profiles | cell types and tissues with high or low expression of FGFR2 protein relative to other cell types and tissues from the HPM Cell Type and Tissue Protein Expression Profiles dataset. | |
HPO Gene-Disease Associations | phenotypes associated with FGFR2 gene by mapping known disease genes to disease phenotypes from the HPO Gene-Disease Associations dataset. | |
Hub Proteins Protein-Protein Interactions | interacting hub proteins for FGFR2 from the curated Hub Proteins Protein-Protein Interactions dataset. | |
HuBMAP ASCT+B Annotations | cell types associated with FGFR2 gene from the HuBMAP ASCT+B dataset. | |
HuBMAP ASCT+B Augmented with RNA-seq Coexpression | cell types associated with FGFR2 gene from the HuBMAP ASCT+B Augmented with RNA-seq Coexpression dataset. | |
HuBMAP Azimuth Cell Type Annotations | cell types associated with FGFR2 gene from the HuBMAP Azimuth Cell Type Annotations dataset. | |
HuGE Navigator Gene-Phenotype Associations | phenotypes associated with FGFR2 gene by text-mining GWAS publications from the HuGE Navigator Gene-Phenotype Associations dataset. | |
InterPro Predicted Protein Domain Annotations | protein domains predicted for FGFR2 protein from the InterPro Predicted Protein Domain Annotations dataset. | |
JASPAR Predicted Transcription Factor Targets | transcription factors regulating expression of FGFR2 gene predicted using known transcription factor binding site motifs from the JASPAR Predicted Transcription Factor Targets dataset. | |
KEA Substrates of Kinases | kinases that phosphorylate FGFR2 protein from the curated KEA Substrates of Kinases dataset. | |
KEGG Pathways | pathways involving FGFR2 protein from the KEGG Pathways dataset. | |
Kinase Library Tyrosine Kinome Atlas | kinases that phosphorylate FGFR2 protein from the Kinase Library Tyrosine Kinome Atlas dataset. | |
Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene CNV Profiles | cell lines with high or low copy number of FGFR2 gene relative to other cell lines from the Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene CNV Profiles dataset. | |
Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene Mutation Profiles | cell lines with FGFR2 gene mutations from the Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene Mutation Profiles dataset. | |
KnockTF Gene Expression Profiles with Transcription Factor Perturbations | transcription factor perturbations changing expression of FGFR2 gene from the KnockTF Gene Expression Profiles with Transcription Factor Perturbations dataset. | |
LINCS KinomeScan Kinase Inhibitor Targets | small molecules inhibiting FGFR2 kinase from the KinomeScan Kinase Inhibitor Targets dataset. | |
LINCS L1000 CMAP Chemical Perturbation Consensus Signatures | small molecule perturbations changing expression of FGFR2 gene from the LINCS L1000 CMAP Chemical Perturbations Consensus Signatures dataset. | |
LINCS L1000 CMAP CRISPR Knockout Consensus Signatures | gene perturbations changing expression of FGFR2 gene from the LINCS L1000 CMAP CRISPR Knockout Consensus Signatures dataset. | |
LINCS L1000 CMAP Signatures of Differentially Expressed Genes for Small Molecules | small molecule perturbations changing expression of FGFR2 gene from the LINCS L1000 CMAP Signatures of Differentially Expressed Genes for Small Molecules dataset. | |
LOCATE Curated Protein Localization Annotations | cellular components containing FGFR2 protein in low- or high-throughput protein localization assays from the LOCATE Curated Protein Localization Annotations dataset. | |
LOCATE Predicted Protein Localization Annotations | cellular components predicted to contain FGFR2 protein from the LOCATE Predicted Protein Localization Annotations dataset. | |
MGI Mouse Phenotype Associations 2023 | phenotypes of transgenic mice caused by FGFR2 gene mutations from the MGI Mouse Phenotype Associations 2023 dataset. | |
MiRTarBase microRNA Targets | microRNAs targeting FGFR2 gene in low- or high-throughput microRNA targeting studies from the MiRTarBase microRNA Targets dataset. | |
MotifMap Predicted Transcription Factor Targets | transcription factors regulating expression of FGFR2 gene predicted using known transcription factor binding site motifs from the MotifMap Predicted Transcription Factor Targets dataset. | |
MoTrPAC Rat Endurance Exercise Training | tissue samples with high or low expression of FGFR2 gene relative to other tissue samples from the MoTrPAC Rat Endurance Exercise Training dataset. | |
MPO Gene-Phenotype Associations | phenotypes of transgenic mice caused by FGFR2 gene mutations from the MPO Gene-Phenotype Associations dataset. | |
MSigDB Signatures of Differentially Expressed Genes for Cancer Gene Perturbations | gene perturbations changing expression of FGFR2 gene from the MSigDB Signatures of Differentially Expressed Genes for Cancer Gene Perturbations dataset. | |
NIBR DRUG-seq U2OS MoA Box Gene Expression Profiles | drug perturbations changing expression of FGFR2 gene from the NIBR DRUG-seq U2OS MoA Box dataset. | |
NURSA Protein Complexes | protein complexs containing FGFR2 protein recovered by IP-MS from the NURSA Protein Complexes dataset. | |
OMIM Gene-Disease Associations | phenotypes associated with FGFR2 gene from the curated OMIM Gene-Disease Associations dataset. | |
PANTHER Pathways | pathways involving FGFR2 protein from the PANTHER Pathways dataset. | |
Pathway Commons Protein-Protein Interactions | interacting proteins for FGFR2 from the Pathway Commons Protein-Protein Interactions dataset. | |
PerturbAtlas Signatures of Differentially Expressed Genes for Gene Perturbations | gene perturbations changing expression of FGFR2 gene from the PerturbAtlas Signatures of Differentially Expressed Genes for Gene Perturbations dataset. | |
PerturbAtlas Signatures of Differentially Expressed Genes for Mouse Gene Perturbations | gene perturbations changing expression of FGFR2 gene from the PerturbAtlas Signatures of Differentially Expressed Genes for Gene Perturbations dataset. | |
PFOCR Pathway Figure Associations 2023 | pathways involving FGFR2 protein from the PFOCR Pathway Figure Associations 2023 dataset. | |
PFOCR Pathway Figure Associations 2024 | pathways involving FGFR2 protein from the Wikipathways PFOCR 2024 dataset. | |
Phosphosite Textmining Biological Term Annotations | biological terms co-occuring with FGFR2 protein in abstracts of publications describing phosphosites from the Phosphosite Textmining Biological Term Annotations dataset. | |
PhosphoSitePlus Substrates of Kinases | kinases that phosphorylate FGFR2 protein from the curated PhosphoSitePlus Substrates of Kinases dataset. | |
PID Pathways | pathways involving FGFR2 protein from the PID Pathways dataset. | |
Reactome Pathways 2014 | pathways involving FGFR2 protein from the Reactome Pathways dataset. | |
Reactome Pathways 2024 | pathways involving FGFR2 protein from the Reactome Pathways 2024 dataset. | |
Roadmap Epigenomics Cell and Tissue DNA Methylation Profiles | cell types and tissues with high or low DNA methylation of FGFR2 gene relative to other cell types and tissues from the Roadmap Epigenomics Cell and Tissue DNA Methylation Profiles dataset. | |
Roadmap Epigenomics Cell and Tissue Gene Expression Profiles | cell types and tissues with high or low expression of FGFR2 gene relative to other cell types and tissues from the Roadmap Epigenomics Cell and Tissue Gene Expression Profiles dataset. | |
Roadmap Epigenomics Histone Modification Site Profiles | histone modification site profiles with high histone modification abundance at FGFR2 gene from the Roadmap Epigenomics Histone Modification Site Profiles dataset. | |
RummaGEO Drug Perturbation Signatures | drug perturbations changing expression of FGFR2 gene from the RummaGEO Drug Perturbation Signatures dataset. | |
RummaGEO Gene Perturbation Signatures | gene perturbations changing expression of FGFR2 gene from the RummaGEO Gene Perturbation Signatures dataset. | |
Sanger Dependency Map Cancer Cell Line Proteomics | cell lines associated with FGFR2 protein from the Sanger Dependency Map Cancer Cell Line Proteomics dataset. | |
Tabula Sapiens Gene-Cell Associations | cell types with high or low expression of FGFR2 gene relative to other cell types from the Tabula Sapiens Gene-Cell Associations dataset. | |
TargetScan Predicted Conserved microRNA Targets | microRNAs regulating expression of FGFR2 gene predicted using conserved miRNA seed sequences from the TargetScan Predicted Conserved microRNA Targets dataset. | |
TargetScan Predicted Nonconserved microRNA Targets | microRNAs regulating expression of FGFR2 gene predicted using nonconserved miRNA seed sequences from the TargetScan Predicted Nonconserved microRNA Targets dataset. | |
TCGA Signatures of Differentially Expressed Genes for Tumors | tissue samples with high or low expression of FGFR2 gene relative to other tissue samples from the TCGA Signatures of Differentially Expressed Genes for Tumors dataset. | |
TISSUES Curated Tissue Protein Expression Evidence Scores | tissues with high expression of FGFR2 protein from the TISSUES Curated Tissue Protein Expression Evidence Scores dataset. | |
TISSUES Curated Tissue Protein Expression Evidence Scores 2025 | tissues with high expression of FGFR2 protein from the TISSUES Curated Tissue Protein Expression Evidence Scores 2025 dataset. | |
TISSUES Experimental Tissue Protein Expression Evidence Scores | tissues with high expression of FGFR2 protein in proteomics datasets from the TISSUES Experimental Tissue Protein Expression Evidence Scores dataset. | |
TISSUES Experimental Tissue Protein Expression Evidence Scores 2025 | tissues with high expression of FGFR2 protein in proteomics datasets from the TISSUES Experimental Tissue Protein Expression Evidence Scores 2025 dataset. | |
TISSUES Text-mining Tissue Protein Expression Evidence Scores | tissues co-occuring with FGFR2 protein in abstracts of biomedical publications from the TISSUES Text-mining Tissue Protein Expression Evidence Scores dataset. | |
TISSUES Text-mining Tissue Protein Expression Evidence Scores 2025 | tissues co-occuring with FGFR2 protein in abstracts of biomedical publications from the TISSUES Text-mining Tissue Protein Expression Evidence Scores 2025 dataset. | |
WikiPathways Pathways 2014 | pathways involving FGFR2 protein from the Wikipathways Pathways 2014 dataset. | |
WikiPathways Pathways 2024 | pathways involving FGFR2 protein from the WikiPathways Pathways 2024 dataset. | |