Рецепторы фактора роста фибробластов при злокачественных опухолях

Автор: Федянин М.Ю., Хмелькова Д.Н., Серебрийская Т.С., Никольская Т.А., Тюляндин С.А.

Журнал: Злокачественные опухоли @malignanttumors

Рубрика: Фундаментальная онкология и экспериментальная медицина

Статья в выпуске: 4 (11), 2014 года.

Бесплатный доступ

Одними из наиболее исследуемых в настоящее время в онкологии маркеров являются рецепторы к фактору роста фибробластов, а также лиганды к нему. В данном обзоре мы сконцентрируемся на молекулярных процессах, возникающих при активации рецепторов к фактору роста фибробластов. А также рассмотрим, с какой частотой встречаются нарушения экспрессии компонентов сигнального пути данного рецептора при различных онкологических заболеваниях.

Солидные опухоли, рецептор к фактору роста фибробластов, предикторные и прогностические факторы

Короткий адрес: https://sciup.org/14045513

IDR: 14045513

Список литературы Рецепторы фактора роста фибробластов при злокачественных опухолях

  • Itoh N., The Fgf families in humans, mice, and zebrafish: their evolutional processes and roles in development, metabolism, and disease. Biol Pharm Bull, 2007. 30 (10): p. 1819-1825
  • Ornitz D. M., Xu J., Colvin J. S., McEwen D.G., MacArthur C.A., Coulier F., et al., Receptor specificity of the fibroblast growth factor family. J Biol Chem, 1996. 271 (25): p. 15292-15297
  • Turner N., Grose R., Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer, 2010. 10 (2): p. 116-129
  • Katoh M., Nakagama H., FGF receptors: cancer biology and therapeutics. Med Res Rev, 2014. 34 (2): p. 280-300
  • Beenken A., Mohammadi M., The FGF family: biology, pathophysiology and therapy. Nat Rev Drug Discov, 2009. 8 (3): p. 235-253
  • Bryant D. M., Stow J. L., Nuclear translocation of cell-surface receptors: lessons from fibroblast growth factor. Traffic, 2005. 6 (10): p. 947-954
  • Imamura T., Engleka K., Zhan X., Tokita Y., Forough R., Roeder D., et al., Recovery of mitogenic activity of a growth factor mutant with a nuclear translocation sequence. Science, 1990. 249 (4976): p. 1567-1570
  • Bikfalvi A., Klein S., Pintucci G., Rifkin D. B., Biological roles of fibroblast growth factor-2. Endocr Rev, 1997. 18 (1): p. 26-45
  • Miller D. L., Ortega S., Bashayan O., Basch R., Basilico C., Compensation by fibroblast growth factor 1 (FGF1) does not account for the mild phenotypic defects observed in FGF2 null mice. Mol Cell Biol, 2000. 20 (6): p. 2260-2268
  • Dono R., Texido G., Dussel R., Ehmke H., Zeller R., Impaired cerebral cortex development and blood pressure regulation in FGF-2-deficient mice. EMBO J, 1998. 17 (15): p. 4213-4225
  • Simons M., Horowitz A., Syndecan-4-mediated signalling. Cell Signal, 2001. 13 (12): p. 855-862
  • Ong S. H., Guy G. R., Hadari Y. R., 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 (3): p. 979-989
  • Ong S. H., Hadari Y. R., Gotoh N., Guy G. R., Schlessinger J., Lax I., Stimulation of phosphatidylinositol 3-kinase by fibroblast growth factor receptors is mediated by coordinated recruitment of multiple docking proteins. Proc Natl Acad Sci U S A, 2001. 98 (11): p. 6074-6079
  • Lin N., Chen S., Pan W., Xu L., Hu K., Xu R., NP603, a novel and potent inhibitor of FGFR1 tyrosine kinase, inhibits hepatic stellate cell proliferation and ameliorates hepatic fibrosis in rats. Am J Physiol Cell Physiol, 2011. 301 (2): p. C469-477
  • Meloche S., Pouyssegur J., The ERK1/2 mitogen-activated protein kinase pathway as a master regulator of the G1-to S-phase transition. Oncogene, 2007. 26 (22): p. 3227-3239
  • Koziczak M., Holbro T., Hynes N. E., Blocking of FGFR signaling inhibits breast cancer cell proliferation through downregulation of D-type cyclins. Oncogene, 2004. 23 (20): p. 3501-3508
  • Lee J. G., Kay E. P., PI 3-kinase/Rac1 and ERK1/2 regulate FGF-2-mediated cell proliferation through phosphorylation of p27 at Ser10 by KIS and at Thr187 by Cdc25A/Cdk2. Invest Ophthalmol Vis Sci, 2011. 52 (1): p. 417-426
  • Miyake M., Ishii M., Koyama N., Kawashima K., Kodama T., Anai S., et al., 1-tert-butyl-3- pyrimidin-7-yl] -urea (PD173074), a selective tyrosine kinase inhibitor of fibroblast growth factor receptor-3 (FGFR3), inhibits cell proliferation of bladder cancer carrying the FGFR3 gene mutation along with up-regulation of p27/Kip1 and G1/G0 arrest. J Pharmacol Exp Ther, 2010. 332 (3): p. 795-802
  • Pardo O. E., Arcaro A., Salerno G., Raguz S., Downward J., Seckl M. J., Fibroblast growth factor-2 induces translational regulation of Bcl-XL and Bcl-2 via a MEK-dependent pathway: correlation with resistance to etoposide-induced apoptosis. J Biol Chem, 2002. 277 (14): p.12040-12046
  • Goetz R., Mohammadi M., Exploring mechanisms of FGF signalling through the lens of structural biology. Nat Rev Mol Cell Biol, 2013. 14 (3): p. 166-180
  • Balmanno K., Cook S. J., Tumour cell survival signalling by the ERK1/2 pathway. Cell Death Differ, 2009. 16 (3): p. 368-377
  • Mohammadi M., Honegger A. M., Rotin D., Fischer R., Bellot F., Li W., et al A tyrosine-phosphorylated carboxy-terminal peptide of the fibroblast growth factor receptor (Flg) is a binding site for the SH2 domain of phospholipase C-gamma 1. Mol Cell Biol, 1991. 11 (10): p. 5068-5078.
  • Hart K. C., Robertson S. C., Kanemitsu M. Y., Meyer A. N., Tynan J. A., Donoghue D. J., Transformation and Stat activation by derivatives of FGFR1, FGFR3, and FGFR4. Oncogene, 2000. 19 (29): p. 3309-3320
  • Smith T. G., Karlsson M., Lunn J. S., Eblaghie M. C., Keenan I. D., Farrell E. R., et al., Negative feedback predominates over cross-regulation to control ERK MAPK activity in response to FGF signalling in embryos. FEBS Lett, 2006. 580 (17): p. 4242-4245
  • Mason J. M., Morrison D. J., Basson M. A., Licht J. D., Sprouty proteins: multifaceted negative-feedback regulators of receptor tyrosine kinase signaling. Trends Cell Biol, 2006. 16 (1): p. 45-54
  • Kovalenko D., Yang X., Nadeau R. J., Harkins L. K., Friesel R., Sef inhibits fibroblast growth factor signaling by inhibiting FGFR1 tyrosine phosphorylation and subsequent ERK activation. J Biol Chem, 2003. 278 (16): p. 14087-14091
  • Baird A., Bhlen P., Fibroblast growth factors, in Peptide growth factors and their receptors I. 1990, Springer. p. 369-418
  • Matthews D. J., Gerritsen M. E., Targeting protein kinases for cancer therapy. 2011: John Wiley & Sons
  • Iyer G., Milowsky M. I., Fibroblast growth factor receptor-3 in urothelial tumorigenesis. Urol Oncol, 2013. 31 (3): p. 303-311
  • Cappellen D., De Oliveira C., Ricol D., de Medina S., Bourdin J., Sastre-Garau X., et al., Frequent activating mutations of FGFR3 in human bladder and cervix carcinomas. Nat Genet, 1999. 23 (1): p. 18-20
  • Hernandez S., Lopez-Knowles E., Lloreta J., Kogevinas M., Amoros A., Tardon A., et al., Prospective study of FGFR3 mutations as a prognostic factor in nonmuscle invasive urothelial bladder carcinomas. J Clin Oncol, 2006. 24 (22): p. 3664-3671
  • di Martino E., Tomlinson D. C., Knowles M. A., A Decade of FGF Receptor Research in Bladder Cancer: Past, Present, and Future Challenges. Adv Urol, 2012. 2012: p. 429213
  • Welm B. E., Freeman K. W., Chen M., Contreras A., Spencer D. M., Rosen J. M., Inducible dimerization of FGFR1: development of a mouse model to analyze progressive transformation of the mammary gland. J Cell Biol, 2002. 157 (4): p. 703-714
  • Elbauomy Elsheikh S., Green A. R., Lambros M. B., Turner N. C., Grainge M. J., Powe D., et al., FGFR1 amplification in breast carcinomas: a chromogenic in situ hybridisation analysis. Breast Cancer Res, 2007. 9 (2): p. R23
  • Turner N., Pearson A., Sharpe R., Lambros M., Geyer F., Lopez-Garcia M.A., et al., FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer. Cancer Res, 2010. 70 (5): p. 2085-2094
  • Xian W., Pappas L., Pandya D., Selfors L. M., Derksen P. W., de Bruin M., et al., Fibroblast growth factor receptor 1-transformed mammary epithelial cells are dependent on RSK activity for growth and survival. Cancer Res, 2009. 69 (6): p. 2244-2251
  • Brunello E., Brunelli M., Bogina G., Calio A., Manfrin E., Nottegar A., et al., FGFR-1 amplification in metastatic lymph-nodal and haematogenous lobular breast carcinoma. J Exp Clin Cancer Res, 2012. 31: p. 103
  • Jang M., Kim E., Choi Y., Lee H., Kim Y., Kim J., et al., FGFR1 is amplified during the progression of in situ to invasive breast carcinoma. Breast Cancer Res, 2012. 14 (4): p. R115
  • Reis-Filho J.S., Simpson P. T., Turner N. C., Lambros M. B., Jones C., Mackay A., et al., FGFR1 emerges as a potential therapeutic target for lobular breast carcinomas. Clin Cancer Res, 2006. 12 (22): p. 6652-6662
  • Courjal F., Cuny M., Simony-Lafontaine J., Louason G., Speiser P., Zeillinger R., et al., Mapping of DNA amplifications at 15 chromosomal localizations in 1875 breast tumors: definition of phenotypic groups. Cancer Res, 1997. 57 (19): p. 4360-4367
  • Cuny M., Kramar A., Courjal F., Johannsdottir V., Iacopetta B., Fontaine H., et al., Relating genotype and phenotype in breast cancer: an analysis of the prognostic significance of amplification at eight different genes or loci and of p 53 mutations. Cancer Res, 2000. 60 (4): p. 1077-1083
  • Ray M. E., Yang Z. Q., Albertson D., Kleer C. G., Washburn J. G., Macoska J. A., et al., Genomic and expression analysis of the 8p11-12 amplicon in human breast cancer cell lines. Cancer Res, 2004. 64 (1): p. 40-47
  • Easton D. F., Pooley K. A., Dunning A. M., Pharoah P. D., Thompson D., Ballinger D. G., et al., Genome-wide association study identifies novel breast cancer susceptibility loci. Nature, 2007. 447 (7148): p. 1087-1093
  • Boyarskikh U. A., Zarubina N. A., Biltueva J. A., Sinkina T. V., Voronina E. N., Lazarev A. F., et al., Association of FGFR2 gene polymorphisms with the risk of breast cancer in population of West Siberia. Eur J Hum Genet, 2009. 17 (12): p. 1688-1691
  • Fanale D., Amodeo V., Corsini L. R., Rizzo S., Bazan V., Russo A., Breast cancer genome-wide association studies: there is strength in numbers. Oncogene, 2012. 31 (17): p. 2121-2128
  • Sun S., Jiang Y., Zhang G., Song H., Zhang X., Zhang Y., et al., Increased expression of fibroblastic growth factor receptor 2 is correlated with poor prognosis in patients with breast cancer. J Surg Oncol, 2012. 105 (8): p. 773-779
  • Sibley K., Stern P., Knowles M. A., Frequency of fibroblast growth factor receptor 3 mutations in sporadic tumours. Oncogene, 2001. 20 (32): p. 4416-4418
  • Greenman C., Stephens P., Smith R., Dalgliesh G. L., Hunter C., Bignell G., et al., Patterns of somatic mutation in human cancer genomes. Nature, 2007. 446 (7132): p. 153-158
  • Kuroso K., Imai Y., Kobayashi M., Yanagimoto K., Suzuki T., Kojima M., et al., Immunohistochemical detection of fibroblast growth factor receptor 3 in human breast cancer: correlation with clinicopathological/molecular parameters and prognosis. Pathobiology, 2010. 77 (5): p. 231-240
  • Tomlinson D. C., Knowles M. A., Speirs V., Mechanisms of FGFR3 actions in endocrine resistant breast cancer. Int J Cancer, 2012. 130 (12): p. 2857-2866
  • Lafitte M., Moranvillier I., Garcia S., Peuchant E., Iovanna J., Rousseau B., et al., FGFR3 has tumor suppressor properties in cells with epithelial phenotype. Mol Cancer, 2013. 12: p. 83
  • Jaakkola S., Salmikangas P., Nylund S., Partanen J., Armstrong E., Pyrhonen S., et al., Amplification of fgfr4 gene in human breast and gynecological cancers. Int J Cancer, 1993. 54 (3): p. 378-382
  • Meijer D., Sieuwerts A. M., Look M. P., van Agthoven T., Foekens J. A., Dorssers L. C., Fibroblast growth factor receptor 4 predicts failure on tamoxifen therapy in patients with recurrent breast cancer. Endocr Relat Cancer, 2008. 15 (1): p. 101-111
  • Roidl A., Berger H. J., Kumar S., Bange J., Knyazev P., Ullrich A., Resistance to chemotherapy is associated with fibroblast growth factor receptor 4 up-regulation. Clin Cancer Res, 2009. 15 (6): p. 2058-2066
  • Becker N., Nieters A., Chang-Claude J., The fibroblast growth factor receptor gene Arg388 allele is not associated with early lymph node metastasis of breast cancer. Cancer Epidemiol Biomarkers Prev, 2003. 12 (6): p. 582-583
  • Jezequel P., Campion L., Joalland M. P., Millour M., Dravet F., Classe J. M., et al., G388R mutation of the FGFR4 gene is not relevant to breast cancer prognosis. Br J Cancer, 2004. 90 (1): p. 189-193
  • Spinola M., Leoni V. P., Tanuma J., Pettinicchio A., Frattini M., Signoroni S., et al., FGFR4 Gly388Arg polymorphism and prognosis of breast and colorectal cancer. Oncol Rep, 2005. 14 (2): p. 415-419
  • Moffa A. B., Tannheimer S. L., Ethier S. P., Transforming potential of alternatively spliced variants of fibroblast growth factor receptor 2 in human mammary epithelial cells. Mol Cancer Res, 2004. 2 (11): p. 643-652
  • Zisman-Rozen S., Fink D., Ben-Izhak O., Fuchs Y., Brodski A., Kraus M. H., et al., Downregulation of Sef, an inhibitor of receptor tyrosine kinase signaling, is common to a variety of human carcinomas. Oncogene, 2007. 26 (41): p. 6093-6098
  • McLeskey S.W., Zhang L., El-Ashry D., Trock B. J., Lopez C. A., Kharbanda S., et al., Tamoxifen-resistant fibroblast growth factor-transfected MCF-7 cells are cross-resistant in vivo to the antiestrogen ICI 182,780 and two aromatase inhibitors. Clin Cancer Res, 1998. 4 (3): p. 697-711
  • Ding L., Getz G., Wheeler D. A., Mardis E. R., McLellan M.D., Cibulskis K., et al., Somatic mutations affect key pathways in lung adenocarcinoma. Nature, 2008. 455 (7216): p. 1069-1075
  • Marks J. L., McLellan M.D., Zakowski M. F., Lash A. E., Kasai Y., Broderick S., et al., Mutational analysis of EGFR and related signaling pathway genes in lung adenocarcinomas identifies a novel somatic kinase domain mutation in FGFR4. PLoS One, 2007. 2 (5): p. e426
  • Weiss J., Sos M. L., Seidel D., Peifer M., Zander T., Heuckmann J. M., et al., Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci Transl Med, 2010. 2 (62): p. 62ra93.
  • Thomson S., Petti F., Sujka-Kwok I., Epstein D., Haley J. D., Kinase switching in mesenchymal-like non-small cell lung cancer lines contributes to EGFR inhibitor resistance through pathway redundancy. Clin Exp Metastasis, 2008. 25 (8): p. 843-854
  • Spinola M., Leoni V., Pignatiello C., Conti B., Ravagnani F., Pastorino U., et al., Functional FGFR4 Gly388Arg polymorphism predicts prognosis in lung adenocarcinoma patients. J Clin Oncol, 2005. 23 (29): p. 7307-7311
  • Falvella F. S., Frullanti E., Galvan A., Spinola M., Noci S., De Cecco L., et al., FGFR4 Gly388Arg polymorphism may affect the clinical stage of patients with lung cancer by modulating the transcriptional profile of normal lung. Int J Cancer, 2009. 124 (12): p. 2880-2885
  • Matakidou A., El Galta R., Rudd M. F., Webb E. L., Bridle H., Eisen T., et al., Further observations on the relationship between the FGFR4 Gly388Arg polymorphism and lung cancer prognosis. Br J Cancer, 2007. 96 (12): p. 1904-1907
  • Sasaki H., Okuda K., Kawano O., Yukiue H., Yano M., Fujii Y., Fibroblast growth factor receptor 4 mutation and polymorphism in Japanese lung cancer. Oncol Rep, 2008. 20 (5): p. 1125-1130
  • Shou Y., Hirano T., Gong Y., Kato Y., Yoshida K., Ohira T., et al., Influence of angiogenetic factors and matrix metalloproteinases upon tumour progression in non-small-cell lung cancer. Br J Cancer, 2001. 85 (11): p. 1706-1712
  • Takanami I., Tanaka F., Hashizume T., Kikuchi K., Yamamoto Y., Yamamoto T., et al., The basic fibroblast growth factor and its receptor in pulmonary adenocarcinomas: an investigation of their expression as prognostic markers. Eur J Cancer, 1996. 32A (9): p. 1504-1509
  • Iwasaki A., Kuwahara M., Yoshinaga Y., Shirakusa T., Basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) levels, as prognostic indicators in NSCLC. Eur J Cardiothorac Surg, 2004. 25 (3): p. 443-448
  • Brattstrom D., Bergqvist M., Larsson A., Holmertz J., Hesselius P., Rosenberg L., et al., Basic fibroblast growth factor and vascular endothelial growth factor in sera from non-small cell lung cancer patients. Anticancer Res, 1998. 18 (2A): p. 1123-1127
  • Isa S., Kawaguchi T., Teramukai S., Minato K., Ohsaki Y., Shibata K., et al., Serum osteopontin levels are highly prognostic for survival in advanced non-small cell lung cancer: results from JMTO LC 0004. J Thorac Oncol, 2009. 4 (9): p. 1104-1110
  • Brattstrom D., Bergqvist M., Hesselius P., Larsson A., Lamberg K., Wernlund J., et al., Elevated preoperative serum levels of angiogenic cytokines correlate to larger primary tumours and poorer survival in non-small cell Lung Cancer patients. Lung Cancer, 2002. 37 (1): p. 57-63
  • Donnem T., Al-Shibli K., Al-Saad S., Busund L. T., Bremnes R. M., Prognostic impact of fibroblast growth factor 2 in non-small cell lung cancer: coexpression with VEGFR-3 and PDGF-B predicts poor survival. J Thorac Oncol, 2009. 4 (5): p. 578-585
  • Bremnes R. M., Camps C., Sirera R., Angiogenesis in non-small cell Lung Cancer: the prognostic impact of neoangiogenesis and the cytokines VEGF and bFGF in tumours and blood. Lung Cancer, 2006. 51 (2): p. 143-158
  • Kojima H., Shijubo N., Abe S., Thymidine phosphorylase and vascular endothelial growth factor in patients with Stage I lung adenocarcinoma. Cancer, 2002. 94 (4): p. 1083-1093
  • Oki M., Yamamoto H., Taniguchi H., Adachi Y., Imai K., Shinomura Y., Overexpression of the receptor tyrosine kinase EphA4 in human gastric cancers. World J Gastroenterol, 2008. 14 (37): p. 5650-5656
  • Yokota J., Yamamoto T., Miyajima N., Toyoshima K., Nomura N., Sakamoto H., et al., Genetic alterations of the c-erbB-2 Oncogene occur frequently in tubular adenocarcinoma of the stomach and are often accompanied by amplification of the v-erbA homologue. Oncogene, 1988. 2 (3): p. 283-287
  • Yashiro M., Nishioka N., Hirakawa K., K-ras mutation influences macroscopic features of gastric carcinoma. J Surg Res, 2005. 124 (1): p. 74-78
  • Kuniyasu H., Yasui W., Kitadai Y., Yokozaki H., Ito H., Tahara E., Frequent amplification of the c-met gene in scirrhous type stomach cancer. Biochem Biophys Res Commun, 1992. 189 (1): p. 227-232
  • Tsujimoto H., Sugihara H., Hagiwara A., Hattori T., Amplification of growth factor receptor genes and DNA ploidy pattern in the progression of gastric cancer. Virchows Arch, 1997. 431 (6): p. 383-389
  • Hattori Y., Odagiri H., Nakatani H., Miyagawa K., Naito K., Sakamoto H., et al., K-sam, an amplified gene in stomach cancer, is a member of the heparin-binding growth factor receptor genes. Proc Natl Acad Sci U S A, 1990. 87 (15): p. 5983-5987
  • Yoshida T., Sakamoto H., Terada M., Amplified genes in cancer in upper digestive tract. Semin Cancer Biol, 1993. 4 (1): p. 33-40
  • Jang J. H., Shin K. H., Park J. G., Mutations in fibroblast growth factor receptor 2 and fibroblast growth factor receptor 3 genes associated with human gastric and colorectal cancers. Cancer Res, 2001. 61 (9): p. 3541-3543
  • Kunii K., Davis L., Gorenstein J., Hatch H., Yashiro M., Di Bacco A., et al., FGFR2-amplified gastric cancer cell lines require FGFR2 and Erbb3 signaling for growth and survival. Cancer Res, 2008. 68 (7): p. 2340-2348
  • Nakazawa K., Yashiro M., Hirakawa K., Keratinocyte growth factor produced by gastric fibroblasts specifically stimulates proliferation of cancer cells from scirrhous gastric carcinoma. Cancer Res, 2003. 63 (24): p. 8848-8852
  • Cha J. Y., Maddileti S., Mitin N., Harden T. K., Der C. J., Aberrant receptor internalization and enhanced FRS2-dependent signaling contribute to the transforming activity of the fibroblast growth factor receptor 2 IIIb C3 isoform. J Biol Chem, 2009. 284 (10): p. 6227-6240
  • Shin E. Y., Lee B. H., Yang J. H., Shin K. S., Lee G. K., Yun H. Y., et al., Up-regulation and co-expression of fibroblast growth factor receptors in human gastric cancer. J Cancer Res Clin Oncol, 2000. 126 (9): p. 519-528
  • Murase H., Inokuchi M., Takagi Y., Kato K., Kojima K., Sugihara K., Prognostic significance of the co-overexpression of fibroblast growth factor receptors 1, 2 and 4 in gastric cancer. Mol Clin Oncol, 2014. 2 (4): p. 509-517
  • Ye Y., Shi Y., Zhou Y., Du C., Wang C., Zhan H., et al., The fibroblast growth factor receptor-4 Arg388 allele is associated with gastric cancer progression. Ann Surg Oncol, 2010. 17 (12): p. 3354-3361
  • Yoshino M., Ishiwata T., Watanabe M., Komine O., Shibuya T., Tokunaga A., et al., Keratinocyte growth factor receptor expression in normal colorectal epithelial cells and differentiated type of colorectal cancer. Oncol Rep, 2005. 13 (2): p. 247-252
  • Otte J. M., Schmitz F., Banasiewicz T., Drews M., Folsch U. R., Herzig K. H., Expression of keratinocyte growth factor and its receptor in colorectal cancer. Eur J Clin Invest, 2000. 30 (3): p. 222-229
  • Jayson G. C., Vives C., Paraskeva C., Schofield K., Coutts J., Fleetwood A., et al., Coordinated modulation of the fibroblast growth factor dual receptor mechanism during transformation from human colon adenoma to carcinoma. Int J Cancer, 1999. 82 (2): p. 298-304
  • Goke F., Goke A., von Massenhausen A., Franzen A., Sharma R., Kirsten R., et al., Fibroblast growth factor receptor 1 as a putative therapy target in colorectal cancer. Digestion, 2013. 88 (3): p. 172-181
  • Matsuda Y., Ishiwata T., Yamahatsu K., Kawahara K., Hagio M., Peng W. X., et al., Overexpressed fibroblast growth factor receptor 2 in the invasive front of colorectal cancer: a potential therapeutic target in colorectal cancer. Cancer Lett, 2011. 309 (2): p. 209-219
  • Visco V., Belleudi F., Marchese C., Leone L., Aimati L., Cardinali G., et al., Differential response to keratinocyte growth factor receptor and epidermal growth factor receptor ligands of proliferating and differentiating intestinal epithelial cells. J Cell Physiol, 2004. 200 (1): p. 31-44
  • Watanabe M., Ishiwata T., Nishigai K., Moriyama Y., Asano G., Overexpression of keratinocyte growth factor in cancer cells and enterochromaffin cells in human colorectal cancer. Pathol Int, 2000. 50 (5): p. 363-372
  • Narita K., Fujii T., Ishiwata T., Yamamoto T., Kawamoto Y., Kawahara K., et al., Keratinocyte growth factor induces vascular endothelial growth factor-A expression in colorectal cancer cells. Int J Oncol, 2009. 34 (2): p. 355-360
  • Kudo M., Ishiwata T., Nakazawa N., Kawahara K., Fujii T., Teduka K., et al., Keratinocyte growth factor-transfection-stimulated adhesion of colorectal cancer cells to extracellular matrices. Exp Mol Pathol, 2007. 83 (3): p. 443-452
  • Matsuike A., Ishiwata T., Watanabe M., Asano G., Expression of fibroblast growth factor (FGF) -10 in human colorectal adenocarcinoma cells. J Nippon Med Sch, 2001. 68 (5): p. 397-404
  • Sato T., Oshima T., Yoshihara K., Yamamoto N., Yamada R., Nagano Y., et al., Overexpression of the fibroblast growth factor receptor-1 gene correlates with liver metastasis in colorectal cancer. Oncol Rep, 2009. 21 (1): p. 211-216
  • Jang J. H., Shin K. H., Park Y. J., Lee R. J., McKeehan W.L., Park J. G., Novel transcripts of fibroblast growth factor receptor 3 reveal aberrant splicing and activation of cryptic splice sequences in colorectal cancer. Cancer Res, 2000. 60 (15): p. 4049-4052.
  • Desnoyers L. R., Pai R., Ferrando R. E., Hotzel K., Le T., Ross J., et al., Targeting FGF19 inhibits tumor growth in colon cancer xenograft and FGF19 transgenic hepatocellular carcinoma models. Oncogene, 2008. 27 (1): p. 85-97
  • Henriksson M. L., Edin S., Dahlin A. M., Oldenborg P. A., Oberg A., Van Guelpen B., et al., Colorectal cancer cells activate adjacent fibroblasts resulting in FGF1/FGFR3 signaling and increased invasion. Am J Pathol, 2011. 178 (3): p. 1387-1394
  • Liu R., Li J., Xie K., Zhang T., Lei Y., Chen Y., et al., FGFR4 promotes stroma-induced epithelial-to-mesenchymal transition in colorectal cancer. Cancer Res, 2013. 73 (19): p. 5926-5935
  • Bange J., Prechtl D., Cheburkin Y., Specht K., Harbeck N., Schmitt M., et al., Cancer progression and tumor cell motility are associated with the FGFR4 Arg (388) allele. Cancer Res, 2002. 62 (3): p. 840-847
  • Turkington R. C., Longley D. B., Allen W. L., Stevenson L., McLaughlin K., Dunne P. D., et al., Fibroblast growth factor receptor 4 (FGFR4): a targetable regulator of drug resistance in colorectal cancer. Cell Death Dis, 2014. 5: p. e1046
  • Knights, V. and S. J. Cook, De-regulated FGF receptors as therapeutic targets in cancer. Pharmacol Ther, 2010. 125 (1): p. 105-17
  • Taylor, S.K., et al., A phase II study of pazopanib in patients with recurrent or metastatic invasive breast carcinoma: a trial of the Princess Margaret Hospital phase II consortium. Oncologist, 2010. 15 (8): p. 810-8
  • Cristofanilli, M., et al., A randomized phase II study of lapatinib + pazopanib versus lapatinib in patients with HER2+ inflammatory breast cancer. Breast Cancer Res Treat, 2013. 137 (2): p. 471-82
  • Brady, J., et al., An open-label study of the safety and tolerability of pazopanib in combination with FOLFOX6 or CapeOx in patients with colorectal cancer. Invest New Drugs, 2013. 31 (5): p. 1228-35
  • Shiang, C.Y., et al., Amplification of fibroblast growth factor receptor-1 in breast cancer and the effects of brivanib alaninate. Breast Cancer Res Treat, 2010. 123 (3): p. 747-55
  • Siu, L., et al., NCIC Clinical Trials Group and AGITG: Phase III randomized trial of cetuximab (CET) plus either brivanib alaninate (BRIV) or placebo in patients (pts) with metastatic (MET) chemotherapy refractory K-RAS wild-type (WT) colorectal carcinoma (CRC): The NCIC Clinical Trials Group and AGITG CO. 20 trial. J Clin Oncol, 2012. 30 (suppl 4): p. 3504
  • Dempke, W.C. and R. Zippel, Brivanib, a novel dual VEGF-R2/bFGF-R inhibitor. Anticancer Res, 2010. 30 (11): p. 4477-83
  • Du Bois, A., et al. AGO-OVAR 12: A randomized placebo-controlled GCIG/ENGOT-INTERGROUP phase III trial of standard frontline chemotherapy plus/-nintedanib for advanced ovarian cancer. Internatioanal Journal of Gynecological Cancer. 2013
  • Raja, F., et al., Randomized double-blind phase III trial of cediranib (AZD 2171) in relapsed platinum sensetive ovarisn cancer: results of the ICON6 trial. International Journal of Cynecological Cancer, 2013. 23 (8)
  • Konecny G, F.N., Garcia A, et al., Phase 2 study of second-line dovitinib (TKI258) in patients with fibroblast growth factor receptor 2 (FGFR2) -mutated or nonmutated advanced and/or metastatic endometrial cancer.ESMO2014. LBA27
  • Zhang, H., et al., FP-1039 (FGFR1: Fc), a soluble FGFR1 receptor antagonist, inhibits tumor growth and angiogenesis. Molecular Cancer Therapeutics, 2007. 6 (11 Supplement): p. B55-B55
Еще
Статья научная