Skip to main content
Top
Published in: Molecular Cancer 1/2018

Open Access 01-12-2018 | Review

Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities

Authors: Yazhuo Zhang, Mengfang Xia, Ke Jin, Shufei Wang, Hang Wei, Chunmei Fan, Yingfen Wu, Xiaoling Li, Xiayu Li, Guiyuan Li, Zhaoyang Zeng, Wei Xiong

Published in: Molecular Cancer | Issue 1/2018

Login to get access

Abstract

c-Met is a receptor tyrosine kinase belonging to the MET (MNNG HOS transforming gene) family, and is expressed on the surfaces of various cells. Hepatocyte growth factor (HGF) is the ligand for this receptor. The binding of HGF to c-Met initiates a series of intracellular signals that mediate embryogenesis and wound healing in normal cells. However, in cancer cells, aberrant HGF/c-Met axis activation, which is closely related to c-Met gene mutations, overexpression, and amplification, promotes tumor development and progression by stimulating the PI3K/AKT, Ras/MAPK, JAK/STAT, SRC, Wnt/β-catenin, and other signaling pathways. Thus, c-Met and its associated signaling pathways are clinically important therapeutic targets. In this review, we elaborate on the molecular structure of c-Met and HGF and the mechanism through which their interaction activates the PI3K/AKT, Ras/MAPK, and Wnt signaling pathways. We also summarize the connection between c-Met and RON and EGFR, which are also receptor tyrosine kinases. Finally, we introduce the current therapeutic drugs that target c-Met in primary tumors, and their use in clinical research.
Literature
1.
go back to reference Salgia R. MET in lung cancer: biomarker selection based on scientific rationale. Mol Cancer Ther. 2017;16:555–65.PubMedCrossRef Salgia R. MET in lung cancer: biomarker selection based on scientific rationale. Mol Cancer Ther. 2017;16:555–65.PubMedCrossRef
2.
go back to reference Fu YT, Zheng HB, Zhou L, Zhang DQ, Liu XL, Sun H. Valproic acid, targets papillary thyroid cancer through inhibition of c-Met signalling pathway. Am J Transl Res. 2017;9:3138–47.PubMedPubMedCentral Fu YT, Zheng HB, Zhou L, Zhang DQ, Liu XL, Sun H. Valproic acid, targets papillary thyroid cancer through inhibition of c-Met signalling pathway. Am J Transl Res. 2017;9:3138–47.PubMedPubMedCentral
3.
go back to reference Rucki AA, Xiao Q, Muth S, Chen J, Che X, Kleponis J, Sharma R, Anders RA, Jaffee EM, Zheng L. Dual Inhibition of Hedgehog and c-Met Pathways for Pancreatic Cancer Treatment. Mol Cancer Ther. 2017;16:2399-409. Rucki AA, Xiao Q, Muth S, Chen J, Che X, Kleponis J, Sharma R, Anders RA, Jaffee EM, Zheng L. Dual Inhibition of Hedgehog and c-Met Pathways for Pancreatic Cancer Treatment. Mol Cancer Ther. 2017;16:2399-409.
4.
go back to reference Zhu L, Xiong X, Kim Y, Okada N, Lu F, Zhang H, Sun H. Acid sphingomyelinase is required for cell surface presentation of met receptor tyrosine kinase in cancer cells. J Cell Sci. 2016;129:4238–51.PubMedPubMedCentralCrossRef Zhu L, Xiong X, Kim Y, Okada N, Lu F, Zhang H, Sun H. Acid sphingomyelinase is required for cell surface presentation of met receptor tyrosine kinase in cancer cells. J Cell Sci. 2016;129:4238–51.PubMedPubMedCentralCrossRef
5.
go back to reference Liu WT, Jing YY, Yu GF, Chen H, Han ZP, Yu DD, Fan QM, Ye F, Li R, Gao L, et al. Hepatic stellate cell promoted hepatoma cell invasion via the HGF/c-Met signaling pathway regulated by p53. Cell Cycle. 2016;15:886–94.PubMedPubMedCentralCrossRef Liu WT, Jing YY, Yu GF, Chen H, Han ZP, Yu DD, Fan QM, Ye F, Li R, Gao L, et al. Hepatic stellate cell promoted hepatoma cell invasion via the HGF/c-Met signaling pathway regulated by p53. Cell Cycle. 2016;15:886–94.PubMedPubMedCentralCrossRef
7.
go back to reference Szturz P, Raymond E, Abitbol C, Albert S, de Gramont A, Faivre S. Understanding c-MET signalling in squamous cell carcinoma of the head & neck. Crit Rev Oncol Hematol. 2017;111:39–51.PubMedCrossRef Szturz P, Raymond E, Abitbol C, Albert S, de Gramont A, Faivre S. Understanding c-MET signalling in squamous cell carcinoma of the head & neck. Crit Rev Oncol Hematol. 2017;111:39–51.PubMedCrossRef
8.
go back to reference Kim B, Jung N, Lee S, Sohng JK, Jung HJ. Apigenin inhibits cancer stem cell-like phenotypes in human Glioblastoma cells via suppression of c-Met Signaling. Phytother Res. 2016;30:1833–40.PubMedCrossRef Kim B, Jung N, Lee S, Sohng JK, Jung HJ. Apigenin inhibits cancer stem cell-like phenotypes in human Glioblastoma cells via suppression of c-Met Signaling. Phytother Res. 2016;30:1833–40.PubMedCrossRef
9.
go back to reference International Cancer Genome Consortium PedBrain Tumor P. Recurrent MET fusion genes represent a drug target in pediatric glioblastoma. Nat Med. 2016;22:1314–20.CrossRef International Cancer Genome Consortium PedBrain Tumor P. Recurrent MET fusion genes represent a drug target in pediatric glioblastoma. Nat Med. 2016;22:1314–20.CrossRef
10.
go back to reference Imura Y, Nakai T, Yamada S, Outani H, Takenaka S, Hamada K, Araki N, Itoh K, Yoshikawa H, Naka N. Functional and therapeutic relevance of hepatocyte growth factor/c-MET signaling in synovial sarcoma. Cancer Sci. 2016;107:1867–76.PubMedPubMedCentralCrossRef Imura Y, Nakai T, Yamada S, Outani H, Takenaka S, Hamada K, Araki N, Itoh K, Yoshikawa H, Naka N. Functional and therapeutic relevance of hepatocyte growth factor/c-MET signaling in synovial sarcoma. Cancer Sci. 2016;107:1867–76.PubMedPubMedCentralCrossRef
11.
go back to reference Liang Y, Liu J, Liu T, Yang X. Anti-c-Met antibody bioconjugated with hollow gold nanospheres as a novel nanomaterial for targeted radiation ablation of human cervical cancer cell. Oncol Lett. 2017;14:2254–60.PubMedPubMedCentralCrossRef Liang Y, Liu J, Liu T, Yang X. Anti-c-Met antibody bioconjugated with hollow gold nanospheres as a novel nanomaterial for targeted radiation ablation of human cervical cancer cell. Oncol Lett. 2017;14:2254–60.PubMedPubMedCentralCrossRef
12.
go back to reference Pilotto S, Carbognin L, Karachaliou N, Ma PC, Rosell R, Tortora G, Bria E. Tracking MET de-addiction in lung cancer: a road towards the oncogenic target. Cancer Treat Rev. 2017;60:1–11.PubMedCrossRef Pilotto S, Carbognin L, Karachaliou N, Ma PC, Rosell R, Tortora G, Bria E. Tracking MET de-addiction in lung cancer: a road towards the oncogenic target. Cancer Treat Rev. 2017;60:1–11.PubMedCrossRef
13.
go back to reference Wu JC, Wang CT, Hung HC, Wu WJ, Wu DC, Chang MC, Sung PJ, Chou YW, Wen ZH, Tai MH. Heteronemin is a novel c-Met /STAT3 inhibitor against advanced prostate cancer cells. Prostate. 2016;76:1469–83.PubMedCrossRef Wu JC, Wang CT, Hung HC, Wu WJ, Wu DC, Chang MC, Sung PJ, Chou YW, Wen ZH, Tai MH. Heteronemin is a novel c-Met /STAT3 inhibitor against advanced prostate cancer cells. Prostate. 2016;76:1469–83.PubMedCrossRef
14.
go back to reference Safaie Qamsari E, Safaei Ghaderi S, Zarei B, Dorostkar R, Bagheri S, Jadidi-Niaragh F, Somi MH, Yousefi M. The c-Met receptor: implication for targeted therapies in colorectal cancer. Tumour Biol. 2017;39:1010428317699118.PubMedCrossRef Safaie Qamsari E, Safaei Ghaderi S, Zarei B, Dorostkar R, Bagheri S, Jadidi-Niaragh F, Somi MH, Yousefi M. The c-Met receptor: implication for targeted therapies in colorectal cancer. Tumour Biol. 2017;39:1010428317699118.PubMedCrossRef
15.
go back to reference Hughes PE, Rex K, Caenepeel S, Yang Y, Zhang Y, Broome MA, Kha HT, Burgess TL, Amore B, Kaplan-Lefko PJ, et al. In vitro and in vivo activity of AMG 337, a potent and selective MET Kinase inhibitor, in MET-dependent cancer models. Mol Cancer Ther. 2016;15:1568–79.PubMedCrossRef Hughes PE, Rex K, Caenepeel S, Yang Y, Zhang Y, Broome MA, Kha HT, Burgess TL, Amore B, Kaplan-Lefko PJ, et al. In vitro and in vivo activity of AMG 337, a potent and selective MET Kinase inhibitor, in MET-dependent cancer models. Mol Cancer Ther. 2016;15:1568–79.PubMedCrossRef
16.
go back to reference Kuang W, Deng Q, Deng C, Li W, Shu S, Zhou M. Hepatocyte growth factor induces breast cancer cell invasion via the PI3K/Akt and p38 MAPK signaling pathways to up-regulate the expression of COX2. Am J Transl Res. 2017;9:3816–26.PubMedPubMedCentral Kuang W, Deng Q, Deng C, Li W, Shu S, Zhou M. Hepatocyte growth factor induces breast cancer cell invasion via the PI3K/Akt and p38 MAPK signaling pathways to up-regulate the expression of COX2. Am J Transl Res. 2017;9:3816–26.PubMedPubMedCentral
17.
go back to reference Leung E, Xue A, Wang Y, Rougerie P, Sharma VP, Eddy R, Cox D, Condeelis J. Blood vessel endothelium-directed tumor cell streaming in breast tumors requires the HGF/C-Met signaling pathway. Oncogene. 2017;36:2680–92.PubMedCrossRef Leung E, Xue A, Wang Y, Rougerie P, Sharma VP, Eddy R, Cox D, Condeelis J. Blood vessel endothelium-directed tumor cell streaming in breast tumors requires the HGF/C-Met signaling pathway. Oncogene. 2017;36:2680–92.PubMedCrossRef
18.
go back to reference Bahrami A, Shahidsales S, Khazaei M, Ghayour-Mobarhan M, Maftouh M, Hassanian SM, Avan A. c-Met as a potential target for the treatment of gastrointestinal cancer: current status and future perspectives. J Cell Physiol. 2017;232:2657–73.PubMedCrossRef Bahrami A, Shahidsales S, Khazaei M, Ghayour-Mobarhan M, Maftouh M, Hassanian SM, Avan A. c-Met as a potential target for the treatment of gastrointestinal cancer: current status and future perspectives. J Cell Physiol. 2017;232:2657–73.PubMedCrossRef
19.
go back to reference Barrow-McGee R, Kishi N, Joffre C, Menard L, Hervieu A, Bakhouche BA, Noval AJ, Mai A, Guzman C, Robert-Masson L, et al. Beta 1-integrin-c-Met cooperation reveals an inside-in survival signalling on autophagy-related endomembranes. Nat Commun. 2016;7:11942.PubMedPubMedCentralCrossRef Barrow-McGee R, Kishi N, Joffre C, Menard L, Hervieu A, Bakhouche BA, Noval AJ, Mai A, Guzman C, Robert-Masson L, et al. Beta 1-integrin-c-Met cooperation reveals an inside-in survival signalling on autophagy-related endomembranes. Nat Commun. 2016;7:11942.PubMedPubMedCentralCrossRef
20.
go back to reference Caenepeel S, Cooke K, Wadsworth S, Huang G, Robert L, Moreno BH, Parisi G, Cajulis E, Kendall R, Beltran P, et al. MAPK pathway inhibition induces MET and GAB1 levels, priming BRAF mutant melanoma for rescue by hepatocyte growth factor. Oncotarget. 2017;8:17795–809.PubMedPubMedCentralCrossRef Caenepeel S, Cooke K, Wadsworth S, Huang G, Robert L, Moreno BH, Parisi G, Cajulis E, Kendall R, Beltran P, et al. MAPK pathway inhibition induces MET and GAB1 levels, priming BRAF mutant melanoma for rescue by hepatocyte growth factor. Oncotarget. 2017;8:17795–809.PubMedPubMedCentralCrossRef
21.
go back to reference Li Y, Liu H, Chen J. Dysregulation of HGF/ c-Met signal pathway and their targeting drugs in lung cancer. Zhongguo Fei Ai Za Zhi. 2014;17:625–34.PubMed Li Y, Liu H, Chen J. Dysregulation of HGF/ c-Met signal pathway and their targeting drugs in lung cancer. Zhongguo Fei Ai Za Zhi. 2014;17:625–34.PubMed
22.
go back to reference Yin B, Liu Z, Wang Y, Wang X, Liu W, Yu P, Duan X, Liu C, Chen Y, Zhang Y, et al. RON and c-Met facilitate metastasis through the ERK signaling pathway in prostate cancer cells. Oncol Rep. 2017;37:3209–18.PubMedPubMedCentralCrossRef Yin B, Liu Z, Wang Y, Wang X, Liu W, Yu P, Duan X, Liu C, Chen Y, Zhang Y, et al. RON and c-Met facilitate metastasis through the ERK signaling pathway in prostate cancer cells. Oncol Rep. 2017;37:3209–18.PubMedPubMedCentralCrossRef
23.
go back to reference Hass R, Jennek S, Yang Y, Friedrich K. c-Met expression and activity in urogenital cancers - novel aspects of signal transduction and medical implications. Cell Commun Signal. 2017;15:10.PubMedPubMedCentralCrossRef Hass R, Jennek S, Yang Y, Friedrich K. c-Met expression and activity in urogenital cancers - novel aspects of signal transduction and medical implications. Cell Commun Signal. 2017;15:10.PubMedPubMedCentralCrossRef
24.
go back to reference Al-U'datt DGF, Al-Husein BAA, Qasaimeh GR. A mini-review of c-Met as a potential therapeutic target in melanoma. Biomed Pharmacother. 2017;88:194–202.PubMedCrossRef Al-U'datt DGF, Al-Husein BAA, Qasaimeh GR. A mini-review of c-Met as a potential therapeutic target in melanoma. Biomed Pharmacother. 2017;88:194–202.PubMedCrossRef
27.
go back to reference Rosário M. How to make tubes: signaling by the met receptor tyrosine kinase. Trends Cell Biol. 2003;13:328–35.PubMedCrossRef Rosário M. How to make tubes: signaling by the met receptor tyrosine kinase. Trends Cell Biol. 2003;13:328–35.PubMedCrossRef
28.
go back to reference Vogel W, Ullrich A. Multiple in vivo phosphorylated tyrosine phosphatase SHP-2 engages binding to Grb2 via tyrosine 584. Cell Growth Differ. 1996;7:1589–97.PubMed Vogel W, Ullrich A. Multiple in vivo phosphorylated tyrosine phosphatase SHP-2 engages binding to Grb2 via tyrosine 584. Cell Growth Differ. 1996;7:1589–97.PubMed
29.
go back to reference Wang M, Zhao J, Zhang L, Wei F, Lian Y, Wu Y, Gong Z, Zhang S, Zhou J, Cao K, et al. Role of tumor microenvironment in tumorigenesis. J Cancer. 2017;8:761–73.PubMedPubMedCentralCrossRef Wang M, Zhao J, Zhang L, Wei F, Lian Y, Wu Y, Gong Z, Zhang S, Zhou J, Cao K, et al. Role of tumor microenvironment in tumorigenesis. J Cancer. 2017;8:761–73.PubMedPubMedCentralCrossRef
30.
go back to reference Liebmann C. Regulation of MAP kinase activity by peptide receptor signalling pathway: paradigms of multiplicity. Cell Signal. 2001;13:777–85.PubMedCrossRef Liebmann C. Regulation of MAP kinase activity by peptide receptor signalling pathway: paradigms of multiplicity. Cell Signal. 2001;13:777–85.PubMedCrossRef
31.
go back to reference Shaw RJ, Cantley LC. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature. 2006;441:424–30.PubMedCrossRef Shaw RJ, Cantley LC. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature. 2006;441:424–30.PubMedCrossRef
32.
go back to reference Mottet D, Dumont V, Deccache Y, Demazy C, Ninane N, Raes M, Michiels C. Regulation of hypoxia-inducible factor-1alpha protein level during hypoxic conditions by the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3beta pathway in HepG2 cells. J Biol Chem. 2003;278:31277–85.PubMedCrossRef Mottet D, Dumont V, Deccache Y, Demazy C, Ninane N, Raes M, Michiels C. Regulation of hypoxia-inducible factor-1alpha protein level during hypoxic conditions by the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3beta pathway in HepG2 cells. J Biol Chem. 2003;278:31277–85.PubMedCrossRef
33.
go back to reference Jiang BH, Liu LZ. AKT signaling in regulating angiogenesis. Curr Cancer Drug Targets. 2008;8:19–26.PubMedCrossRef Jiang BH, Liu LZ. AKT signaling in regulating angiogenesis. Curr Cancer Drug Targets. 2008;8:19–26.PubMedCrossRef
34.
go back to reference Holland JD, Gyorffy B, Vogel R, Eckert K, Valenti G, Fang L, Lohneis P, Elezkurtaj S, Ziebold U, Birchmeier W. Combined Wnt/beta-catenin, met, and CXCL12/CXCR4 signals characterize basal breast cancer and predict disease outcome. Cell Rep. 2013;5:1214–27.PubMedCrossRef Holland JD, Gyorffy B, Vogel R, Eckert K, Valenti G, Fang L, Lohneis P, Elezkurtaj S, Ziebold U, Birchmeier W. Combined Wnt/beta-catenin, met, and CXCL12/CXCR4 signals characterize basal breast cancer and predict disease outcome. Cell Rep. 2013;5:1214–27.PubMedCrossRef
35.
go back to reference Tuynman JB, Vermeulen L, Boon EM, Kemper K, Zwinderman AH, Peppelenbosch MP, Richel DJ. Cyclooxygenase-2 inhibition inhibits c-Met kinase activity and Wnt activity in colon cancer. Cancer Res. 2008;68:1213–20.PubMedCrossRef Tuynman JB, Vermeulen L, Boon EM, Kemper K, Zwinderman AH, Peppelenbosch MP, Richel DJ. Cyclooxygenase-2 inhibition inhibits c-Met kinase activity and Wnt activity in colon cancer. Cancer Res. 2008;68:1213–20.PubMedCrossRef
36.
go back to reference Kim KH, Seol HJ, Kim EH, Rheey J, Jin HJ, Lee Y, Joo KM, Lee J, Nam DH. Wnt/beta-catenin signaling is a key downstream mediator of MET signaling in glioblastoma stem cells. Neuro-Oncology. 2013;15:161–71.PubMedCrossRef Kim KH, Seol HJ, Kim EH, Rheey J, Jin HJ, Lee Y, Joo KM, Lee J, Nam DH. Wnt/beta-catenin signaling is a key downstream mediator of MET signaling in glioblastoma stem cells. Neuro-Oncology. 2013;15:161–71.PubMedCrossRef
37.
go back to reference Previdi S, Maroni P, Matteucci E, Broggini M, Bendinelli P, Desiderio MA. Interaction between human-breast cancer metastasis and bone microenvironment through activated hepatocyte growth factor/met and beta-catenin/Wnt pathways. Eur J Cancer. 2010;46:1679–91.PubMedCrossRef Previdi S, Maroni P, Matteucci E, Broggini M, Bendinelli P, Desiderio MA. Interaction between human-breast cancer metastasis and bone microenvironment through activated hepatocyte growth factor/met and beta-catenin/Wnt pathways. Eur J Cancer. 2010;46:1679–91.PubMedCrossRef
38.
go back to reference Tortelote GG, Reis RR, de Almeida MF, Abreu JG. Complexity of the Wnt/betacatenin pathway: searching for an activation model. Cell Signal. 2017;40:30–43.PubMedCrossRef Tortelote GG, Reis RR, de Almeida MF, Abreu JG. Complexity of the Wnt/betacatenin pathway: searching for an activation model. Cell Signal. 2017;40:30–43.PubMedCrossRef
39.
go back to reference Wu C, Zhuang Y, Jiang S, Liu S, Zhou J, Wu J, Teng Y, Xia B, Wang R, Zou X. Interaction between Wnt/beta-catenin pathway and microRNAs regulates epithelial-mesenchymal transition in gastric cancer (review). Int J Oncol. 2016;48:2236–46.PubMedCrossRef Wu C, Zhuang Y, Jiang S, Liu S, Zhou J, Wu J, Teng Y, Xia B, Wang R, Zou X. Interaction between Wnt/beta-catenin pathway and microRNAs regulates epithelial-mesenchymal transition in gastric cancer (review). Int J Oncol. 2016;48:2236–46.PubMedCrossRef
40.
go back to reference Yan Q, Zeng Z, Gong Z, Zhang W, Li X, He B, Song Y, Li Q, Zeng Y, Liao Q, et al. EBV-miR-BART10-3p facilitates epithelial-mesenchymal transition and promotes metastasis of nasopharyngeal carcinoma by targeting BTRC. Oncotarget. 2015;6:41766–82.PubMedPubMedCentral Yan Q, Zeng Z, Gong Z, Zhang W, Li X, He B, Song Y, Li Q, Zeng Y, Liao Q, et al. EBV-miR-BART10-3p facilitates epithelial-mesenchymal transition and promotes metastasis of nasopharyngeal carcinoma by targeting BTRC. Oncotarget. 2015;6:41766–82.PubMedPubMedCentral
41.
go back to reference Arend RC, Londono-Joshi AI, Straughn JM Jr, Buchsbaum DJ. The Wnt/beta-catenin pathway in ovarian cancer: a review. Gynecol Oncol. 2013;131:772–9.PubMedCrossRef Arend RC, Londono-Joshi AI, Straughn JM Jr, Buchsbaum DJ. The Wnt/beta-catenin pathway in ovarian cancer: a review. Gynecol Oncol. 2013;131:772–9.PubMedCrossRef
42.
go back to reference Mariani M, McHugh M, Petrillo M, Sieber S, He S, Andreoli M, Wu Z, Fiedler P, Scambia G, Shahabi S, Ferlini C. HGF/c-Met axis drives cancer aggressiveness in the neo-adjuvant setting of ovarian cancer. Oncotarget. 2014;5:4855–67.PubMedPubMedCentralCrossRef Mariani M, McHugh M, Petrillo M, Sieber S, He S, Andreoli M, Wu Z, Fiedler P, Scambia G, Shahabi S, Ferlini C. HGF/c-Met axis drives cancer aggressiveness in the neo-adjuvant setting of ovarian cancer. Oncotarget. 2014;5:4855–67.PubMedPubMedCentralCrossRef
43.
go back to reference Raman JD. Re: prognostic value of MET, RON and histoprognostic factors for urothelial carcinoma in the upper urinary tract. E. Comperat, M. Roupret, E. Chartier-Kastler, M. O. Bitker, F. Richard, P. Camparo, F. Capron and O. Cussenot. J Urol 2008; 179: 868-872. J Urol. 2008;180:1183. author reply 1183-1184PubMedCrossRef Raman JD. Re: prognostic value of MET, RON and histoprognostic factors for urothelial carcinoma in the upper urinary tract. E. Comperat, M. Roupret, E. Chartier-Kastler, M. O. Bitker, F. Richard, P. Camparo, F. Capron and O. Cussenot. J Urol 2008; 179: 868-872. J Urol. 2008;180:1183. author reply 1183-1184PubMedCrossRef
44.
go back to reference Cheng HL, Liu HS, Lin YJ, Chen HH, Hsu PY, Chang TY, Ho CL, Tzai TS, Chow NH. Co-expression of RON and MET is a prognostic indicator for patients with transitional-cell carcinoma of the bladder. Br J Cancer. 2005;92:1906–14.PubMedPubMedCentralCrossRef Cheng HL, Liu HS, Lin YJ, Chen HH, Hsu PY, Chang TY, Ho CL, Tzai TS, Chow NH. Co-expression of RON and MET is a prognostic indicator for patients with transitional-cell carcinoma of the bladder. Br J Cancer. 2005;92:1906–14.PubMedPubMedCentralCrossRef
45.
go back to reference Maggiora P. The RON and MET oncogenes are co-expressed in human ovarian carcinomas and cooperate in activating invasiveness. Exp Cell Res. 2003;288:382–9.PubMedCrossRef Maggiora P. The RON and MET oncogenes are co-expressed in human ovarian carcinomas and cooperate in activating invasiveness. Exp Cell Res. 2003;288:382–9.PubMedCrossRef
46.
go back to reference McClaine RJ, Marshal AM, Wagh PK, Waltz SE. Ron receptor tyrosine Kinase activation confers resistance to Tamoxifen in breast cancer cell lines. Neoplasia. 2010;12:650–8.PubMedPubMedCentralCrossRef McClaine RJ, Marshal AM, Wagh PK, Waltz SE. Ron receptor tyrosine Kinase activation confers resistance to Tamoxifen in breast cancer cell lines. Neoplasia. 2010;12:650–8.PubMedPubMedCentralCrossRef
47.
go back to reference Previdi S, Abbadessa G, Dalo F, France DS, Broggini M. Breast cancer-derived bone metastasis can be effectively reduced through specific c-MET inhibitor tivantinib (ARQ 197) and shRNA c-MET knockdown. Mol Cancer Ther. 2012;11:214–23.PubMedCrossRef Previdi S, Abbadessa G, Dalo F, France DS, Broggini M. Breast cancer-derived bone metastasis can be effectively reduced through specific c-MET inhibitor tivantinib (ARQ 197) and shRNA c-MET knockdown. Mol Cancer Ther. 2012;11:214–23.PubMedCrossRef
48.
go back to reference Takeuchi H, Bilchik A, Saha S, Turner R, Wiese D, Tanaka M, Kuo C, Wang HJ, Hoon DS. c-MET expression level in primary colon cancer: a predictor of tumor invasion and lymph node metastases. Clin Cancer Res. 2003;9:1480–8.PubMed Takeuchi H, Bilchik A, Saha S, Turner R, Wiese D, Tanaka M, Kuo C, Wang HJ, Hoon DS. c-MET expression level in primary colon cancer: a predictor of tumor invasion and lymph node metastases. Clin Cancer Res. 2003;9:1480–8.PubMed
49.
go back to reference Zhao S, Ammanamanchi S, Brattain M, Cao L, Thangasamy A, Wang J, Freeman JW. Smad4-dependent TGF-beta signaling suppresses RON receptor tyrosine kinase-dependent motility and invasion of pancreatic cancer cells. J Biol Chem. 2008;283:11293–301.PubMedPubMedCentralCrossRef Zhao S, Ammanamanchi S, Brattain M, Cao L, Thangasamy A, Wang J, Freeman JW. Smad4-dependent TGF-beta signaling suppresses RON receptor tyrosine kinase-dependent motility and invasion of pancreatic cancer cells. J Biol Chem. 2008;283:11293–301.PubMedPubMedCentralCrossRef
50.
go back to reference Benvenuti S, Lazzari L, Arnesano A, Li Chiavi G, Gentile A, Comoglio PM. Ron kinase transphosphorylation sustains MET oncogene addiction. Cancer Res. 2011;71:1945–55.PubMedCrossRef Benvenuti S, Lazzari L, Arnesano A, Li Chiavi G, Gentile A, Comoglio PM. Ron kinase transphosphorylation sustains MET oncogene addiction. Cancer Res. 2011;71:1945–55.PubMedCrossRef
51.
go back to reference Gaudino G, Follenzi A, Naldini L, Collesi C, Santoro M, Gallo KA, Godowski PJ, Comoglio PM. RON is a heterodimeric tyrosine kinase receptor activated by the HGF homologue MSP. EMBO J. 1994;13:3524–32.PubMedPubMedCentral Gaudino G, Follenzi A, Naldini L, Collesi C, Santoro M, Gallo KA, Godowski PJ, Comoglio PM. RON is a heterodimeric tyrosine kinase receptor activated by the HGF homologue MSP. EMBO J. 1994;13:3524–32.PubMedPubMedCentral
52.
go back to reference Follenzi A, Bakovic S, Gual P, Stella MC, Longati P, Comoglio PM. Cross-talk between the proto-oncogenes met and Ron. Oncogene. 2000;19:3041–9.PubMedCrossRef Follenzi A, Bakovic S, Gual P, Stella MC, Longati P, Comoglio PM. Cross-talk between the proto-oncogenes met and Ron. Oncogene. 2000;19:3041–9.PubMedCrossRef
53.
54.
go back to reference Peace BE, Hughes MJ, Degen SJ, Waltz SE. Point mutations and overexpression of Ron induce transformation, tumor formation, and metastasis. Oncogene. 2001;20:6142–51.PubMedCrossRef Peace BE, Hughes MJ, Degen SJ, Waltz SE. Point mutations and overexpression of Ron induce transformation, tumor formation, and metastasis. Oncogene. 2001;20:6142–51.PubMedCrossRef
55.
go back to reference Pinkas-Kramarski R, Shelly M, Glathe S, Ratzkin BJ, Yarden Y. Neu differentiation factor/neuregulin isoforms activate distinct receptor combinations. J Biol Chem. 1996;271:19029–32.PubMedCrossRef Pinkas-Kramarski R, Shelly M, Glathe S, Ratzkin BJ, Yarden Y. Neu differentiation factor/neuregulin isoforms activate distinct receptor combinations. J Biol Chem. 1996;271:19029–32.PubMedCrossRef
56.
go back to reference Zhao S, Cao L, Freeman JW. Knockdown of RON receptor kinase delays but does not prevent tumor progression while enhancing HGF/MET signaling in pancreatic cancer cell lines. Oncogene. 2013;2:e76.CrossRef Zhao S, Cao L, Freeman JW. Knockdown of RON receptor kinase delays but does not prevent tumor progression while enhancing HGF/MET signaling in pancreatic cancer cell lines. Oncogene. 2013;2:e76.CrossRef
57.
go back to reference Wang MH, Wang D, Chen YQ. Oncogenic and invasive potentials of human macrophage-stimulating protein receptor, the RON receptor tyrosine kinase. Carcinogenesis. 2003;24:1291–300.PubMedCrossRef Wang MH, Wang D, Chen YQ. Oncogenic and invasive potentials of human macrophage-stimulating protein receptor, the RON receptor tyrosine kinase. Carcinogenesis. 2003;24:1291–300.PubMedCrossRef
58.
go back to reference Chaudhuri A, Xie MH, Yang B, Mahapatra K, Liu J, Marsters S, Bodepudi S, Ashkenazi A. Distinct involvement of the Gab1 and Grb2 adaptor proteins in signal transduction by the related receptor tyrosine kinases RON and MET. J Biol Chem. 2011;286:32762–74.PubMedPubMedCentralCrossRef Chaudhuri A, Xie MH, Yang B, Mahapatra K, Liu J, Marsters S, Bodepudi S, Ashkenazi A. Distinct involvement of the Gab1 and Grb2 adaptor proteins in signal transduction by the related receptor tyrosine kinases RON and MET. J Biol Chem. 2011;286:32762–74.PubMedPubMedCentralCrossRef
59.
go back to reference Logan-Collins J, Thomas RM, Yu P, Jaquish D, Mose E, French R, Stuart W, McClaine R, Aronow B, Hoffman RM, et al. Silencing of RON receptor signaling promotes apoptosis and gemcitabine sensitivity in pancreatic cancers. Cancer Res. 2010;70:1130–40.PubMedPubMedCentralCrossRef Logan-Collins J, Thomas RM, Yu P, Jaquish D, Mose E, French R, Stuart W, McClaine R, Aronow B, Hoffman RM, et al. Silencing of RON receptor signaling promotes apoptosis and gemcitabine sensitivity in pancreatic cancers. Cancer Res. 2010;70:1130–40.PubMedPubMedCentralCrossRef
60.
go back to reference Velpula KK, Dasari VR, Asuthkar S, Gorantla B, Tsung AJ. EGFR and c-Met cross talk in Glioblastoma and its regulation by human cord blood stem cells. Transl Oncol. 2012;5:379–IN318.PubMedPubMedCentralCrossRef Velpula KK, Dasari VR, Asuthkar S, Gorantla B, Tsung AJ. EGFR and c-Met cross talk in Glioblastoma and its regulation by human cord blood stem cells. Transl Oncol. 2012;5:379–IN318.PubMedPubMedCentralCrossRef
61.
go back to reference McDermott U, Pusapati RV, Christensen JG, Gray NS, Settleman J. Acquired resistance of non-small cell lung cancer cells to MET kinase inhibition is mediated by a switch to epidermal growth factor receptor dependency. Cancer Res. 2010;70:1625–34.PubMedPubMedCentralCrossRef McDermott U, Pusapati RV, Christensen JG, Gray NS, Settleman J. Acquired resistance of non-small cell lung cancer cells to MET kinase inhibition is mediated by a switch to epidermal growth factor receptor dependency. Cancer Res. 2010;70:1625–34.PubMedPubMedCentralCrossRef
62.
go back to reference Agarwal S, Zerillo C, Kolmakova J, Christensen JG, Harris LN, Rimm DL, Digiovanna MP, Stern DF. Association of constitutively activated hepatocyte growth factor receptor (met) with resistance to a dual EGFR/Her2 inhibitor in non-small-cell lung cancer cells. Br J Cancer. 2009;100:941–9.PubMedPubMedCentralCrossRef Agarwal S, Zerillo C, Kolmakova J, Christensen JG, Harris LN, Rimm DL, Digiovanna MP, Stern DF. Association of constitutively activated hepatocyte growth factor receptor (met) with resistance to a dual EGFR/Her2 inhibitor in non-small-cell lung cancer cells. Br J Cancer. 2009;100:941–9.PubMedPubMedCentralCrossRef
63.
go back to reference Breindel JL, Haskins JW, Cowell EP, Zhao M, Nguyen DX, Stern DF. EGF receptor activates MET through MAPK to enhance non-small cell lung carcinoma invasion and brain metastasis. Cancer Res. 2013;73:5053–65.PubMedPubMedCentralCrossRef Breindel JL, Haskins JW, Cowell EP, Zhao M, Nguyen DX, Stern DF. EGF receptor activates MET through MAPK to enhance non-small cell lung carcinoma invasion and brain metastasis. Cancer Res. 2013;73:5053–65.PubMedPubMedCentralCrossRef
64.
go back to reference Mueller KL, Hunter LA, Ethier SP, Boerner JL. Met and c-Src cooperate to compensate for loss of epidermal growth factor receptor kinase activity in breast cancer cells. Cancer Res. 2008;68:3314–22.PubMedCrossRef Mueller KL, Hunter LA, Ethier SP, Boerner JL. Met and c-Src cooperate to compensate for loss of epidermal growth factor receptor kinase activity in breast cancer cells. Cancer Res. 2008;68:3314–22.PubMedCrossRef
65.
go back to reference Ramalingam SS, Owonikoko TK, Khuri FR. Lung cancer: new biological insights and recent therapeutic advances. CA Cancer J Clin. 2011;61:91–112.PubMedCrossRef Ramalingam SS, Owonikoko TK, Khuri FR. Lung cancer: new biological insights and recent therapeutic advances. CA Cancer J Clin. 2011;61:91–112.PubMedCrossRef
66.
go back to reference Wang W, Li Q, Takeuchi S, Yamada T, Koizumi H, Nakamura T, Matsumoto K, Mukaida N, Nishioka Y, Sone S, et al. Met kinase inhibitor E7050 reverses three different mechanisms of hepatocyte growth factor-induced tyrosine kinase inhibitor resistance in EGFR mutant lung cancer. Clin Cancer Res. 2012;18:1663–71.PubMedCrossRef Wang W, Li Q, Takeuchi S, Yamada T, Koizumi H, Nakamura T, Matsumoto K, Mukaida N, Nishioka Y, Sone S, et al. Met kinase inhibitor E7050 reverses three different mechanisms of hepatocyte growth factor-induced tyrosine kinase inhibitor resistance in EGFR mutant lung cancer. Clin Cancer Res. 2012;18:1663–71.PubMedCrossRef
67.
go back to reference Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–43.PubMedCrossRef Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–43.PubMedCrossRef
68.
go back to reference Benedettini E, Sholl LM, Peyton M, Reilly J, Ware C, Davis L, Vena N, Bailey D, Yeap BY, Fiorentino M, et al. Met activation in non-small cell lung cancer is associated with de novo resistance to EGFR inhibitors and the development of brain metastasis. Am J Pathol. 2010;177:415–23.PubMedPubMedCentralCrossRef Benedettini E, Sholl LM, Peyton M, Reilly J, Ware C, Davis L, Vena N, Bailey D, Yeap BY, Fiorentino M, et al. Met activation in non-small cell lung cancer is associated with de novo resistance to EGFR inhibitors and the development of brain metastasis. Am J Pathol. 2010;177:415–23.PubMedPubMedCentralCrossRef
69.
go back to reference Sequist LV, Martins RG, Spigel D, Grunberg SM, Spira A, Janne PA, Joshi VA, McCollum D, Evans TL, Muzikansky A, et al. First-line gefitinib in patients with advanced non-small-cell lung cancer harboring somatic EGFR mutations. J Clin Oncol. 2008;26:2442–9.PubMedCrossRef Sequist LV, Martins RG, Spigel D, Grunberg SM, Spira A, Janne PA, Joshi VA, McCollum D, Evans TL, Muzikansky A, et al. First-line gefitinib in patients with advanced non-small-cell lung cancer harboring somatic EGFR mutations. J Clin Oncol. 2008;26:2442–9.PubMedCrossRef
70.
go back to reference Turke AB, Zejnullahu K, Wu YL, Song Y, Dias-Santagata D, Lifshits E, Toschi L, Rogers A, Mok T, Sequist L, et al. Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell. 2010;17:77–88.PubMedPubMedCentralCrossRef Turke AB, Zejnullahu K, Wu YL, Song Y, Dias-Santagata D, Lifshits E, Toschi L, Rogers A, Mok T, Sequist L, et al. Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell. 2010;17:77–88.PubMedPubMedCentralCrossRef
71.
go back to reference Raghav KP, Gonzalez-Angulo AM, Blumenschein GR Jr. Role of HGF/MET axis in resistance of lung cancer to contemporary management. Transl Lung Cancer Res. 2012;1:179–93.PubMedPubMedCentral Raghav KP, Gonzalez-Angulo AM, Blumenschein GR Jr. Role of HGF/MET axis in resistance of lung cancer to contemporary management. Transl Lung Cancer Res. 2012;1:179–93.PubMedPubMedCentral
72.
go back to reference Kaposi-Novak P, Lee JS, Gomez-Quiroz L, Coulouarn C, Factor VM, Thorgeirsson SS. Met-regulated expression signature defines a subset of human hepatocellular carcinomas with poor prognosis and aggressive phenotype. J Clin Invest. 2006;116:1582–95.PubMedPubMedCentralCrossRef Kaposi-Novak P, Lee JS, Gomez-Quiroz L, Coulouarn C, Factor VM, Thorgeirsson SS. Met-regulated expression signature defines a subset of human hepatocellular carcinomas with poor prognosis and aggressive phenotype. J Clin Invest. 2006;116:1582–95.PubMedPubMedCentralCrossRef
73.
go back to reference Ke AW, Shi GM, Zhou J, Wu FZ, Ding ZB, Hu MY, Xu Y, Song ZJ, Wang ZJ, Wu JC, et al. Role of overexpression of CD151 and/or c-Met in predicting prognosis of hepatocellular carcinoma. Hepatology. 2009;49:491–503.PubMedCrossRef Ke AW, Shi GM, Zhou J, Wu FZ, Ding ZB, Hu MY, Xu Y, Song ZJ, Wang ZJ, Wu JC, et al. Role of overexpression of CD151 and/or c-Met in predicting prognosis of hepatocellular carcinoma. Hepatology. 2009;49:491–503.PubMedCrossRef
74.
go back to reference Wang ZL, Liang P, Dong BW, Yu XL, Yu DJ. Prognostic factors and recurrence of small hepatocellular carcinoma after hepatic resection or microwave ablation: a retrospective study. J Gastrointest Surg. 2008;12:327–37.PubMedCrossRef Wang ZL, Liang P, Dong BW, Yu XL, Yu DJ. Prognostic factors and recurrence of small hepatocellular carcinoma after hepatic resection or microwave ablation: a retrospective study. J Gastrointest Surg. 2008;12:327–37.PubMedCrossRef
75.
go back to reference Ueki T, Fujimoto J, Suzuki T, Yamamoto H, Okamoto E. Expression of hepatocyte growth factor and its receptor c-met proto-oncogene in hepatocellular carcinoma. Hepatology. 1997;25:862–6.PubMedCrossRef Ueki T, Fujimoto J, Suzuki T, Yamamoto H, Okamoto E. Expression of hepatocyte growth factor and its receptor c-met proto-oncogene in hepatocellular carcinoma. Hepatology. 1997;25:862–6.PubMedCrossRef
76.
go back to reference Viticchie G, PAJ M. c-Met and other cell surface molecules: interaction, activation and functional consequences. Biomedicine. 2015;3:46–70.CrossRef Viticchie G, PAJ M. c-Met and other cell surface molecules: interaction, activation and functional consequences. Biomedicine. 2015;3:46–70.CrossRef
77.
go back to reference You H, Ding W, Dang H, Jiang Y, Rountree CB. c-Met represents a potential therapeutic target for personalized treatment in hepatocellular carcinoma. Hepatology. 2011;54:879–89.PubMedPubMedCentralCrossRef You H, Ding W, Dang H, Jiang Y, Rountree CB. c-Met represents a potential therapeutic target for personalized treatment in hepatocellular carcinoma. Hepatology. 2011;54:879–89.PubMedPubMedCentralCrossRef
78.
go back to reference Yarden Y. The EGFR family and its ligands in human cancer. Signalling mechanisms and therapeutic opportunities. Eur J Cancer. 2001;37(Suppl 4):S3–8.PubMedCrossRef Yarden Y. The EGFR family and its ligands in human cancer. Signalling mechanisms and therapeutic opportunities. Eur J Cancer. 2001;37(Suppl 4):S3–8.PubMedCrossRef
79.
go back to reference Merchant M, Ma X, Maun HR, Zheng Z, Peng J, Romero M, Huang A, Yang NY, Nishimura M, Greve J, et al. Monovalent antibody design and mechanism of action of onartuzumab, a MET antagonist with anti-tumor activity as a therapeutic agent. Proc Natl Acad Sci U S A. 2013;110:E2987–96.PubMedPubMedCentralCrossRef Merchant M, Ma X, Maun HR, Zheng Z, Peng J, Romero M, Huang A, Yang NY, Nishimura M, Greve J, et al. Monovalent antibody design and mechanism of action of onartuzumab, a MET antagonist with anti-tumor activity as a therapeutic agent. Proc Natl Acad Sci U S A. 2013;110:E2987–96.PubMedPubMedCentralCrossRef
80.
go back to reference Gelsomino F, Facchinetti F, Haspinger ER, Garassino MC, Trusolino L, De Braud F, Tiseo M. Targeting the MET gene for the treatment of non-small-cell lung cancer. Crit Rev Oncol Hematol. 2014;89:284–99.PubMedCrossRef Gelsomino F, Facchinetti F, Haspinger ER, Garassino MC, Trusolino L, De Braud F, Tiseo M. Targeting the MET gene for the treatment of non-small-cell lung cancer. Crit Rev Oncol Hematol. 2014;89:284–99.PubMedCrossRef
81.
go back to reference Camidge DR, Ou SHI, Shapiro G, Otterson GA, Villaruz LC, Villalona-Calero MA. Efficacy and safety of crizotinib in patients with advanced c-MET-amplified non-small cell lung cancer (NSCLC). 2014. Camidge DR, Ou SHI, Shapiro G, Otterson GA, Villaruz LC, Villalona-Calero MA. Efficacy and safety of crizotinib in patients with advanced c-MET-amplified non-small cell lung cancer (NSCLC). 2014.
82.
go back to reference Shea M, Costa DB, Rangachari D. Management of advanced non-small cell lung cancers with known mutations or rearrangements: latest evidence and treatment approaches. Ther Adv Respir Dis. 2016;10:113–29.PubMedCrossRef Shea M, Costa DB, Rangachari D. Management of advanced non-small cell lung cancers with known mutations or rearrangements: latest evidence and treatment approaches. Ther Adv Respir Dis. 2016;10:113–29.PubMedCrossRef
83.
go back to reference Facchinetti F, Rossi G, Bria E, Soria JC, Besse B, Minari R, Friboulet L, Tiseo M. Oncogene addiction in non-small cell lung cancer: focus on ROS1 inhibition. Cancer Treat Rev. 2017;55:83–95.PubMedCrossRef Facchinetti F, Rossi G, Bria E, Soria JC, Besse B, Minari R, Friboulet L, Tiseo M. Oncogene addiction in non-small cell lung cancer: focus on ROS1 inhibition. Cancer Treat Rev. 2017;55:83–95.PubMedCrossRef
84.
go back to reference Huang XX, Xie FF, Hou LJ, Chen XX, Ou RY, Yu JT, Qiu JG, Zhang WJ, Jiang QW, Yang Y, et al. Crizotinib synergizes with cisplatin in preclinical models of ovarian cancer. Am J Transl Res. 2017;9:1667–79.PubMedPubMedCentral Huang XX, Xie FF, Hou LJ, Chen XX, Ou RY, Yu JT, Qiu JG, Zhang WJ, Jiang QW, Yang Y, et al. Crizotinib synergizes with cisplatin in preclinical models of ovarian cancer. Am J Transl Res. 2017;9:1667–79.PubMedPubMedCentral
85.
go back to reference Stanley A, Ashrafi GH, Seddon AM, Modjtahedi H. Synergistic effects of various her inhibitors in combination with IGF-1R, c-Met and Src targeting agents in breast cancer cell lines. Sci Rep. 2017;7:3964.PubMedPubMedCentralCrossRef Stanley A, Ashrafi GH, Seddon AM, Modjtahedi H. Synergistic effects of various her inhibitors in combination with IGF-1R, c-Met and Src targeting agents in breast cancer cell lines. Sci Rep. 2017;7:3964.PubMedPubMedCentralCrossRef
86.
go back to reference Vergote IB, Smith DC, Berger R, Kurzrock R, Vogelzang NJ, Sella A, Wheler J, Lee Y, Foster PG, Weitzman R, Buckanovich RJ. A phase 2 randomised discontinuation trial of cabozantinib in patients with ovarian carcinoma. Eur J Cancer. 2017;83:229–36.PubMedCrossRef Vergote IB, Smith DC, Berger R, Kurzrock R, Vogelzang NJ, Sella A, Wheler J, Lee Y, Foster PG, Weitzman R, Buckanovich RJ. A phase 2 randomised discontinuation trial of cabozantinib in patients with ovarian carcinoma. Eur J Cancer. 2017;83:229–36.PubMedCrossRef
87.
go back to reference Escudier B, Lougheed JC, Albiges L. Cabozantinib for the treatment of renal cell carcinoma. Expert Opin Pharmacother. 2016;17:2499–504.PubMedCrossRef Escudier B, Lougheed JC, Albiges L. Cabozantinib for the treatment of renal cell carcinoma. Expert Opin Pharmacother. 2016;17:2499–504.PubMedCrossRef
88.
go back to reference Bentzien F, Zuzow M, Heald N, Gibson A, Shi Y, Goon L, Yu P, Engst S, Zhang W, Huang D, et al. In vitro and in vivo activity of cabozantinib (XL184), an inhibitor of RET, MET, and VEGFR2, in a model of medullary thyroid cancer. Thyroid. 2013;23:1569–77.PubMedPubMedCentralCrossRef Bentzien F, Zuzow M, Heald N, Gibson A, Shi Y, Goon L, Yu P, Engst S, Zhang W, Huang D, et al. In vitro and in vivo activity of cabozantinib (XL184), an inhibitor of RET, MET, and VEGFR2, in a model of medullary thyroid cancer. Thyroid. 2013;23:1569–77.PubMedPubMedCentralCrossRef
89.
go back to reference Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring PN, Rini BI, Hammers HJ, Donskov F, Roth BJ, Peltola K, et al. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. The Lancet Oncology. 2016;17:917–27.PubMedCrossRef Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring PN, Rini BI, Hammers HJ, Donskov F, Roth BJ, Peltola K, et al. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. The Lancet Oncology. 2016;17:917–27.PubMedCrossRef
90.
go back to reference Shintani T, Kusuhara Y, Daizumoto K, Dondoo TO, Yamamoto H, Mori H, Fukawa T, Nakatsuji H, Fukumori T, Takahashi M, Kanayama H. The involvement of Hepatocyte growth factor-MET-matrix metalloproteinase 1 Signaling in bladder cancer invasiveness and proliferation. Effect of the MET inhibitor, Cabozantinib (XL184), on bladder cancer cells. Urology. 2017;101:169 e167–13.CrossRef Shintani T, Kusuhara Y, Daizumoto K, Dondoo TO, Yamamoto H, Mori H, Fukawa T, Nakatsuji H, Fukumori T, Takahashi M, Kanayama H. The involvement of Hepatocyte growth factor-MET-matrix metalloproteinase 1 Signaling in bladder cancer invasiveness and proliferation. Effect of the MET inhibitor, Cabozantinib (XL184), on bladder cancer cells. Urology. 2017;101:169 e167–13.CrossRef
91.
go back to reference Shah MA, Wainberg ZA, Catenacci DV, Hochster HS, Ford J, Kunz P, Lee FC, Kallender H, Cecchi F, Rabe DC, et al. Phase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancer. PLoS One. 2013;8:e54014.PubMedPubMedCentralCrossRef Shah MA, Wainberg ZA, Catenacci DV, Hochster HS, Ford J, Kunz P, Lee FC, Kallender H, Cecchi F, Rabe DC, et al. Phase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancer. PLoS One. 2013;8:e54014.PubMedPubMedCentralCrossRef
92.
go back to reference Choueiri TK, Vaishampayan U, Rosenberg JE, Logan TF, Harzstark AL, Bukowski RM, Rini BI, Srinivas S, Stein MN, Adams LM, et al. Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma. J Clin Oncol. 2013;31:181–6.PubMedCrossRef Choueiri TK, Vaishampayan U, Rosenberg JE, Logan TF, Harzstark AL, Bukowski RM, Rini BI, Srinivas S, Stein MN, Adams LM, et al. Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma. J Clin Oncol. 2013;31:181–6.PubMedCrossRef
93.
go back to reference Chia SK, Ellard SL, Mates M, Welch S, Mihalcioiu C, Miller WH Jr, Gelmon K, Lohrisch C, Kumar V, Taylor S, et al. A phase-I study of lapatinib in combination with foretinib, a c-MET, AXL and vascular endothelial growth factor receptor inhibitor, in human epidermal growth factor receptor 2 (HER-2)-positive metastatic breast cancer. Breast Cancer Res. 2017;19:54.PubMedPubMedCentralCrossRef Chia SK, Ellard SL, Mates M, Welch S, Mihalcioiu C, Miller WH Jr, Gelmon K, Lohrisch C, Kumar V, Taylor S, et al. A phase-I study of lapatinib in combination with foretinib, a c-MET, AXL and vascular endothelial growth factor receptor inhibitor, in human epidermal growth factor receptor 2 (HER-2)-positive metastatic breast cancer. Breast Cancer Res. 2017;19:54.PubMedPubMedCentralCrossRef
94.
go back to reference Cheng H, Chua V, Liao C, Purwin TJ, Terai M, Kageyama K, Davies MA, Sato T, Aplin AE. Co-targeting HGF/cMET Signaling with MEK inhibitors in metastatic Uveal melanoma. Mol Cancer Ther. 2017;16:516–28.PubMedPubMedCentralCrossRef Cheng H, Chua V, Liao C, Purwin TJ, Terai M, Kageyama K, Davies MA, Sato T, Aplin AE. Co-targeting HGF/cMET Signaling with MEK inhibitors in metastatic Uveal melanoma. Mol Cancer Ther. 2017;16:516–28.PubMedPubMedCentralCrossRef
95.
go back to reference Barat S, Bozko P, Chen X, Scholta T, Hanert F, Gotze J, Malek NP, Wilkens L, Plentz RR. Targeting c-MET by LY2801653 for treatment of cholangiocarcinoma. Mol Carcinog. 2016;55:2037–50.PubMedCrossRef Barat S, Bozko P, Chen X, Scholta T, Hanert F, Gotze J, Malek NP, Wilkens L, Plentz RR. Targeting c-MET by LY2801653 for treatment of cholangiocarcinoma. Mol Carcinog. 2016;55:2037–50.PubMedCrossRef
96.
go back to reference Wang J, Cheng JX. c-Met inhibition enhances chemosensitivity of human ovarian cancer cells. Clin Exp Pharmacol Physiol. 2017;44:79–87.PubMedCrossRef Wang J, Cheng JX. c-Met inhibition enhances chemosensitivity of human ovarian cancer cells. Clin Exp Pharmacol Physiol. 2017;44:79–87.PubMedCrossRef
97.
go back to reference Liu X, Wang Q, Yang G, Marando C, Koblish HK, Hall LM, Fridman JS, Behshad E, Wynn R, Li Y, et al. A novel kinase inhibitor, INCB28060, blocks c-MET-dependent signaling, neoplastic activities, and cross-talk with EGFR and HER-3. Clin Cancer Res. 2011;17:7127–38.PubMedCrossRef Liu X, Wang Q, Yang G, Marando C, Koblish HK, Hall LM, Fridman JS, Behshad E, Wynn R, Li Y, et al. A novel kinase inhibitor, INCB28060, blocks c-MET-dependent signaling, neoplastic activities, and cross-talk with EGFR and HER-3. Clin Cancer Res. 2011;17:7127–38.PubMedCrossRef
98.
go back to reference Okusaka T, Aramaki T, Inaba Y, Nakamura S, Morimoto M, Moriguchi M, Sato T, Ikawa Y, Ikeda M, Furuse J. Phase I study of tivantinib in Japanese patients with advanced hepatocellular carcinoma: distinctive pharmacokinetic profiles from other solid tumors. Cancer Sci. 2015;106:611–7.PubMedPubMedCentralCrossRef Okusaka T, Aramaki T, Inaba Y, Nakamura S, Morimoto M, Moriguchi M, Sato T, Ikawa Y, Ikeda M, Furuse J. Phase I study of tivantinib in Japanese patients with advanced hepatocellular carcinoma: distinctive pharmacokinetic profiles from other solid tumors. Cancer Sci. 2015;106:611–7.PubMedPubMedCentralCrossRef
99.
go back to reference Xiang Q, Zhen Z, Deng DY, Wang J, Chen Y, Li J, Zhang Y, Wang F, Chen N, Chen H, Chen Y. Tivantinib induces G2/M arrest and apoptosis by disrupting tubulin polymerization in hepatocellular carcinoma. J Exp Clin Cancer Res. 2015;34:118.PubMedPubMedCentralCrossRef Xiang Q, Zhen Z, Deng DY, Wang J, Chen Y, Li J, Zhang Y, Wang F, Chen N, Chen H, Chen Y. Tivantinib induces G2/M arrest and apoptosis by disrupting tubulin polymerization in hepatocellular carcinoma. J Exp Clin Cancer Res. 2015;34:118.PubMedPubMedCentralCrossRef
100.
go back to reference Best J, Schotten C, Lohmann G, Gerken G, Dechene A. Tivantinib for the treatment of hepatocellular carcinoma. Expert Opin Pharmacother. 2017;18:727–33.PubMedCrossRef Best J, Schotten C, Lohmann G, Gerken G, Dechene A. Tivantinib for the treatment of hepatocellular carcinoma. Expert Opin Pharmacother. 2017;18:727–33.PubMedCrossRef
101.
go back to reference Sequist LV, von Pawel J, Garmey EG, Akerley WL, Brugger W, Ferrari D, Chen Y, Costa DB, Gerber DE, Orlov S, et al. Randomized phase II study of erlotinib plus tivantinib versus erlotinib plus placebo in previously treated non-small-cell lung cancer. J Clin Oncol. 2011;29:3307–15.PubMedCrossRef Sequist LV, von Pawel J, Garmey EG, Akerley WL, Brugger W, Ferrari D, Chen Y, Costa DB, Gerber DE, Orlov S, et al. Randomized phase II study of erlotinib plus tivantinib versus erlotinib plus placebo in previously treated non-small-cell lung cancer. J Clin Oncol. 2011;29:3307–15.PubMedCrossRef
102.
go back to reference Yoshioka H, Azuma K, Yamamoto N, Takahashi T, Nishio M, Katakami N, Ahn MJ, Hirashima T, Maemondo M, Kim SW, et al. A randomized, double-blind, placebo-controlled, phase III trial of erlotinib with or without a c-Met inhibitor tivantinib (ARQ 197) in Asian patients with previously treated stage IIIB/IV nonsquamous nonsmall-cell lung cancer harboring wild-type epidermal growth factor receptor (ATTENTION study). Ann Oncol. 2015;26:2066–72.PubMedCrossRef Yoshioka H, Azuma K, Yamamoto N, Takahashi T, Nishio M, Katakami N, Ahn MJ, Hirashima T, Maemondo M, Kim SW, et al. A randomized, double-blind, placebo-controlled, phase III trial of erlotinib with or without a c-Met inhibitor tivantinib (ARQ 197) in Asian patients with previously treated stage IIIB/IV nonsquamous nonsmall-cell lung cancer harboring wild-type epidermal growth factor receptor (ATTENTION study). Ann Oncol. 2015;26:2066–72.PubMedCrossRef
103.
go back to reference Scagliotti G, von Pawel J, Novello S, Ramlau R, Favaretto A, Barlesi F, Akerley W, Orlov S, Santoro A, Spigel D, et al. Phase III multinational, randomized, double-blind, placebo-controlled study of Tivantinib (ARQ 197) plus Erlotinib versus Erlotinib alone in previously treated patients with locally advanced or metastatic Nonsquamous non-small-cell lung cancer. J Clin Oncol. 2015;33:2667–74.PubMedCrossRef Scagliotti G, von Pawel J, Novello S, Ramlau R, Favaretto A, Barlesi F, Akerley W, Orlov S, Santoro A, Spigel D, et al. Phase III multinational, randomized, double-blind, placebo-controlled study of Tivantinib (ARQ 197) plus Erlotinib versus Erlotinib alone in previously treated patients with locally advanced or metastatic Nonsquamous non-small-cell lung cancer. J Clin Oncol. 2015;33:2667–74.PubMedCrossRef
104.
go back to reference Puzanov I, Sosman J, Santoro A, Saif MW, Goff L, Dy GK, Zucali P, Means-Powell JA, Ma WW, Simonelli M, et al. Phase 1 trial of tivantinib in combination with sorafenib in adult patients with advanced solid tumors. Investig New Drugs. 2015;33:159–68.CrossRef Puzanov I, Sosman J, Santoro A, Saif MW, Goff L, Dy GK, Zucali P, Means-Powell JA, Ma WW, Simonelli M, et al. Phase 1 trial of tivantinib in combination with sorafenib in adult patients with advanced solid tumors. Investig New Drugs. 2015;33:159–68.CrossRef
105.
go back to reference Klempner SJ, Borghei A, Hakimian B, Ali SM, Ou SI. Intracranial activity of Cabozantinib in MET exon 14-positive NSCLC with brain metastases. J Thorac Oncol. 2017;12:152–6.PubMedCrossRef Klempner SJ, Borghei A, Hakimian B, Ali SM, Ou SI. Intracranial activity of Cabozantinib in MET exon 14-positive NSCLC with brain metastases. J Thorac Oncol. 2017;12:152–6.PubMedCrossRef
106.
go back to reference Pedersen L, Christensen JF, Hojman P. Effects of exercise on tumor physiology and metabolism. Cancer J. 2015;21:111–6.PubMedCrossRef Pedersen L, Christensen JF, Hojman P. Effects of exercise on tumor physiology and metabolism. Cancer J. 2015;21:111–6.PubMedCrossRef
107.
go back to reference Corbet C, Feron O. Tumour acidosis: from the passenger to the driver's seat. Nat Rev Cancer. 2017;17:577–93.PubMedCrossRef Corbet C, Feron O. Tumour acidosis: from the passenger to the driver's seat. Nat Rev Cancer. 2017;17:577–93.PubMedCrossRef
108.
go back to reference Fan C, Tang Y, Wang J, Xiong F, Guo C, Wang Y, Zhang S, Gong Z, Wei F, Yang L, et al. Role of long non-coding RNAs in glucose metabolism in cancer. Mol Cancer. 2017;16:130.PubMedPubMedCentralCrossRef Fan C, Tang Y, Wang J, Xiong F, Guo C, Wang Y, Zhang S, Gong Z, Wei F, Yang L, et al. Role of long non-coding RNAs in glucose metabolism in cancer. Mol Cancer. 2017;16:130.PubMedPubMedCentralCrossRef
109.
110.
go back to reference Song Y, Li X, Zeng Z, Li Q, Gong Z, Liao Q, Li X, Chen P, Xiang B, Zhang W, et al. Epstein-Barr virus encoded miR-BART11 promotes inflammation-induced carcinogenesis by targeting FOXP1. Oncotarget. 2016;7:36783–99.PubMedPubMedCentral Song Y, Li X, Zeng Z, Li Q, Gong Z, Liao Q, Li X, Chen P, Xiang B, Zhang W, et al. Epstein-Barr virus encoded miR-BART11 promotes inflammation-induced carcinogenesis by targeting FOXP1. Oncotarget. 2016;7:36783–99.PubMedPubMedCentral
Metadata
Title
Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities
Authors
Yazhuo Zhang
Mengfang Xia
Ke Jin
Shufei Wang
Hang Wei
Chunmei Fan
Yingfen Wu
Xiaoling Li
Xiayu Li
Guiyuan Li
Zhaoyang Zeng
Wei Xiong
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2018
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0796-y

Other articles of this Issue 1/2018

Molecular Cancer 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

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

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