Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-2lccl Total loading time: 0 Render date: 2024-04-29T01:12:41.415Z Has data issue: false hasContentIssue false

17 - Hepatocyte growth factor/Met signaling in cancer

from Part 2.1 - Molecular pathways underlying carcinogenesis: signal transduction

Published online by Cambridge University Press:  05 February 2015

Fabiola Cecchi
Affiliation:
Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda,MD, USA
Young H. Lee
Affiliation:
Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
Donald P. Bottaro
Affiliation:
Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Hepatocyte growth factor (HGF), also known as scatter factor (SF), was discovered on the basis of its ability to promote liver regeneration, and independently for its mitogenic activity on epithelial cells and its ability to induce cell scatter (1). HGF is secreted primarily by mesenchymal cells and drives cell motility, proliferation, survival, and morphogenesis by binding to the Met receptor tyrosine kinase (TK) present on a variety of target cell types (1–6). HGF/Met signaling is critical for normal development and adult homeostasis: deletion of either gene lethally disrupts embryogenesis (4,6) and up-regulation of HGF expression after kidney, liver, or heart injury protects against tissue damage and promotes repair and regeneration in adults (1,7–11). Under normal conditions, Met activation is tightly regulated by paracrine ligand delivery, ligand activation, and receptor internalization, dephosphorylation, and degradation (1). Despite this, HGF/Met signaling contributes to tumorigenesis, tumor angiogenesis, and metastasis in several prevalent cancers, a realization that has driven rapid growth in the development of experimental therapeutics targeting the pathway.

HGF and Met structure and function

The human HGF gene consists of 18 exons and 16 introns spanning 68 Mb on chromosome 7q21.11 (1). Five mRNA transcripts arise from alternative splicing: two encode full-length HGF forms and three encode truncated isoforms that bind Met, but differ in their biological activities (1). HGF protein is a plasminogen family member consisting of an amino-terminal heparin-binding domain (N), four kringle domains (K1–4) and a carboxyl-terminal serine-protease-like domain (Figure 17.1a). Unlike other plasminogen family members, HGF has no proteolytic activity (1). The HGF N and K1 domains contain the primary Met binding sites (12), and the protease-like domain contains an important secondary Met binding site (13). Proteolytic processing of the single-chain HGF precursor results in the active disulfide-linked heterodimer; the amino-terminal α-chain contains N and K1–4, and the β-chain contains the protease-like region (1).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 204 - 217
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Rubin, JS, Bottaro, DP. UCSD Molecule Pages: HGF. URL: (Accessed September 2013.)
Peschard, P, Park, M. From Tpr-Met to Met, tumorigenesis and tubes. Oncogene 2007;26:1276–85.CrossRef
Corso, S, Comoglio, PM, Giordano, S. Cancer therapy: can the challenge be MET? Trends in Molecular Medicine 2005;11:284–92.
Rosario, M, Birchmeier, W. How to make tubes: signaling by the Met receptor tyrosine kinase. Trends in Cell Biology 2003;13:328–35.CrossRef
Zhang, YW and Vande, Woude GF.HGF/SF-met signaling in the control of branching morphogenesis and invasion. Journal of Cell Biochemistry 2003;88:408–17.CrossRefGoogle ScholarPubMed
Birchmeier, C, Birchmeier, W, Gherardi, E, Vande, Woude GF. Met, metastasis, motility and more. Nature Reviews Molecular and Cellular Biology 2003;4:915–25.CrossRef
Liu, Y. Renal fibrosis: new insights into the pathogenesis and therapeutics. Kidney International 2006;69:213–17.CrossRef
Borowiak, M, Garratt, AN, Wustefeld, T, et al. Met provides essential signals for liver regeneration. Proceedings of the National Academy of Sciences USA 2004;101:10 608–13.
Huh, CG, Factor, VM, Sanchez, A, et al. Hepatocyte growth factor/c-met signaling pathway is required for efficient liver regeneration and repair. Proceedings of the National Academy of Sciences USA 2004;101:4477–82.CrossRef
Morishita, R, Aoki, M, Hashiya, N, et al. Therapeutic angiogenesis using hepatocyte growth factor (HGF). Current Gene Therapy 2004;4:199–206.CrossRef
Matsumoto, K, Nakamura, T. Hepatocyte growth factor: renotropic role and potential therapeutics for renal diseases. Kidney International 2001;59:2023–38.CrossRef
Lokker, NA, Mark, MR, Luis, EA, et al. Structure-function analysis of hepatocyte growth factor: identification of variants that lack mitogenic activity yet retain high affinity receptor binding. EMBO Journal1992;11:2503–10.
Niemann, HH, Jager, V, Butler, PJ, et al. Structure of the human receptor tyrosine kinase met in complex with the Listeria invasion protein InlB. Cell 2007;130:235–46.CrossRef
Dharmawardana, PG, Giubellino, A, Bottaro, DP. Hereditary papillary renal carcinoma type I. Current Molecular Medicine 2004;4:855–68.CrossRef
Liu, Y. The human hepatocyte growth factor receptor gene: complete structural organization and promoter characterization. Gene 1998;215(1):159–69.CrossRef
Gherardi, E, Youles, ME, Miguel, RN, et al. Functional map and domain structure of MET, the product of the c-met protooncogene and receptor for hepatocyte growth factor/scatter factor. Proceedings of the National Academy of Sciences USA 2003;100:12 039–44.
Antipenko, A, Himanen, JP, van Leyen, K, et al. Structure of the semaphorin-3A receptor binding module. Neuron 2003;39:589–98.CrossRef
Love, CA, Harlos, K, Mavaddat, N, et al. The ligand-binding face of the semaphorins revealed by the high-resolution crystal structure of SEMA4D. Nature Structural Biology 2003;10:843–8.CrossRef
Basilico, C, Arnesano, A, Galluzzo, M, et al. A high affinity hepatocyte growth factor-binding site in the immunoglobulin-like region of Met. Journal of Biological Chemistry 2008;283:21 267–77.CrossRefGoogle ScholarPubMed
Ma, PC, Maulik, G, Christensen, J, Salgia, R. c-Met: structure, functions and potential for therapeutic inhibition. Cancer Metastasis Reviews 2003;22:309–25.CrossRef
Villa-Moruzzi, E, Puntoni, F, Bardelli, A, et al. Protein tyrosine phosphatase PTP-S binds to the juxtamembrane region of the hepatocyte growth factor receptor Met. Biochemical Journal 1998;336:235–9.CrossRef
Comoglio, PM, Boccaccio, C. Scatter factors and invasive growth. Seminars in Cancer Biology 2001;11:153–65.CrossRef
Giordano, S, Bardelli, A, Zhen, Z, et al. A point mutation in the MET oncogene abrogates metastasis without affecting transformation. Proceedings of the National Academy of Sciences USA 1997;94:13 868–72.
Gu, H, Neel, BG. The “Gab” in signal transduction. Trends in Cell Biology 2003;13:122–30.CrossRef
Van Andel Research Institute, Research Tools: Met. URL: (Accessed September 2013.)
Lengyel, E, Sawada, K, Salgia, R. Tyrosine kinase mutations in human cancer. Current Molecular Medicine 2007;7:77–84.CrossRef
Bardelli, A, Longati, P, Gramaglia, D, et al. Uncoupling signal transducers from oncogenic MET mutants abrogates cell transformation and inhibits invasive growth. Proceedings of the National Academy of Sciences USA 1998;95:14 379–83.
Giordano, S, Maffe, A, Williams, TA, et al. Different point mutations in the met oncogene elicit distinct biological properties. FASEB Journal 2000;14:399–406.CrossRef
Michieli, P, Basilico, C, Pennacchietti, S, et al. Mutant Met-mediated transformation is ligand-dependent and can be inhibited by HGF antagonists. Oncogene 1999;18:5221–31.CrossRef
Graveel, CR, London, CA, Vande Woude GF. A mouse model of activating Met mutations. Cell Cycle 2005;4518–20.
Joffre, C, Barrow, R, Menard, L, et al. A direct role for Met endocytosis in tumorigenesis. Nature Cell Biology 2011;13:827–37.CrossRef
Kong-Beltran, M, Seshagiri, S, Zha, J, et al. Somatic mutations lead to an oncogenic deletion of met in lung cancer. Cancer Research 2006;66:283–9.CrossRef
Ma, PC, Kijima, T, Maulik, G, et al. c-MET mutational analysis in small cell lung cancer: novel juxtamembrane domain mutations regulating cytoskeletal functions. Cancer Research 2003;63:6272–81.
Ma, PC, Jagadeeswaran, R, Jagadeesh, S, Tretiakova, MS, et al. Functional expression and mutations of c-Met and its therapeutic inhibition with SU11274 and small interfering RNA in non-small cell lung cancer. Cancer Research 2005; 65:1479–88.CrossRef
Lee, JH, Han, SU, Cho, H, et al. A novel germ line juxtamembrane Met mutation in human gastric cancer. Oncogene 2000; 19:4947–53.CrossRef
Tyner, JW, Fletcher, LB, Wang, EQ, et al. MET receptor sequence variants R970C and T992I lack transforming capacity. Cancer Research 2010;70:6233–7.CrossRef
Corso, S, Migliore, C, Ghiso, E, et al. Silencing the MET oncogene leads to regression of experimental tumors and metastases. Oncogene 2008;27:684–93.CrossRef
Pennacchietti, S, Michieli, P, Galluzzo, M, et al. Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell 2003;3:347–61.CrossRef
Peruzzi, B, Athauda, G, Bottaro, DP. The von Hippel-Lindau tumor suppressor gene product represses oncogenic beta-catenin signaling in renal carcinoma cells. Proceedings of the National Academy of Sciences USA 2006;103:14 531–6.
Cecchi, F, Wright, C, Bottaro, DP. Experimental cancer therapeutics targeting the hepatocyte growth factor/Met signaling pathway. Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892 USA. URL: (Accessed March 25, 2013.)
Jun, HT, Sun, J, Rex, K, et al. AMG 102, a fully human anti-hepatocyte growth factor/scatter factor neutralizing antibody, enhances the efficacy of temozolomide or docetaxel in U-87 MG cells and xenografts. Clinical Cancer Research 2007;13:6735–42.CrossRef
Wen, PY, Schiff, D, Cloughesy, TF, et al. A Phase II study evaluating the efficacy and safety of AMG 102 (rilotumumab) in patients with recurrent glioblastoma. Neurological Oncology 2011;13:437–46.
Eng, C, Van, Cutsem E, Nowara, E, et al. A randomized, Phase Ib/II trial of rilotumumab (AMG 102; ril) or ganitumab (AMG 479; gan) with panitumumab (pmab) versus pmab alone in patients (pts) with wild-type (WT) KRAS metastatic colorectal cancer (mCRC): Primary and biomarker analyses. Journal of Clinical Oncology 29, 2011 (Suppl. Abstract 3500), American Society of Clinical Oncology (ASCO) Annual Meeting; Chicago, IL, 2011.CrossRefGoogle Scholar
Jones, SF, Cohen, RB, Bendell, JC, et al. Safety, tolerability, and pharmacokinetics of TAK-701, a humanized anti-hepatocyte growth factor (HGF) monoclonal antibody, in patients with advanced nonhematologic malignancies: First-in-human Phase I dose-escalation study. Journal of Clinical Oncology 28:15s, 2010 (Suppl. Abstract 3081), American Society of Clinical Oncology (ASCO) Annual Meeting; Chicago, IL, 2010.CrossRefGoogle Scholar
Tan, K, Park, K, Lim, M, et al. Phase Ib study of ficlatuzumab (formerly AV-299), an anti-hepatocyte growth factor (HGF) monoclonal antibody (MAb) in combination with gefitinib (G) in Asian patients (pts) with NSCLC. Journal of Clinical Oncology 29: 2011 (Suppl. Abstract 7571), American Society of Clinical Oncology (ASCO) Annual Meeting; Chicago, IL, 2011.CrossRefGoogle Scholar
Martens, T, Schmidt, NO, Eckerich, C, et al. A novel one-armed anti-c-Met antibody inhibits glioblastoma growth in vivo. Clinical Cancer Research 2006;12:6144–52.CrossRef
US Food and Drug Administration. Cabozantinib. URL: (Accessed March 25, 2013.)
Traynor, K.Cabozantinib approved for advanced medullary thyroid cancer. American Journal of Health-System Pharmacy 2013;70:88.Google ScholarPubMed
Gordon, MS, Vogelzang, NJ, Schoffski, P, et al. Activity of cabozantinib (XL184) in soft tissue and bone: results of a Phase II randomized discontinuation trial (RDT) in patients (pts) with advanced solid tumors. Journal of Clinical Oncology 2011;29:(Suppl.; Abstract 3010).CrossRefGoogle Scholar
Wen, P, et al. Phase II study of XL184 (BMS 907351), an inhibitor of MET, VEGFR2, and RET, in patients (pts) with progressive glioblastoma (GB). Journal of Clinical Oncology 2010;28 (Suppl. Abstract 2006). American Society of Clinical Oncology (ASCO) Annual Meeting; Chicago, IL, 2010.CrossRefGoogle Scholar
Vaishampayan, U. Cabozantinib as a novel therapy for renal cell carcinoma. Current Oncology Reports. 2013;15:76–82.CrossRef
Christensen, JG, Zou, HY, Arango, ME, et al. Cytoreductive antitumor activity of PF-2341066, a novel inhibitor of anaplastic lymphoma kinase and c-Met, in experimental models of anaplastic large-cell lymphoma. Molecular Cancer Therapeutics 2007;6:3314–22.CrossRef
Kwak, EL, Bang, YJ, Camidge, DR, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. New England Journal of Medicine 2010;363:1693–1703.CrossRefGoogle ScholarPubMed
Munshi, N, Jeay, S, Li, Y, et al. ARQ 197, a novel and selective inhibitor of the human c-Met receptor tyrosine kinase with antitumor activity. Molecular Cancer Therapeutics 2010;9:1544–53.CrossRef
(Accessed September 2013.)
Wagner, AJ, Goldberg, JM, Dubois, SG, et al. Tivantinib (ARQ 197), a selective inhibitor of MET, in patients with microphthalmia transcription factor-associated tumors: results of a multicenter phase 2 trial. Cancer 2012;118:5894–902.CrossRef
(Accessed September 2013.)
Santoro, A, Rimassa, L, Borbath, I, et al. Tivantinib for second-line treatment of advanced hepatocellular carcinoma: a randomised, placebo-controlled phase 2 study. Lancet Oncology 2013;14:55–63.CrossRef
Choueiri, TK, Vaishampayan, U, Rosenberg, JE, et al. Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma. Journal of Clinical Oncology 2013;31:181–6.CrossRefGoogle ScholarPubMed
Liu, L, Shi, H, Liu, Y, et al. Synergistic effects of foretinib with HER-targeted agents in MET and HER1- or HER2-coactivated tumor cells. Molecular Cancer Therapeutics 2011;10:518–30.CrossRef
Nakagawa, T, Tohyama, O, Yamaguchi, A, et al. E7050: a dual c-Met and VEGFR-2 tyrosine kinase inhibitor promotes tumor regression and prolongs survival in mouse xenograft models. Cancer Science 2010;101:210–15.CrossRef
Qi, W, Cooke, LS, Stejskal, A, et al. MP470, a novel receptor tyrosine kinase inhibitor, in combination with Erlotinib inhibits the HER family/PI3K/Akt pathway and tumor growth in prostate cancer. BMC Cancer 2009;9:142.CrossRef
Welsh, P, Mahadevan, D, Bearss, D, Stea, B.Sensitization of a glioblastoma multiforme (GBM) cell line by MP470: a novel c-Met antagonist. International Journal of Radiation Oncology 2007;69:S100.CrossRefGoogle Scholar
Hong, D, LoRusso, P, Kurzrock, R, et al. Phase I study of MGCD265 administered intermittently to patients with advanced malignancies (Study 265–102). Journal of Clinical Oncology (Meeting Abstracts) 2009; 27:e14516. American Society of Clinical Oncology (ASCO), Annual Meeting; Chicago, IL, 2009.Google Scholar
Kollmannsberger, CK, Hurwitz, H, Vlahovic, G, et al. Phase I study of daily administration of MGCD265 to patients with advanced malignancies (Study 265–101). Journal of Clinical Oncology (Meeting Abstracts) 2009; 27:e14525. American Society of Clinical Oncology (ASCO), Annual Meeting; Chicago, IL, 2009.Google Scholar
Donehower, RC, Sacrdina, M, Hill, M, et al. A Phase I dose-escalation study of INCB028060, an inhibitor of c-MET receptor tyrosine kinase, in patients with advanced solid tumors. Journal of Clinical Oncology 29 (Suppl. Abstract 3091) American Society of Clinical Oncology (ASCO), Annual Meeting; Chicago, IL, 2011.CrossRefGoogle Scholar
Yang, WJ, Credille, K, Gao, H, et al. LY2801653, an orally available small molecule inhibitor of c-Met, demonstrated broad antitumor efficacy in patients derived xenograft models. Cancer Research 2010;70: Suppl. 1, American Association for Cancer Research (AACR) Annual Meeting; Washington, DC, 2010.
Diamond, S, Boer, J, Maduskuie, TP, et al. Species-specific metabolism of SGX523 by aldehyde oxidase and the toxicological implications. Drug Metabolism and Disposition 2010;38:1277–85.CrossRef
Timofeevski, SL, McTigue, MA., Ryan, K., et al. Enzymatic characterization of c-Met receptor tyrosine kinase oncogenic mutants and kinetic studies with aminopyridine and triazolopyrazine inhibitors. Biochemistry 2009;48:5339–49.CrossRef
Qi, J, McTigue, MA, Rogers, A, et al. Multiple mutations and bypass mechanisms can contribute to development of acquired resistance to MET inhibitors. Cancer Research. 2011;71:1081–91.CrossRef
Peach, ML, Tan, N, Choyke, S, et al. Directed discovery of agents targeting the Met tyrosine kinase domain by virtual screening. Journal of Medicinal Chemistry 2009;52:943–51.CrossRefGoogle ScholarPubMed
Ross, RW, Stein, M, Sarantopoulos, J, et al. A Phase II study of the c-Met RTK inhibitor XL880 in patients (pts) with papillary renal-cell carcinoma (PRC). Journal of Clinical Oncology, ASCO Annual Meeting Proceedings Part I. Vol 25, No. 18S (June 20 Supplement): 15601, American Society of Clinical Oncology (ASCO), Annual Meeting; Chicago, IL, 2007.Google Scholar
Srinivasan, R, Choueiri, TK, Vaishampayan, U, et al. A Phase II study of the dual MET/VEGFR2 inhibitor XL880 in patients (pts) with papillary renal carcinoma (PRC). Journal of Clinical Oncology 26 (Suppl. Abstract 5103), American Society of Clinical Oncology (ASCO), Annual Meeting; Chicago, IL, 2008.CrossRefGoogle Scholar
Cecchi, F, Liu, Y, Gagnon, RC, et al. ShedMET (sMet), VEGFA, and sVEGFR2 are markers of foretinib treatment in metastatic gastric cancer patients. Molecular Cancer Therapeutics 2009;8 Suppl. 1, AACR-NCI-EORTC International Conference Molecular Target and Cancer Therapeutics; Boston, MA, 2009.
Kollmannsberger, CK, Hurwitz, H, Vlahovic, G, et al. Phase I study of daily administration of MGCD265 to patients with advanced malignancies (study 265–101). Journal of Clinical Oncology 27 (Suppl. Abstract e14525), American Society of Clinical Oncology (ASCO) Annual Meeting; Chicago, IL, 2009.Google Scholar
DePrimo, S, Wu, B, Huang, S, et al. Correlative tumor molecular profiling and plasma biomarker analysis in a Phase II study of XL184 in patients with progressive or recurrent glioblastoma multiforme (GBM). Journal of Clinical Oncology 27:15s, 2 (Suppl. Abstract 2049), American Society of Clinical Oncology (ASCO), Annual Meeting; Chicago, IL, 2009.Google Scholar
Muller, T.DePrimo, S.McGrath, G, Yu, P, et al. Pharmacodynamic and correlative biomarker analyses in clinical trials of XL184 and oral, potent inhibitor of MET VEGFR2, and RET. AACR-NCI-EORTC International Conference Molecular Target and Cancer Therapeutics; Boston, MA, 2009.
Pena, C, Shan, M, Bukowski, RM, et al. Plasma biomarkers predicting outcome in patients with advanced RCC: results from the sorafenib Phase II TARGET trial. AACR-NCI-EORTC International Conference Molecular Target and Cancer Therapeutics; Boston, MA, 2009.
Heymach, JV, Fritsche, HA, Gornet, TG, et al. Lower baseline levels of plasm hepatocyte growth factor, IL-6, IL-8 are correlated with greater tumor shrinkage in renal cell carcinoma patients treated with pazopanib. Molecular Cancer Therapeutics 2009;8, Suppl. 1, AACR-NCI-EORTC International Conference Molecular Target and Cancer Therapeutics; Boston, MA, 2009.
Tanimoto, S, Fukumori, T, El-Moula, G, et al. Prognostic significance of serum hepatocyte growth factor in clear cell renal cell carcinoma: comparison with serum vascular endothelial growth factor. Journal of Medical Investigation 2008;55:106–11.CrossRefGoogle ScholarPubMed
Yano, S, Wang, W., Li, Q, et al. Hepatocyte growth factor induces gefitinib resistance of lung adenocarcinoma with epidermal growth factor receptor-activating mutations. Cancer Research 2008;68:9479–87.CrossRef
Engelman, JA, Zejnullahu, K., Misudomi, T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007;316:1039–43.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Hepatocyte growth factor/Met signaling in cancer
    • By Fabiola Cecchi, Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda,MD, USA, Young H. Lee, Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA, Donald P. Bottaro, Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.018
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Hepatocyte growth factor/Met signaling in cancer
    • By Fabiola Cecchi, Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda,MD, USA, Young H. Lee, Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA, Donald P. Bottaro, Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.018
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Hepatocyte growth factor/Met signaling in cancer
    • By Fabiola Cecchi, Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda,MD, USA, Young H. Lee, Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA, Donald P. Bottaro, Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.018
Available formats
×