Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Original Article
  • Published:

Effective inhibition of c-MET-mediated signaling, growth and migration of ovarian cancer cells is influenced by the ovarian tissue microenvironment

Abstract

The signaling mediated by c-MET and its ligand, hepatocyte growth factor (HGF), has been implicated in malignant progression of cancer involving stimulation of proliferation, invasion and metastasis. We studied the c-MET/HGF axis as a mediator of tumor–stromal interaction in ovarian cancer and the value of targeting c-MET for the treatment of ovarian cancer. To assess c-MET signaling, we established in vitro models of the microenvironment using primary and immortalized human fibroblasts from normal ovary and tumor samples and epithelial ovarian cancer cell lines. We found that fibroblast from normal ovaries secreted high levels of HGF (1500–3800 pg/ml) as compared with tumor-derived fibroblasts (undetectable level) and could elicit cellular biological responses on c-MET-expressing ovarian cancer cells including increase of cell proliferation and migration (2- to 140-fold increase). HGF secreted by fibroblasts was also found sequestered within extracellular matrices (ECMs) and when degraded this ECM-derived HGF stimulated cancer cell migration (1.5- to 24-fold). In cells containing constitutive c-MET phosphorylation, recombinant HGF and fibroblast-derived HGF negligibly affect c-MET phosphorylation on Tyr1234 and Tyr1003. However, both sources of HGF increased the phosphorylation of c-MET on Tyr1349, the multi-substrate docking site, by more than sixfold and led to activation of downstream signaling transducers. DCC-2701 (Deciphera Pharmaceuticals, LLC), a novel c-MET/TIE-2/VEGFR inhibitor was able to effectively reduce tumor burden in vivo and block c-MET pTyr1349-mediated signaling, cell growth and migration as compared with a HGF antagonist in vitro. Importantly, DCC-2701’s anti-proliferative activity was dependent on c-MET activation induced by stromal human fibroblasts and to a lesser extent exogenous HGF. Our data suggest for the first time that DCC-2701 may be superior to HGF antagonists that are in clinical trials and that pTyr1349 levels might be a good indicator of c-MET activation and likely response to targeted therapy as a result of signals from the microenvironment.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6
Figure 7

Similar content being viewed by others

References

  1. Siegel R, Naishadham D, Jemal A . Cancer statistics, 2013. CA Cancer J Clin 2013; 63: 11–30.

    Article  Google Scholar 

  2. Ozols RF, Bookman MA, Connolly DC, Daly MB, Godwin AK, Schilder RJ et al. Focus on epithelial ovarian cancer. Cancer Cell 2004; 5: 19–24.

    Article  CAS  Google Scholar 

  3. Bhowmick NA, Moses HL . Tumor-stroma interactions. Curr Opin Genet Dev 2005; 15: 97–101.

    Article  CAS  Google Scholar 

  4. Liotta LA, Kohn EC . The microenvironment of the tumour-host interface. Nature 2001; 411: 375–379.

    Article  CAS  Google Scholar 

  5. Beacham DA, Cukierman E . Stromagenesis: the changing face of fibroblastic microenvironments during tumor progression. Semin Cancer Biol 2005; 15: 329–341.

    Article  Google Scholar 

  6. Hynes RO . The extracellular matrix: not just pretty fibrils. Science 2009; 326: 1216–1219.

    Article  CAS  Google Scholar 

  7. Matsumoto K, Nakamura T . Hepatocyte growth factor and the Met system as a mediator of tumor-stromal interactions. Int J Cancer 2006; 119: 477–483.

    Article  CAS  Google Scholar 

  8. Sierra JR, Tsao MS . c-MET as a potential therapeutic target and biomarker in cancer. Ther Adv Med Oncol 2011; 3 (1 Suppl): S21–S35.

    Article  CAS  Google Scholar 

  9. Corps AN, Sowter HM, Smith SK . Hepatocyte growth factor stimulates motility, chemotaxis and mitogenesis in ovarian carcinoma cells expressing high levels of c-met. Int J Cancer 1997; 73: 151–155.

    Article  CAS  Google Scholar 

  10. Taipale J, Keski-Oja J . Growth factors in the extracellular matrix. FASEB J 1997; 11: 51–59.

    Article  CAS  Google Scholar 

  11. Tatsumi R, Allen RE . Active hepatocyte growth factor is present in skeletal muscle extracellular matrix. Muscle Nerve 2004; 30: 654–658.

    Article  CAS  Google Scholar 

  12. Comoglio PM, Giordano S, Trusolino L . Drug development of MET inhibitors: targeting oncogene addiction and expedience. Nat Rev Drug Discov 2008; 7: 504–516.

    Article  CAS  Google Scholar 

  13. Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, Du J et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 2012; 487: 500–504.

    Article  CAS  Google Scholar 

  14. Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature 2012; 487: 505–509.

    Article  CAS  Google Scholar 

  15. Ayhan A, Ertunc D, Tok EC, Ayhan A . Expression of the c-Met in advanced epithelial ovarian cancer and its prognostic significance. Int J Gynecol Cancer 2005; 15: 618–623.

    Article  CAS  Google Scholar 

  16. Zillhardt M, Christensen JG, Lengyel E . An orally available small-molecule inhibitor of c-Met, PF-2341066, reduces tumor burden and metastasis in a preclinical model of ovarian cancer metastasis. Neoplasia 2010; 12: 1–10.

    Article  CAS  Google Scholar 

  17. Zillhardt M, Park SM, Romero IL, Sawada K, Montag A, Krausz T et al. Foretinib (GSK1363089), an orally available multikinase inhibitor of c-Met and VEGFR-2, blocks proliferation, induces anoikis, and impairs ovarian cancer metastasis. Clin Cancer Res 2011; 17: 4042–4051.

    Article  CAS  Google Scholar 

  18. Gordon MS, Sweeney CS, Mendelson DS, Eckhardt SG, Anderson A, Beaupre DM et al. Safety, pharmacokinetics, and pharmacodynamics of AMG 102, a fully human hepatocyte growth factor-neutralizing monoclonal antibody, in a first-in-human study of patients with advanced solid tumors. Clin Cancer Res 2010; 16: 699–710.

    Article  CAS  Google Scholar 

  19. Liu X, Newton RC, Scherle PA . Developing c-MET pathway inhibitors for cancer therapy: progress and challenges. Trends Mol Med 2009; 16: 37–45.

    Article  Google Scholar 

  20. Lyon M, Deakin JA, Mizuno K, Nakamura T, Gallagher JT . Interaction of hepatocyte growth factor with heparan sulfate. Elucidation of the major heparan sulfate structural determinants. J Biol Chem 1994; 269: 11216–11223.

    CAS  PubMed  Google Scholar 

  21. Ashikari S, Habuchi H, Kimata K . Characterization of heparan sulfate oligosaccharides that bind to hepatocyte growth factor. J Biol Chem 1995; 270: 29586–29593.

    Article  CAS  Google Scholar 

  22. Zioncheck TF, Richardson L, Liu J, Chang L, King KL, Bennett GL et al. Sulfated oligosaccharides promote hepatocyte growth factor association and govern its mitogenic activity. J Biol Chem 1995; 270: 16871–16878.

    Article  CAS  Google Scholar 

  23. Flynn DL, Ahn YM, Berger MS, Caldwell T, Hood MM, Kaufman MD et alRebastinib and DCC-2701: targeting of resistance mechanisms in cancer treatment. ACS National Meeting, New Orleans, 8 April, MEDI 209 2013.

  24. Sawada K, Radjabi AR, Shinomiya N, Kistner E, Kenny H, Becker AR et al. c-Met overexpression is a prognostic factor in ovarian cancer and an effective target for inhibition of peritoneal dissemination and invasion. Cancer Res 2007; 67: 1670–1679.

    Article  CAS  Google Scholar 

  25. Kwon Y, Cukierman E, Godwin AK . Differential expressions of adhesive molecules and proteases define mechanisms of ovarian tumor cell matrix penetration/invasion. PLoS One 2011; 6: e18872.

    Article  CAS  Google Scholar 

  26. Furge KA, Zhang YW, Vande Woude GF . Met receptor tyrosine kinase: enhanced signaling through adapter proteins. Oncogene 2000; 19: 5582–5589.

    Article  CAS  Google Scholar 

  27. Pan BS, Chan GK, Chenard M, Chi A, Davis LJ, Deshmukh SV et al. MK-2461, a novel multitargeted kinase inhibitor, preferentially inhibits the activated c-Met receptor. Cancer Res 2010; 70: 1524–1533.

    Article  CAS  Google Scholar 

  28. Ponzetto C, Bardelli A, Zhen Z, Maina F, dalla Zonca P, Giordano S et al. A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family. Cell 1994; 77: 261–271.

    Article  CAS  Google Scholar 

  29. Michieli P, Basilico C, Pennacchietti S, Maffe A, Tamagnone L, Giordano S et al. Mutant Met-mediated transformation is ligand-dependent and can be inhibited by HGF antagonists. Oncogene 1999; 18: 5221–5231.

    Article  CAS  Google Scholar 

  30. Miyata Y, Kanetake H, Kanda S . Presence of phosphorylated hepatocyte growth factor receptor/c-Met is associated with tumor progression and survival in patients with conventional renal cell carcinoma. Clin Cancer Res 2006; 12: 4876–4881.

    Article  CAS  Google Scholar 

  31. Marchand-Adam S, Marchal J, Cohen M, Soler P, Gerard B, Castier Y et al. Defect of hepatocyte growth factor secretion by fibroblasts in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2003; 168: 1156–1161.

    Article  Google Scholar 

  32. Matsuoka H, Sisson TH, Nishiuma T, Simon RH . Plasminogen-mediated activation and release of hepatocyte growth factor from extracellular matrix. Am J Respir Cell Mol Biol 2006; 35: 705–713.

    Article  CAS  Google Scholar 

  33. Lyon M, Deakin JA, Rahmoune H, Fernig DG, Nakamura T, Gallagher JT . Hepatocyte growth factor/scatter factor binds with high affinity to dermatan sulfate. J Biol Chem 1998; 273: 271–278.

    Article  CAS  Google Scholar 

  34. Nayeri F, Nayeri T, Aili D, Brudin L, Liedberg B . Clinical impact of real-time evaluation of the biological activity and degradation of hepatocyte growth factor. Growth Factors 2008; 26: 163–171.

    Article  CAS  Google Scholar 

  35. Hamilton TC, Young RC, McKoy WM, Grotzinger KR, Green JA, Chu EW et al. Characterization of a human ovarian carcinoma cell line (NIH:OVCAR-3) with androgen and estrogen receptors. Cancer Res 1983; 43: 5379–5389.

    CAS  PubMed  Google Scholar 

  36. Langdon SP, Lawrie SS, Hay FG, Hawkes MM, McDonald A, Hayward IP et al. Characterization and properties of nine human ovarian adenocarcinoma cell lines. Cancer Res 1988; 48: 6166–6172.

    CAS  Google Scholar 

  37. Quiros RM, Valianou M, Kwon Y, Brown KM, Godwin AK, Cukierman E . Ovarian normal and tumor-associated fibroblasts retain in vivo stromal characteristics in a 3-D matrix-dependent manner. Gynecol Oncol 2008; 110: 99–109.

    Article  CAS  Google Scholar 

  38. Amatangelo MD, Bassi DE, Klein-Szanto AJ, Cukierman E . Stroma-derived three-dimensional matrices are necessary and sufficient to promote desmoplastic differentiation of normal fibroblasts. Am J Pathol 2005; 167: 475–488.

    Article  CAS  Google Scholar 

  39. Beacham DA, Amatangelo MD, Cukierman E . Preparation of extracellular matrices produced by cultured and primary fibroblasts. Curr Protoc Cell Biol 2007, Chapter 10 Unit 10 9.

  40. Castello-Cros R, Cukierman E . Stromagenesis during tumorigenesis: characterization of tumor-associated fibroblasts and stroma-derived 3D matrices. Methods Mol Biol 2009; 522: 275–305.

    Article  CAS  Google Scholar 

  41. Cukierman E, Pankov R, Stevens DR, Yamada KM . Taking cell-matrix adhesions to the third dimension. Science 2001; 294: 1708–1712.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Dr Ziyan Pessetto for her expertise in the cell viability assays and Dr Daniel Flynn for his helpful comments and guidance in regards the biology and activity of DCC-2701. The study was supported in part by a program project grant from Ovarian Cancer Research Fund (http://www.ocrf.org) and a grant from the NCI (R01 CA140323) to AKG. We would also like to acknowledge support from the University of Kansas Cancer Center’s CCSG (P30 CA168524), the Kansas Bioscience Authority Eminent Scholar Program. AKG is the Chancellors Distinguished Chair in Biomedical Sciences endowed Professor.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to A K Godwin.

Ethics declarations

Competing interests

Dr AKG’s work has been funded by the NIH/NCI, the state of Kansas and non-profit organizations. BSD and MDK are employees of and receive compensation from Deciphera Pharmaceuticals, LLC. The remaining authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on the Oncogene website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kwon, Y., Smith, B., Zhou, Y. et al. Effective inhibition of c-MET-mediated signaling, growth and migration of ovarian cancer cells is influenced by the ovarian tissue microenvironment. Oncogene 34, 144–153 (2015). https://doi.org/10.1038/onc.2013.539

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/onc.2013.539

Keywords

This article is cited by

Search

Quick links