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.

Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion

Abstract

Drug resistance presents a challenge to the treatment of cancer patients. Many studies have focused on cell-autonomous mechanisms of drug resistance. By contrast, we proposed that the tumour micro-environment confers innate resistance to therapy. Here we developed a co-culture system to systematically assay the ability of 23 stromal cell types to influence the innate resistance of 45 cancer cell lines to 35 anticancer drugs. We found that stroma-mediated resistance is common, particularly to targeted agents. We characterized further the stroma-mediated resistance of BRAF-mutant melanoma to RAF inhibitors because most patients with this type of cancer show some degree of innate resistance1,2,3,4. Proteomic analysis showed that stromal cell secretion of hepatocyte growth factor (HGF) resulted in activation of the HGF receptor MET, reactivation of the mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-OH kinase (PI(3)K)–AKT signalling pathways, and immediate resistance to RAF inhibition. Immunohistochemistry experiments confirmed stromal cell expression of HGF in patients with BRAF-mutant melanoma and showed a significant correlation between HGF expression by stromal cells and innate resistance to RAF inhibitor treatment. Dual inhibition of RAF and either HGF or MET resulted in reversal of drug resistance, suggesting RAF plus HGF or MET inhibitory combination therapy as a potential therapeutic strategy for BRAF-mutant melanoma. A similar resistance mechanism was uncovered in a subset of BRAF-mutant colorectal and glioblastoma cell lines. More generally, this study indicates that the systematic dissection of interactions between tumours and their micro-environment can uncover important mechanisms underlying drug resistance.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: The effect of stromal cells on the chemoresistance of cancer cell lines.
Figure 2: HGF rescues melanoma cancer cell lines from RAF and MEK inhibitors.
Figure 3: HGF is present in the stromal cells of melanoma and correlates with a poor response to therapy.
Figure 4: Characterizing the molecular mechanism of HGF-induced primary resistance.

Similar content being viewed by others

References

  1. Bollag, G. et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature 467, 596–599 (2010)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  2. Chapman, P. B. et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N. Engl. J. Med. 364, 2507–2516 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Flaherty, K. T. et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N. Engl. J. Med. 363, 809–819 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sosman, J. A. et al. Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N. Engl. J. Med. 366, 707–714 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Joyce, J. A. & Pollard, J. W. Microenvironmental regulation of metastasis. Nature Rev. Cancer 9, 239–252 (2009)

    Article  CAS  Google Scholar 

  6. Kalluri, R. & Zeisberg, M. Fibroblasts in cancer. Nature Rev. Cancer 6, 392–401 (2006)

    Article  CAS  Google Scholar 

  7. McMillin, D. W. et al. Tumor cell-specific bioluminescence platform to identify stroma-induced changes to anticancer drug activity. Nature Med. 16, 483–489 (2010)

    Article  CAS  PubMed  Google Scholar 

  8. Shekhar, M. P., Santner, S., Carolin, K. A. & Tait, L. Direct involvement of breast tumor fibroblasts in the modulation of tamoxifen sensitivity. Am. J. Pathol. 170, 1546–1560 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Teicher, B. A. et al. Tumor resistance to alkylating agents conferred by mechanisms operative only in vivo. Science 247, 1457–1461 (1990)

    Article  CAS  ADS  PubMed  Google Scholar 

  10. Wang, W. et al. Crosstalk to stromal fibroblasts induces resistance of lung cancer to epidermal growth factor receptor tyrosine kinase inhibitors. Clin. Cancer Res. 15, 6630–6638 (2009)

    Article  CAS  PubMed  Google Scholar 

  11. Williams, R. T., Roussel, M. F. & Sherr, C. J. Arf gene loss enhances oncogenicity and limits imatinib response in mouse models of Bcr–Abl-induced acute lymphoblastic leukemia. Proc. Natl Acad. Sci. USA 103, 6688–6693 (2006)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  12. Puri, N. et al. c-Met is a potentially new therapeutic target for treatment of human melanoma. Clin. Cancer Res. 13, 2246–2253 (2007)

    Article  CAS  PubMed  Google Scholar 

  13. Lee, Y. J. et al. Expression of the c-Met proteins in malignant skin cancers. Ann. Dermatol. 23, 33–38 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Engelman, J. A. et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316, 1039–1043 (2007)

    Article  CAS  ADS  PubMed  Google Scholar 

  15. Wilson, T. R. et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Naturehttp://dx.doi.org/10.1038/nature11249 (2012)

  16. Jiang, C. C. et al. MEK-independent survival of B-RAFV600E melanoma cells selected for resistance to apoptosis induced by the RAF inhibitor PLX4720. Clin. Cancer Res. 17, 721–730 (2011)

    Article  CAS  PubMed  Google Scholar 

  17. Shao, Y. & Aplin, A. E. Akt3-mediated resistance to apoptosis in B-RAF-targeted melanoma cells. Cancer Res. 70, 6670–6681 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Villanueva, J. et al. Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell 18, 683–695 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Johannessen, C. M. et al. COT drives resistance to RAF inhibition through MAP kinase pathway reactivation. Nature 468, 968–972 (2010)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  20. Nazarian, R. et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 468, 973–977 (2010)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  21. Poulikakos, P. I. et al. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E). Nature 480, 387–390 (2011)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  22. Wang, H. et al. Identification of the MEK1(F129L) activating mutation as a potential mechanism of acquired resistance to MEK inhibition in human cancers carrying the B-RafV600E mutation. Cancer Res. 71, 5535–5545 (2011)

    Article  CAS  PubMed  Google Scholar 

  23. Corcoran, R. B. et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2, 227–235 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Prahallad, A. et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 483, 100–103 (2012)

    Article  CAS  ADS  PubMed  Google Scholar 

  25. Kammula, U. S. et al. Molecular co-expression of the c-Met oncogene and hepatocyte growth factor in primary colon cancer predicts tumor stage and clinical outcome. Cancer Lett. 248, 219–228 (2007)

    Article  CAS  PubMed  Google Scholar 

  26. Di Renzo, M. F. et al. Overexpression and amplification of the met/HGF receptor gene during the progression of colorectal cancer. Clin. Cancer Res. 1, 147–154 (1995)

    CAS  PubMed  Google Scholar 

  27. Liu, C., Park, M. & Tsao, M. S. Overexpression of c-met proto-oncogene but not epidermal growth factor receptor or c-erbB-2 in primary human colorectal carcinomas. Oncogene 7, 181–185 (1992)

    CAS  PubMed  Google Scholar 

  28. Chan, A. T., Ogino, S. & Fuchs, C. S. Aspirin and the risk of colorectal cancer in relation to the expression of COX-2. N. Engl. J. Med. 356, 2131–2142 (2007)

    Article  CAS  PubMed  Google Scholar 

  29. Du, J. et al. Bead-based profiling of tyrosine kinase phosphorylation identifies SRC as a potential target for glioblastoma therapy. Nature Biotechnol. 27, 77–83 (2009)

    Article  CAS  Google Scholar 

  30. Keith, C. T., Borisy, A. A. & Stockwell, B. R. Multicomponent therapeutics for networked systems. Nature Rev. Drug Discov. 4, 71–78 (2005)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank all members of the Golub laboratory for discussions. We thank C. M. Johannessen, I. Rabinowitch, N. Shoresh and A. Goren for critical reading of the manuscript, and L. Gaffney for expert assistance with the graphics. This work was supported by the Howard Hughes Medical Institute, National Cancer Institute grants P50CA093683 and U54CA112962 (T.R.G.) and a Melanoma Research Alliance Team Science Award.

Author information

Authors and Affiliations

Authors

Contributions

R.S. and T.R.G. conceived and designed the experiments. R.S. performed the primary cancer–stroma–drugs screen with help from K.S., M.B.-R. and A.D. R.S. carried out the protein array experiments. R.S., K.S., M.B.-R., A.D. and M.M.M. carried out the secondary screens, western blot analysis and enzyme-linked immunosorbent assays (ELISAs). J.D. performed tyrosine kinase phosphorylation profiling. Clinical samples and clinical data were collected by J.A.W., K.T.F., D.T.F., P.B.C., D.B.S., A.R. and R.S.L. The immunohistochemistry experiments were carried out and analysed by S.O., T.M. and Z.R.Q. The immunofluorescence experiments were carried out by J.A.W. and Z.A.C. R.S. and T.R.G. produced the text and figures, including the Supplementary Information. K.S. helped to produce some of the text and figures. All authors discussed the results and contributed to the final manuscript.

Corresponding author

Correspondence to Todd R. Golub.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Figures

This file contains Supplementary Figures 1-24. (PDF 3456 kb)

Supplementary Tables

This zipped file contains Supplementary Tables 1-7 (ZIP 1353 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Straussman, R., Morikawa, T., Shee, K. et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 487, 500–504 (2012). https://doi.org/10.1038/nature11183

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature11183

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer