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.

  • Letter
  • Published:

The Hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies

Abstract

Resistance to RAF- and MEK-targeted therapy is a major clinical challenge1,2,3,4. RAF and MEK inhibitors are initially but only transiently effective in some but not all patients with BRAF gene mutation and are largely ineffective in those with RAS gene mutation because of resistance5,6,7,8,9,10,11,12,13,14. Through a genetic screen in BRAF-mutant tumor cells, we show that the Hippo pathway effector YAP (encoded by YAP1) acts as a parallel survival input to promote resistance to RAF and MEK inhibitor therapy. Combined YAP and RAF or MEK inhibition was synthetically lethal not only in several BRAF-mutant tumor types but also in RAS-mutant tumors. Increased YAP in tumors harboring BRAF V600E was a biomarker of worse initial response to RAF and MEK inhibition in patients, establishing the clinical relevance of our findings. Our data identify YAP as a new mechanism of resistance to RAF- and MEK-targeted therapy. The findings unveil the synthetic lethality of combined suppression of YAP and RAF or MEK as a promising strategy to enhance treatment response and patient survival.

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: A pooled shRNA screen in BRAF-mutant human lung cancer cells identifies new modifiers of the RAF inhibitor response including YAP.
Figure 2: YAP regulates the response to RAF and MEK inhibitors in multiple BRAF-mutant tumor types.
Figure 3: Synthetic lethality and synergistic induction of apoptosis with concurrent inhibition of YAP and oncogenic MAPK signaling.
Figure 4: Increased YAP levels in human tumor specimens encoding BRAF V600E is a biomarker of worse response to RAF inhibitor in patients.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Flaherty, K.T. et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N. Engl. J. Med. 367, 1694–1703 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. 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 

  3. Flaherty, K.T. et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N. Engl. J. Med. 367, 107–114 (2012).

    Article  CAS  PubMed  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. Adjei, A.A. et al. Phase I pharmacokinetic and pharmacodynamic study of the oral, small-molecule mitogen-activated protein kinase kinase 1/2 inhibitor AZD6244 (ARRY-142886) in patients with advanced cancers. J. Clin. Oncol. 26, 2139–2146 (2008).

    Article  CAS  PubMed  Google Scholar 

  6. Jänne, P.A. et al. Selumetinib plus docetaxel for KRAS-mutant advanced non-small-cell lung cancer: a randomised, multicentre, placebo-controlled, phase 2 study. Lancet Oncol. 14, 38–47 (2013).

    Article  PubMed  Google Scholar 

  7. Migliardi, G. et al. Inhibition of MEK and PI3K/mTOR suppresses tumor growth but does not cause tumor regression in patient-derived xenografts of RAS-mutant colorectal carcinomas. Clin. Cancer Res. 18, 2515–2525 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Fordyce, S.L. et al. Genetic diversity among pandemic 2009 influenza viruses isolated from a transmission chain. Virol. J. 10, 116 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Roth, A.D. et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J. Clin. Oncol. 28, 466–474 (2010).

    Article  CAS  PubMed  Google Scholar 

  10. Planchard, D. et al. Interim results of phase II study BRF113928 of dabrafenib in BRAF V600E mutation–positive non–small cell lung cancer (NSCLC) patients. J. Clin. Oncol. 31, 2013 (suppl; abstract 8009) (2013).

    Google Scholar 

  11. Lin, L. et al. Mapping the molecular determinants of BRAF oncogene dependence in human lung cancer. Proc. Natl. Acad. Sci. USA 111, E748–E757 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  13. 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 

  14. Montero-Conde, C. et al. Relief of feedback inhibition of HER3 transcription by RAF and MEK inhibitors attenuates their antitumor effects in BRAF-mutant thyroid carcinomas. Cancer Discov. 3, 520–533 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Rudin, C.M., Hong, K. & Streit, M. Molecular characterization of acquired resistance to the BRAF inhibitor dabrafenib in a patient with BRAF-mutant non-small-cell lung cancer. J. Thorac. Oncol. 8, e41–e42 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Downward, J. Targeting RAS signalling pathways in cancer therapy. Nat. Rev. Cancer 3, 11–22 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Young, A. et al. Ras signaling and therapies. Adv. Cancer Res. 102, 1–17 (2009).

    Article  CAS  PubMed  Google Scholar 

  18. Bivona, T.G. et al. FAS and NF-κB signalling modulate dependence of lung cancers on mutant EGFR. Nature 471, 523–526 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hong, W. & Guan, K.L. The YAP and TAZ transcription co-activators: key downstream effectors of the mammalian Hippo pathway. Semin. Cell Dev. Biol. 23, 785–793 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pan, D. The hippo signaling pathway in development and cancer. Dev. Cell 19, 491–505 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Pylayeva-Gupta, Y., Grabocka, E. & Bar-Sagi, D. RAS oncogenes: weaving a tumorigenic web. Nat. Rev. Cancer 11, 761–774 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kapoor, A. et al. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell 158, 185–197 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhang, W. et al. Downstream of mutant KRAS, the transcription regulator YAP is essential for neoplastic progression to pancreatic ductal adenocarcinoma. Sci. Signal. 7, ra42 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Shao, D.D. et al. KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 158, 171–184 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Huang, J., Wu, S., Barrera, J., Matthews, K. & Pan, D. The Hippo signaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Drosophila homolog of YAP. Cell 122, 421–434 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Vigneron, A.M., Ludwig, R.L. & Vousden, K.H. Cytoplasmic ASPP1 inhibits apoptosis through the control of YAP. Genes Dev. 24, 2430–2439 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Rosenbluh, J. et al. β-catenin–driven cancers require a YAP1 transcriptional complex for survival and tumorigenesis. Cell 151, 1457–1473 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Frederick, D.T. et al. Clinical profiling of BCL-2 family members in the setting of BRAF inhibition offers a rationale for targeting de novo resistance using BH3 mimetics. PLoS ONE 9, e101286 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Corcoran, R.B. et al. Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumor regressions in KRAS mutant cancer models. Cancer Cell 23, 121–128 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Tan, N. et al. Bcl-2/Bcl-xL inhibition increases the efficacy of MEK inhibition alone and in combination with PI3 kinase inhibition in lung and pancreatic tumor models. Mol. Cancer Ther. 12, 853–864 (2013).

    Article  PubMed  Google Scholar 

  31. Feng, X. et al. Hippo-independent activation of YAP by the GNAQ uveal melanoma oncogene through a trio-regulated Rho GTPase signaling circuitry. Cancer Cell 25, 831–845 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yu, F.X. et al. Mutant Gq/11 promote uveal melanoma tumorigenesis by activating YAP. Cancer Cell 25, 822–830 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hall, C.A. et al. Hippo pathway effector Yap is an ovarian cancer oncogene. Cancer Res. 70, 8517–8525 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhao, B. et al. Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev. 21, 2747–2761 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Li, B. & Dewey, C.N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank G. Bollag and P. Lin (Plexxikon) for providing PLX4720. We thank J. Weissman and members of the Bivona laboratory for critical review of the manuscript. The authors acknowledge funding support (to T.G.B.) from the following sources: US National Institutes of Health (NIH) Director's New Innovator Award (DP2CA174497), the Howard Hughes Medical Institute (Collaborative Innovation Award), the Doris Duke Charitable Foundation, the American Lung Association, the National Lung Cancer Partnership, the Addario and Van Auken Private Foundations, the Sidney Kimmel Foundation for Cancer Research and the Searle Scholars Program. The authors also acknowledge funding from La Caixa Foundation (to R.R.) and from US NIH grant R01CA131261 (to M.M.).

Author information

Authors and Affiliations

Authors

Contributions

L.L., E.C., A.J.S., V.O., D.N., J.J.Y., E.P., X.L., M.M.W., L.P. and E.A.C. conducted all experiments and/or analyzed data. L.C. and S.A. conducted the analysis of the shRNA screening data. M.M. provided cell lines and analyzed data. N.K., M.G.C., J.L.M., J.L.R., J.M.S.T., F.B., C.M.R., A.A., E.R., I.O., E.C., E.M.-C., J.C., A.M., R.R., D.T.F., Z.A.C., K.T.F. and J.A.W. provided clinical samples and data. All authors contributed to the design of experiments and to data analysis and interpretation. T.G.B., L.L. and A.J.S. wrote the manuscript with input from all coauthors.

Corresponding author

Correspondence to Trever G Bivona.

Ethics declarations

Competing interests

T.G.B. is a consultant for Driver Group as well as Novartis, Clovis Oncology and Cleave Biosciences and is the recipient of a research grant from Servier.

Integrated supplementary information

Supplementary Figure 1 Effects of YAP1 inhibition on cell growth.

(a) Primary screen data showing that the YAP1 gene was not depleted over 10 d of vehicle (DMSO) treatment in HCC364 cells (DMSO versus T0). (b) Primary screening data showing that shRNAs targeting YAP1 were not depleted over 10 d of vehicle (DMSO) treatment in HCC364 cells (DMSO versus T0). (c) Effect of YAP1 knockdown using two independent shRNAs on cell growth in HCC364, A2058, HT29, WiDr, KHM-5M, HTC/C3, A549, H2347, SW1573, MOR/CPR, SK-MEL-2, MM415, HPAF-II and PANC02.03 cells, measured at 3 d by CellTiter-Glo assay.

Supplementary Figure 2 The effects of YAP silencing were specific to targeted inhibition of RAF-MEK signaling and are through YAP transcriptional activity.

(a) Effects of YAP1 knockdown using two independent shRNAs on pemetrexed sensitivity in HCC364 lung cancer cells. (b) Effects of YAP1 knockdown using two independent shRNAs on cisplatin sensitivity in HCC364 lung cancer cells. (c) Effects of TEAD2 knockdown using two independent shRNAs on vemurafenib sensitivity in HCC364 lung cancer cells (shown are the IC50 and relative cell viability). (d) Validation of the effects of TEAD2 knockdown on trametinib sensitivity in HCC364 BRAF-mutant lung cancer cells (shown are the IC50 and cell viability). (e) Effects of TEAD4 knockdown using two independent shRNAs on vemurafenib sensitivity in HCC364 lung cancer cells (shown are the IC50 and relative cell viability). (f) Validation of the effects of TEAD4 knockdown on trametinib sensitivity in HCC364 BRAF-mutant lung cancer cells (shown are the IC50 and cell viability). (g) mRNA expression of TEAD2 and TEAD4 in cells expressing scrambled control shRNA or shRNA to TEAD2 or TEAD4. (h) Effects of TEAD2 and TEAD4 knockdown on vemurafenib and trametinib sensitivity in HCC364 BRAF-mutant lung cancer cells (shown is 7-d cell growth assessed by crystal violet staining assays, with quantification of the effects on viability under each condition).

Supplementary Figure 3 Nuclear YAP expression in BRAF- and RAS-mutant cancer cell lines.

Nuclear/cytoplasmic fractionation and immunoblot analysis of the indicated proteins in (a) BRAF V600E–mutant cancer cell lines and (b) RAS-mutant cancer cell lines. Data represent three independent experiments.

Supplementary Figure 4 Exogenous expression of YAP or TAZ promotes resistance to RAF-MEK inhibition.

(a) Effects of YAP1 or TAZ overexpression on vemurafenib sensitivity in HCC364 lung cancer cells (shown are the IC50 and relative cell viability). (b) Effects of YAP1 or TAZ overexpression on trametinib sensitivity in HCC364 lung cancer cells (shown are the IC50 and relative cell viability).

Supplementary Figure 5 Increase in maximal growth inhibition upon trametinib treatment in YAP1-depleted cells.

(a) Effects of YAP1 knockdown on trametinib sensitivity in the indicated BRAF-mutant cell lines, shown as percentage of maximal growth inhibition (n = 4, +s.e.m. for all cell viability data shown). (b) Effects of YAP1 knockdown on trametinib sensitivity in the indicated RAS-mutant cell lines, shown as percentage of maximal growth inhibition (n = 4, +s.e.m. for all cell viability data shown).

Supplementary Figure 6 YAP1 knockdown sensitizes cancer cells to RAF-MEK inhibition across multiple tumor types.

(a,b) Effects of YAP1 knockdown using two independent shRNAs on vemurafenib and trametinib sensitivity in (a) A2058 and (b) WM793 BRAF V600E–mutant melanoma cancer cells. (c,d) Effects of YAP1 knockdown on vemurafenib and trametinib sensitivity in (c) A2058 and (d) WM793 BRAF V600E–mutant melanoma cancer cells (shown is 7-d cell growth assessed by crystal violet staining assays, with quantification of the effects on viability under each condition). (e,f) Effects of YAP1 knockdown using two independent shRNAs on vemurafenib and trametinib sensitivity in (e) HT29 and (f) WiDr BRAF V600E–mutant colon cancer cells. (g,h) Effects of YAP1 knockdown using two independent shRNAs on vemurafenib and trametinib sensitivity in (g) KHM-5M and (h) HTC/C3 BRAF V600E–mutant thyroid cancer cells.

Supplementary Figure 7 YAP1 inhibition sensitizes tumors to RAF-MEK inhibition in vivo.

(a) Average tumor volume of xenografts generated from A2058 cells infected with either scrambled control shRNA or YAP1 shRNA (n = 10/group, +s.e.m.). (b) Waterfall plot indicating fold change in tumor volume upon vemurafenib or trametinib treatment of A2058 xenografts infected with either scrambled control shRNA or YAP1 shRNA (n = 8–12/group, ±s.e.m.). (c) Immunoblot analysis for each indicated protein in lysates from representative A2058 xenograft tumors. d, Average tumor volume of xenografts generated from HT29 cells infected with either scrambled control shRNA or YAP1 shRNA (n = 10/group, +s.e.m.). (e) Waterfall plot indicating fold change in tumor volume upon vemurafenib or trametinib treatment of HT29 xenograft model infected with either scrambled control shRNA or YAP1 shRNA (n = 8–12/group, ±s.e.m.). (f) Immunoblot analysis for each indicated protein in lysates from representative HT29 xenograft tumors. (g) Waterfall plot indicating fold change in tumor volume upon trametinib treatment of MOR/CPR xenograft model infected with either scrambled control shRNA or YAP1 shRNA (n = 8–12/group, ±s.e.m.). (h) Immunoblot analysis for each indicated protein in lysates from representative MOR/CPR xenograft tumors.

Supplementary Figure 8 Effect of treatment with the EGFR kinase inhibitor erlotinib on RAF-MEK inhibitor sensitivity.

(a,b) Effect of treatment with the EGFR kinase inhibitor erlotinib on (a) vemurafenib and (b) trametinib sensitivity in A2058 BRAF V600E–mutant melanoma cells. (c,d) Effect of EGFR kinase inhibitor erlotinib on (c) vemurafenib and (d) trametinib sensitivity in KHM-5M BRAF V600E–mutant thyroid cancer cells.

Supplementary Figure 9 YAP1 knockdown sensitizes RAS-mutant melanoma and pancreatic cancer cells to MEK inhibition.

(a,b) Effects of YAP1 knockdown using two independent shRNAs on trametinib sensitivity in (a) MM415 NRAS Q61L–mutant, (b) SK-MEL-2 NRAS Q61R–mutant melanoma cells. (c,d) Effects of YAP1 knockdown using two independent shRNAs on trametinib sensitivity in (c) HPAF-II KRAS G12D–mutant and (d) Panc 02.03 KRAS G12D–mutant pancreatic cancer cells. (e,f) Effects of YAP1 knockdown using two independent shRNAs on trametinib sensitivity in (e) MM415 NRAS Q61L–mutant and (f) SK-MEL-2 NRAS Q61R–mutant melanoma cells (shown is 7-d cell growth assessed by crystal violet staining assays, with quantification of the effects on viability under each condition).

Supplementary Figure 10 YAP1 knockdown sensitizes RAS-mutant NSCLC cells to MEK inhibition.

(af) Effects of YAP1 knockdown using two independent shRNAs on trametinib sensitivity in (a) A549 KRAS G12S–mutant, (b) H23 KRAS G12C–mutant, (c) Calu-6 KRAS G12C–mutant, (d) H2347 NRAS Q61R–mutant, (e) MOR/CPR KRAS G12C–mutant and (f) SW1573 KRAS G12C–mutant NSCLC cell lines. (gi) Effects of YAP1 knockdown using two independent shRNAs on trametinib sensitivity in (g) MOR/CPR KRAS G12C–mutant cells, (h) SW1573 KRAS G12C–mutant cells and (i) H2347 NRAS Q61R–mutant cells (shown is 7-d cell growth assessed by crystal violet staining assays, with quantification of the effects on viability under each condition).

Supplementary Figure 11 YAP1 knockdown induces apoptosis upon RAF-MEK inhibition.

(a,b) Effects of YAP1 knockdown on apoptosis induced upon (a) vemurafenib and (b) trametinib treatment in WiDr BRAF-mutant colon cancer cells as measured by caspase-3/7 activation. (c,d) Effects of YAP1 knockdown on apoptosis induced upon (c) vemurafenib and (d) trametinib treatment in A549 KRAS-mutant lung cancer cells as measured by caspase-3/7 activation. (e,f) Effects of pharmacological inhibition of BCL-xL using TW-37 on vemurafenib or trametinib sensitivity in (e) HCC364 BRAF-mutant lung cancer cells and (f) A2058 BRAF-mutant melanoma cells (n = 3, ±s.e.m. for all cell viability data shown; P values are indicated for statistical analysis). (g,h) Effects of pharmacological inhibition of BCL-xL using ABT-263 or TW-37 on trametinib sensitivity in (g) A549 KRAS G12S–mutant and (h) Calu-1 KRAS G12C–mutant lung cancer cells (n = 3, ±s.e.m. for all cell viability data shown; P values are indicated for statistical analysis).

Supplementary Figure 12 YAP inhibition plus RAF-MEK inhibition suppresses BCL-xL expression.

(ac) Effects of YAP suppression and vemurafenib or trametinib treatment on BCL-xL levels by immunoblot analysis in BRAF V600E–mutant (a) A2058 melanoma, (b) HT29 colon and (c) KHM-5M thyroid cancer cells. Results represent three independent experiments.

Supplementary Figure 13 BCL-xL expression rescues YAP1-depleted cells from growth suppression by RAF-MEK inhibition.

(a,b) Effects of BCL-xL overexpression and YAP1 knockdown on (a) vemurafenib and (b) trametinib sensitivity in HCC364 lung cancer cells (shown are the IC50 and relative cell viability). (c) Immunoblot analysis for each indicated protein in lysates from HCC364 cells overexpressing either GFP or BCL-xL and treated with either control siRNA or YAP1 siRNA.

Supplementary Figure 14 Gene sets enriched among the genes specifically altered by YAP and MEK coinhibition.

Pathway analysis indicating enriched gene sets among those genes whose expression was specifically decreased by YAP and MEK coinhibition (Supplementary Table 5). The top six significantly represented pathways among these genes are shown (significance of pathway representation is shown as –log q, with the red line indicating q < 0.05). The genes highlighted in the inset are the significantly altered apoptosis-related genes.

Supplementary Figure 15 YAP protein expression in human tumor specimens by YAP immunohistochemistry.

(a) Representative images of immunohistochemistry staining of YAP protein in representative human tumor specimens with low, intermediate and high YAP staining (brown). (b) YAP staining by immunohistochemistry in BRAF V600E–mutant melanomas obtained before RAF or MEK inhibitor treatment and upon acquired resistance. Shown are the 4 out of 16 cases in which the pretreatment tumor did not harbor high levels of YAP (showing either low or intermediate staining by immunohistochemistry). Bars represent 50 microns.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 and Supplementary Tables 2–4, 6 and 7 (PDF 2308 kb)

Supplementary Table 1

Gene targets included in the pooled shRNA screening library. (XLSX 101 kb)

Supplementary Table 5

Differentially expressed genes in BRAFV600E NSCLC cells (HCC364) upon either YAP knockdown alone (by shRNA), trametinib treatment alone and combined YAP knockdown and trametinib treatment. (XLSX 398 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, L., Sabnis, A., Chan, E. et al. The Hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies. Nat Genet 47, 250–256 (2015). https://doi.org/10.1038/ng.3218

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3218

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing