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:

Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers

An Erratum to this article was published on 02 October 2013

Abstract

KRAS and BRAF activating mutations drive tumorigenesis through constitutive activation of the MAPK pathway. As these tumours represent an area of high unmet medical need, multiple allosteric MEK inhibitors, which inhibit MAPK signalling in both genotypes, are being tested in clinical trials. Impressive single-agent activity in BRAF-mutant melanoma has been observed; however, efficacy has been far less robust in KRAS-mutant disease1. Here we show that, owing to distinct mechanisms regulating MEK activation in KRAS- versus BRAF-driven tumours2,3, different mechanisms of inhibition are required for optimal antitumour activity in each genotype. Structural and functional analysis illustrates that MEK inhibitors with superior efficacy in KRAS-driven tumours (GDC-0623 and G-573, the former currently in phase I clinical trials) form a strong hydrogen-bond interaction with S212 in MEK that is critical for blocking MEK feedback phosphorylation by wild-type RAF. Conversely, potent inhibition of active, phosphorylated MEK is required for strong inhibition of the MAPK pathway in BRAF-mutant tumours, resulting in superior efficacy in this genotype with GDC-0973 (also known as cobimetinib), a MEK inhibitor currently in phase III clinical trials. Our study highlights that differences in the activation state of MEK in KRAS-mutant tumours versus BRAF-mutant tumours can be exploited through the design of inhibitors that uniquely target these distinct activation states of MEK. These inhibitors are currently being evaluated in clinical trials to determine whether improvements in therapeutic index within KRAS versus BRAF preclinical models translate to improved clinical responses in patients.

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: Allosteric MEK inhibitors show distinct relative potencies in KRAS-mutant and BRAF-mutant cancer models.
Figure 2: MEK inhibitor efficacy in KRAS-mutant models correlates with the ability to block feedback-induced phosphorylation of MEK by RAF.
Figure 3: Distinct inhibitor interactions with MEK determine their individual effects on RAF–MEK complex formation, MEK activation and inhibition of ERK phosphorylation.
Figure 4: Potency of MEK inhibitors in BRAF-mutant versus KRAS-mutant tumours correlates with high inhibitor binding affinity against dually phosphorylated MEK (S218/S222) compared to un-phosphorylated MEK.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

Data deposits

The structure of the MEK1:GDC-0973 complex has been deposited with the PDB and assigned accession code 4LMN.

References

  1. Akinleye, A., Furqan, M., Mukhi, N., Ravella, P. & Liu, D. MEK and the inhibitors: from bench to bedside. J. Hematol. Oncol. 6, 27 (2013)

    Article  CAS  Google Scholar 

  2. Blasco, R. B. et al. c-Raf, but not B-Raf, is essential for development of K-Ras oncogene-driven non-small cell lung carcinoma. Cancer Cell 19, 652–663 (2011)

    Article  CAS  Google Scholar 

  3. Davies, H. et al. Mutations of the BRAF gene in human cancer. Nature 417, 949–954 (2002)

    Article  ADS  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  5. Bamford, S. et al. The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website. Br. J. Cancer 91, 355–358 (2004)

    Article  CAS  Google Scholar 

  6. Hatzivassiliou, G. et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature 464, 431–435 (2010)

    Article  ADS  CAS  Google Scholar 

  7. Heidorn, S. J. et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell 140, 209–221 (2010)

    Article  CAS  Google Scholar 

  8. Poulikakos, P. I., Zhang, C., Bollag, G., Shokat, K. M. & Rosen, N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature 464, 427–430 (2010)

    Article  ADS  CAS  Google Scholar 

  9. Solit, D. B. et al. BRAF mutation predicts sensitivity to MEK inhibition. Nature 439, 358–362 (2006)

    Article  ADS  CAS  Google Scholar 

  10. Wellbrock, C., Karasarides, M. & Marais, R. The RAF proteins take centre stage. Nature Rev. Mol. Cell Biol. 5, 875–885 (2004)

    Article  CAS  Google Scholar 

  11. Chapman, M. S. & Miner, J. N. Novel mitogen-activated protein kinase kinase inhibitors. Expert Opin. Investig. Drugs 20, 209–220 (2011)

    Article  CAS  Google Scholar 

  12. Engelman, J. A. et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nature Med. 14, 1351–1356 (2008)

    Article  CAS  Google Scholar 

  13. Gilmartin, A. G. et al. GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition. Clin. Cancer Res. 17, 989–1000 (2011)

    Article  CAS  Google Scholar 

  14. Hoeflich, K. P. et al. Intermittent administration of MEK inhibitor GDC-0973 plus PI3K inhibitor GDC-0941 triggers robust apoptosis and tumor growth inhibition. Cancer Res. 72, 210–219 (2012)

    Article  CAS  Google Scholar 

  15. Friday, B. B. et al. BRAF V600E disrupts AZD6244-induced abrogation of negative feedback pathways between extracellular signal-regulated kinase and Raf proteins. Cancer Res. 68, 6145–6153 (2008)

    Article  CAS  Google Scholar 

  16. Pratilas, C. A. et al. V600EBRAF is associated with disabled feedback inhibition of RAF–MEK signaling and elevated transcriptional output of the pathway. Proc. Natl Acad. Sci. USA 106, 4519–4524 (2009)

    Article  ADS  CAS  Google Scholar 

  17. Delaney, A. M., Printen, J. A., Chen, H., Fauman, E. B. & Dudley, D. T. Identification of a novel mitogen-activated protein kinase kinase activation domain recognized by the inhibitor PD 184352. Mol. Cell. Biol. 22, 7593–7602 (2002)

    Article  CAS  Google Scholar 

  18. Heald, R. A. et al. Discovery of novel allosteric mitogen-activated protein kinase kinase (MEK) 1,2 inhibitors possessing bidentate Ser212 interactions. J. Med. Chem. 55, 4594–4604 (2012)

    Article  CAS  Google Scholar 

  19. O'Hagan, D. Understanding organofluorine chemistry. An introduction to the C-F bond. Chem. Soc. Rev. 37, 308–319 (2008)

    Article  CAS  Google Scholar 

  20. Gopalbhai, K. et al. Negative regulation of MAPKK by phosphorylation of a conserved serine residue equivalent to Ser212 of MEK1. J. Biol. Chem. 278, 8118–8125 (2003)

    Article  CAS  Google Scholar 

  21. Sheth, P. R. et al. Fully activated MEK1 exhibits compromised affinity for binding of allosteric inhibitors U0126 and PD0325901. Biochemistry 50, 7964–7976 (2011)

    Article  CAS  Google Scholar 

  22. Resing, K. A. & Ahn, N. G. Deuterium exchange mass spectrometry as a probe of protein kinase activation. Analysis of wild-type and constitutively active mutants of MAP kinase kinase-1. Biochemistry 37, 463–475 (1998)

    Article  CAS  Google Scholar 

  23. Rice, K. D. et al. Novel carboxamide-based allosteric MEK inhibitors: discovery and optimization efforts toward XL518 (GDC-0973). ACS Med. Chem. Lett. 3, 416–421 (2012)

    Article  CAS  Google Scholar 

  24. Alessi, D. R., Cuenda, A., Cohen, P., Dudley, D. T. & Saltiel, A. R. PD 098059 is a specific inhibitor of the activation of mitogen-activated protein kinase kinase in vitro and in vivo. J. Biol. Chem. 270, 27489–27494 (1995)

    Article  CAS  Google Scholar 

  25. Ohren, J. F. et al. Structures of human MAP kinase kinase 1 (MEK1) and MEK2 describe novel noncompetitive kinase inhibition. Nature Struct. Mol. Biol. 11, 1192–1197 (2004)

    Article  CAS  Google Scholar 

  26. Garnett, M. J., Rana, S., Paterson, H., Barford, D. & Marais, R. Wild-type and mutant B-RAF activate C-RAF through distinct mechanisms involving heterodimerization. Mol. Cell 20, 963–969 (2005)

    Article  CAS  Google Scholar 

  27. Röring, M. et al. Distinct requirement for an intact dimer interface in wild-type, V600E and kinase-dead B-Raf signalling. EMBO J. 31, 2629–2647 (2012)

    Article  Google Scholar 

  28. Rosen, L. et al. A first-in-human phase 1 study to evaluate the MEK1/2 inhibitor GDC-0973 administered daily in patients with advanced solid tumors. Cancer Res. 71, 4716 (2011)

    Google Scholar 

  29. Choo, E. F. et al. Preclinical disposition and pharmacokinetics-pharmacodynamic modeling of biomarker response and tumour growth inhibition in xenograft mouse models of G-573, a MEK inhibitor. Xenobiotica 40, 751–762 (2010)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge the Genentech IVCC group, IVP dosing group, as well as B. Liu and I. Yen and for technical support. SSRL is supported by the Department of Energy, the National Institutes of Health, the National Institute of General Medical Sciences and the National Center for Research Resources.

Author information

Authors and Affiliations

Authors

Contributions

J.R.H., K.S., S-M.L., J.C., A.P., C.O. J.F.M.H., D.J.A., M.J.C.L., M.Z. and M.U. performed the experiments; M.M., S.P., R.H., C.W., M.Z. and K.P.H. analysed data; L.S.F. provided comments; G.H., J.R.H., H.C., S.M. and M.B. directed the studies, analysed data, and wrote the manuscript.

Corresponding authors

Correspondence to Georgia Hatzivassiliou or Marcia Belvin.

Ethics declarations

Competing interests

The authors were all employees of Genentech or Argenta at the time that the work was carried out.

Supplementary information

Supplementary Information

This file contains Supplementary Methods, Supplementary Figures 1-13 and Supplementary Tables 1-3. This file was replaced on September 11, as Supplementary Figure 7 was corrupted and Supplementary Figures 12 and 13 were missing from the original file posted on line. (PDF 10886 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hatzivassiliou, G., Haling, J., Chen, H. et al. Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers. Nature 501, 232–236 (2013). https://doi.org/10.1038/nature12441

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12441

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: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research