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.

  • Article
  • Published:

An approach to suppress the evolution of resistance in BRAFV600E-mutant cancer

Abstract

The principles that govern the evolution of tumors exposed to targeted therapy are poorly understood. Here we modeled the selection and propagation of an amplification in the BRAF oncogene (BRAFamp) in patient-derived tumor xenografts (PDXs) that were treated with a direct inhibitor of the kinase ERK, either alone or in combination with other ERK signaling inhibitors. Single-cell sequencing and multiplex fluorescence in situ hybridization analyses mapped the emergence of extra-chromosomal amplification in parallel evolutionary trajectories that arose in the same tumor shortly after treatment. The evolutionary selection of BRAFamp was determined by the fitness threshold, the barrier that subclonal populations need to overcome to regain fitness in the presence of therapy. This differed for inhibitors of ERK signaling, suggesting that sequential monotherapy is ineffective and selects for a progressively higher BRAF copy number. Concurrent targeting of the RAF, MEK and ERK kinases, however, imposed a sufficiently high fitness threshold to prevent the propagation of subclones with high-level BRAFamp. When administered on an intermittent schedule, this treatment inhibited tumor growth in 11/11 PDXs of lung cancer or melanoma without apparent toxicity in mice. Thus, gene amplification can be acquired and expanded through parallel evolution, enabling tumors to adapt while maintaining their intratumoral heterogeneity. Treatments that impose the highest fitness threshold will likely prevent the evolution of resistance-causing alterations and, thus, merit testing 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: ERK-inhibitor-resistant populations with extrachromosomal BRAF amplification.
Figure 2: BRAFamp emerges in parallel evolutionary tracts.
Figure 3: BRAFamp is sufficient to confer a selective advantage in the presence of ERKi treatment.
Figure 4: Fitness threshold model.
Figure 5: Identification of a treatment to suppress the evolution of BRAF-amplified clones.
Figure 6: An intermittent combination treatment inhibits tumor growth in BRAFV600E PDX models for lung cancer and melanoma.

Similar content being viewed by others

References

  1. Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature 511, 543–550 (2014).

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

    Article  CAS  PubMed  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. Planchard, D. et al. Dabrafenib in patients with BRAFV600E-positive advanced non-small-cell lung cancer: a single-arm, multicenter, open-label, phase 2 trial. Lancet Oncol. 17, 642–650 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Flaherty, K.T. et al. Combined BRAF and MEK inhibition in melanoma with BRAFV600 mutations. N. Engl. J. Med. 367, 1694–1703 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Larkin, J. et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N. Engl. J. Med. 371, 1867–1876 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Long, G.V. et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N. Engl. J. Med. 371, 1877–1888 (2014).

    Article  CAS  PubMed  Google Scholar 

  8. Planchard, D. et al. Dabrafenib plus trametinib in patients with previously treated BRAFV600E-mutant metastatic non-small-cell lung cancer: an open-label, multicenter phase 2 trial. Lancet Oncol. 17, 984–993 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Poulikakos, P.I. et al. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAFV600E. Nature 480, 387–390 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Shi, H. et al. Melanoma whole-exome sequencing identifies V600EB-RAF amplification-mediated acquired B-RAF inhibitor resistance. Nat. Commun. 3, 724 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Das Thakur, M. et al. Modeling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance. Nature 494, 251–255 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Shi, H. et al. Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy. Cancer Discov. 4, 80–93 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Lito, P., Rosen, N. & Solit, D.B. Tumor adaptation and resistance to RAF inhibitors. Nat. Med. 19, 1401–1409 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. 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  PubMed  PubMed Central  Google Scholar 

  15. Lito, P. et al. Relief of profound feedback inhibition of mitogenic signaling by RAF inhibitors attenuates their activity in BRAFV600E melanomas. Cancer Cell 22, 668–682 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lipinski, K.A. et al. Cancer evolution and the limits of predictability in precision cancer medicine. Trends Cancer 2, 49–63 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  17. McGranahan, N. & Swanton, C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell 27, 15–26 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Merlo, L.M., Pepper, J.W., Reid, B.J. & Maley, C.C. Cancer as an evolutionary and ecological process. Nat. Rev. Cancer 6, 924–935 (2006).

    Article  CAS  PubMed  Google Scholar 

  19. Baslan, T. et al. Genome-wide copy number analysis of single cells. Nat. Protoc. 7, 1024–1041 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Garvin, T. et al. Interactive analysis and assessment of single-cell copy number variations. Nat. Methods 12, 1058–1060 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Navin, N. et al. Tumor evolution inferred by single-cell sequencing. Nature 472, 90–94 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Morris, E.J. et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov. 3, 742–750 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Wong, D.J. et al. Antitumor activity of the ERK inhibitor SCH772984 against BRAF-mutant, NRAS-mutant and wild-type melanoma. Mol. Cancer 13, 194 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Baslan, T. et al. Optimizing sparse sequencing of single cells for highly multiplex copy number profiling. Genome Res. 25, 714–724 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Weir, B.A. et al. Characterizing the cancer genome in lung adenocarcinoma. Nature 450, 893–898 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Yao, Z. et al. BRAF mutants evade ERK-dependent feedback by different mechanisms that determine their sensitivity to pharmacologic inhibition. Cancer Cell 28, 370–383 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Joseph, E.W. et al. The RAF inhibitor PLX4032 inhibits ERK signaling and tumor cell proliferation in a V600E BRAF–selective manner. Proc. Natl. Acad. Sci. USA 107, 14903–14908 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Kim, K.B. et al. Phase 2 study of the MEK1 and MEK2 inhibitor trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitor. J. Clin. Oncol. 31, 482–489 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. 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 

  30. Prahallad, A. et al. Unresponsiveness of colon cancer to BRAFV600E inhibition through feedback activation of EGFR. Nature 483, 100–103 (2012).

    Article  CAS  PubMed  Google Scholar 

  31. Shi, H. et al. A novel AKT1 mutant amplifies an adaptive melanoma response to BRAF inhibition. Cancer Discov. 4, 69–79 (2014).

    Article  CAS  PubMed  Google Scholar 

  32. Sun, C. et al. Reversible and adaptive resistance to BRAFV600E inhibition in melanoma. Nature 508, 118–122 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Maertens, O. et al. Elucidating distinct roles for NF1 in melanomagenesis. Cancer Discov. 3, 338–349 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Johannessen, C.M. et al. A melanocyte lineage program confers resistance to MAP kinase pathway inhibition. Nature 504, 138–142 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Anderson, K. et al. Genetic variegation of clonal architecture and propagating cells in leukemia. Nature 469, 356–361 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Melchor, L. et al. Single-cell genetic analysis reveals the composition of initiating clones and phylogenetic patterns of branching and parallel evolution in myeloma. Leukemia 28, 1705–1715 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Campbell, P.J. et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467, 1109–1113 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Nguyen, A. et al. PKLR promotes colorectal cancer liver colonization through induction of glutathione synthesis. J. Clin. Invest. 126, 681–694 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Poirier, J.T. et al. DNA methylation in small-cell lung cancer defines distinct disease subtypes and correlates with high expression of EZH2. Oncogene 34, 5869–5878 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Einarsdottir, B.O. et al. Melanoma-patient-derived xenografts accurately model the disease and develop fast enough to guide treatment decisions. Oncotarget 5, 9609–9618 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Cheng, D.T. et al. Memorial Sloan Kettering–Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): a hybridization-capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J. Mol. Diagn. 17, 251–264 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Li, H. & Durbin, R. Fast and accurate short-read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Gao, R. et al. Punctuated copy number evolution and clonal stasis in triple-negative breast cancer. Nat. Genet. 48, 1119–1130 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wanjala, J. et al. Identifying actionable targets through integrative analyses of GEM model and human prostate cancer genomic profiling. Mol. Cancer Ther. 14, 278–288 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Hembrough, T. et al. Application of selected reaction monitoring for multiplex quantification of clinically validated biomarkers in formalin-fixed, paraffin-embedded tumor tissue. J. Mol. Diagn. 15, 454–465 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Catenacci, D.V. et al. Absolute quantitation of Met using mass spectrometry for clinical application: assay precision, stability and correlation with MET gene amplification in FFPE tumor tissue. PLoS One 9, e100586 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lito, P., Solomon, M., Li, L.S., Hansen, R. & Rosen, N. Allele-specific inhibitors inactivate mutant KRASG12C by a trapping mechanism. Science 351, 604–608 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank D. Solit, N. Bouvier and the Center for Molecular Oncology at MSKCC for assistance with next-generation sequencing, Z. Yao (MSKCC) for providing the A375 doxycycline-inducible BRAFV600E cells, as well as C. Sawyers, C. Rudin, J. Poirier and M. Mroczkowski for reviewing the manuscript. This work was supported by the US National Institutes of Health (NIH) (grant K08 CA191082-01A1; P.L.), the Uniting Against Lung Cancer Foundation (P.L.), the Damon Runyon Clinical Investigator Award (P.L.), the Josie Robertson Investigator Program at MSKCC (P.L.), the Druckenmiller Center for Lung Cancer Center at MSKCC (P.L.) and a Medical Scientist Training Program grant from the National Institute of General Medical Sciences of the NIH under award number T32GM007739 to the Weill Cornell–Rockefeller–Sloan Kettering Tri-institutional MD–PhD Program (Y.X.). E.d.S. and S.W.L. are supported in part by the MSKCC Pilot Center for Precision Disease Modeling program (U54 OD020355). T.B. is supported by the William C. and Joyce C. O'Neil Charitable Trust, Memorial Sloan Kettering Single-Cell Sequencing Initiative. J.N. is supported by the Knut and Alice Wallenberg Foundation. The authors also acknowledge the MSKCC Support Grant–Core Grant program (P30 CA008748).

Author information

Authors and Affiliations

Authors

Contributions

Y.X., L.M., A.V., M.S., T.T.M. and N.C. performed experiments; L.M., T.B. and J.S.R.-F. performed, analyzed and helped interpret the single-cell sequencing experiments; M.F.B. analyzed the bulk-sequencing data; G.J.R. and B.T.L. provided the patient samples; J.N. and U.S. provided the PDX models; E.d.S. performed the animal experiments; K.S., F.C., T.H. and S.S. performed the mass spectrometry experiments; K.C. performed the FISH experiments, and G.N. analyzed the results. N.R. and S.W.L. provided key scientific insights and reagents; P.L. conceived and supervised the study, designed and performed experiments, and interpreted data; Y.X. and P.L. were the principal writers of the manuscript; and all of the authors reviewed the manuscript and contributed in writing.

Corresponding author

Correspondence to Piro Lito.

Ethics declarations

Competing interests

P.L. is listed as an inventor on a patent application filed by MSKCC that incorporates discoveries described in the manuscript. N.R. is on the scientific advisory board, and has received grant support from, Chugai Pharmaceutical and is on the SAB of Astra-Zeneca, Beigene and Kura. S.S., T.H., K.S. and F.C. are employees of NantOmics.

Supplementary information

Supplementary Tables and Figures

Supplementary Tables 1 and 2 and Supplementary Figures 1–7 (PDF 11679 kb)

Supplementary Dataset (XLSX 2434 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xue, Y., Martelotto, L., Baslan, T. et al. An approach to suppress the evolution of resistance in BRAFV600E-mutant cancer. Nat Med 23, 929–937 (2017). https://doi.org/10.1038/nm.4369

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4369

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