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:

Neoantigen landscape dynamics during human melanoma–T cell interactions

Abstract

Recognition of neoantigens that are formed as a consequence of DNA damage is likely to form a major driving force behind the clinical activity of cancer immunotherapies such as T-cell checkpoint blockade and adoptive T-cell therapy1,2,3,4,5,6,7. Therefore, strategies to selectively enhance T-cell reactivity against genetically defined neoantigens1,8,9,10,11 are currently under development. In mouse models, T-cell pressure can sculpt the antigenicity of tumours, resulting in the emergence of tumours that lack defined mutant antigens12,13. However, whether the T-cell-recognized neoantigen repertoire in human cancers is constant over time is unclear. Here we analyse the stability of neoantigen-specific T-cell responses and the antigens they recognize in two patients with stage IV melanoma treated by adoptive T-cell transfer. The T-cell-recognized neoantigens can be selectively lost from the tumour cell population, either by overall reduced expression of the genes or loss of the mutant alleles. Notably, loss of expression of T-cell-recognized neoantigens was accompanied by development of neoantigen-specific T-cell reactivity in tumour-infiltrating lymphocytes. These data demonstrate the dynamic interactions between cancer cells and T cells, which suggest that T cells mediate neoantigen immunoediting, and indicate that the therapeutic induction of broad neoantigen-specific T-cell responses should be used to avoid tumour resistance.

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: Identification and dynamics of neoepitope-specific T-cell reactivity and neoepitope expression in patient BO.
Figure 2: Identification and dynamics of the dominating intratumoral neoantigen-specific T-cell repertoire in patient AB.
Figure 3: Mutation analysis and neoantigen expression in sequential tumour lines and tumour tissue of patient AB.

Similar content being viewed by others

References

  1. Gubin, M. M. et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515, 577–581 (2014)

    Article  ADS  CAS  Google Scholar 

  2. Le, D. T. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372, 2509–2520 (2015)

    Article  CAS  Google Scholar 

  3. Linnemann, C. et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nat. Med. 21, 81–85 (2015)

    Article  CAS  Google Scholar 

  4. Rizvi, N. A. et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015)

    Article  ADS  CAS  Google Scholar 

  5. Robbins, P. F. et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat. Med. 19, 747–752 (2013)

    Article  CAS  Google Scholar 

  6. Van Allen, E. M. et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 350, 207–211 (2015)

    Article  ADS  CAS  Google Scholar 

  7. van Rooij, N. et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J. Clin. Oncol. 31, e439–e442 (2013)

    Article  Google Scholar 

  8. Carreno, B. M. et al. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science 348, 803–808 (2015)

    Article  ADS  CAS  Google Scholar 

  9. Kreiter, S. et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 520, 692–696 (2015)

    Article  ADS  CAS  Google Scholar 

  10. Schumacher, T. N. & Schreiber, R. D. Neoantigens in cancer immunotherapy. Science 348, 69–74 (2015)

    Article  ADS  CAS  Google Scholar 

  11. Tran, E. et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 344, 641–645 (2014)

    Article  ADS  CAS  Google Scholar 

  12. Matsushita, H. et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 482, 400–404 (2012)

    Article  ADS  CAS  Google Scholar 

  13. Mittal, D., Gubin, M. M., Schreiber, R. D. & Smyth, M. J. New insights into cancer immunoediting and its three component phases—elimination, equilibrium and escape. Curr. Opin. Immunol. 27, 16–25 (2014)

    Article  CAS  Google Scholar 

  14. Andersen, R. S. et al. Dissection of T-cell antigen specificity in human melanoma. Cancer Res. 72, 1642–1650 (2012)

    Article  CAS  Google Scholar 

  15. Kawakami, Y. et al. Recognition of shared melanoma antigens in association with major HLA-A alleles by tumor infiltrating T lymphocytes from 123 patients with melanoma. J. Immunother. 23, 17–27 (2000)

    Article  CAS  Google Scholar 

  16. Lu, Y. C. et al. Efficient identification of mutated cancer antigens recognized by T cells associated with durable tumor regressions. Clin. Cancer Res. 20, 3401–3410 (2014)

    Article  CAS  Google Scholar 

  17. Sharma, P. & Allison, J. P. The future of immune checkpoint therapy. Science 348, 56–61 (2015)

    Article  ADS  CAS  Google Scholar 

  18. Besser, M. J. et al. Clinical responses in a phase II study using adoptive transfer of short-term cultured tumor infiltration lymphocytes in metastatic melanoma patients. Clin. Cancer Res. 16, 2646–2655 (2010)

    Article  CAS  Google Scholar 

  19. Dudley, M. E. et al. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J. Clin. Oncol. 26, 5233–5239 (2008)

    Article  CAS  Google Scholar 

  20. Ellebaek, E. et al. Adoptive cell therapy with autologous tumor infiltrating lymphocytes and low-dose interleukin-2 in metastatic melanoma patients. J. Transl. Med. 10, 169–180 (2012)

    Article  CAS  Google Scholar 

  21. Tran, E. et al. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science 350, 1387–1390 (2015)

    Article  ADS  CAS  Google Scholar 

  22. Gros, A. et al. PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors. J. Clin. Invest. 124, 2246–2259 (2014)

    Article  CAS  Google Scholar 

  23. Verdegaal, E. M. et al. Successful treatment of metastatic melanoma by adoptive transfer of blood-derived polyclonal tumor-specific CD4+ and CD8+ T cells in combination with low-dose interferon-alpha. Cancer Immunol. Immunother. 60, 953–963 (2011)

    Article  CAS  Google Scholar 

  24. Hadrup, S. R. et al. Parallel detection of antigen-specific T-cell responses by multidimensional encoding of MHC multimers. Nat. Methods 6, 520–526 (2009)

    Article  CAS  Google Scholar 

  25. Kwakkenbos, M. J. et al. Generation of stable monoclonal antibody-producing B cell receptor-positive human memory B cells by genetic programming. Nat. Med. 16, 123–128 (2010)

    Article  CAS  Google Scholar 

  26. Wolchok, J. D. et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin. Cancer Res. 15, 7412–7420 (2009)

    Article  CAS  Google Scholar 

  27. Krauthammer, M. et al. Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma. Nat. Genet. 44, 1006–1014 (2012)

    Article  CAS  Google Scholar 

  28. Timm, J. & Walker, C. M. Mutational escape of CD8+ T cell epitopes: implications for prevention and therapy of persistent hepatitis virus infections. Med. Microbiol. Immunol. 204, 29–38 (2015)

    Article  CAS  Google Scholar 

  29. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to G. J. Liefers for handling of patient material, and T. van Wezel and D. Ruano for the setup of the M13-amplicon sequencing technology. This work was supported by Dutch Cancer Society grant UL 2012-5544 (to E.M.E.V., S.H.v.d.B. and J.B.A.G.H), the Anticancer Fund (to E.M.E.V. and S.H.v.d.B.), Dutch Cancer Society grant NKI 2012-5463 (to T.N.S, J.B.A.G.H. and S.H.v.d.B), the Dutch Cancer Society Queen Wilhelmina Award NKI 2013-6122 (T.N.S), Dutch Cancer Society grant UVA 2010-4822 (to H.S.), Fight Colorectal Cancer-Michael’s Mission-AACR Fellowship (2015) and Alpe d’HuZes/KWF Bas Mulder Award (to N.F.C.C.d.M.).

Author information

Authors and Affiliations

Authors

Contributions

E.M.E.V. designed, performed, analysed and interpreted experiments and wrote the paper, N.F.C.C.d.M., M.V., C.E.v.d.M. and T.H. designed, performed, analysed and interpreted the experiments, M.M.v.B. analysed and interpreted next-generation sequencing data and performed peptide selection, R.S.A. and S.R.H. designed, performed and interpreted the combinatorial coding experiments, R.S. generated BCL-6/BCL-XL immortalized B-cell lines. E.H.W.K and J.B.A.G.H. supervised treatment of patients, supplied patient material and provided clinical interpretation of results. H.S. developed the BCL-6/BCL-XL immortalization technology. T.N.S. interpreted the data and wrote the paper, S.H.v.d.B. supervised the project, designed and interpreted the experiments, and wrote the paper.

Corresponding author

Correspondence to Els M. E. Verdegaal.

Ethics declarations

Competing interests

AIMM Therapeutics holds IP to immortalize human B cells, and AIMM Therapeutics/ The Netherlands Cancer Institute hold IP for the use of immortalized human B cells to identify T cell epitopes. H.S. and R.S. are employees and stockholders of AIMM Therapeutics. T.N.S. is a scientific advisor and stockholder of AIMM Therapeutics.

Additional information

Reviewer Information

Nature thanks M. Gubin, U. Sahin and the other anonymous reviewer(s) for their contribution to the peer review of this work.

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Verdegaal, E., de Miranda, N., Visser, M. et al. Neoantigen landscape dynamics during human melanoma–T cell interactions. Nature 536, 91–95 (2016). https://doi.org/10.1038/nature18945

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature18945

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

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