High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma (original) (raw)

Nature Medicine volume 21, pages 81–85 (2015)Cite this article

Subjects

A Corrigendum to this article was published on 06 October 2016

This article has been updated

Abstract

Tumor-specific neo-antigens that arise as a consequence of mutations1,2 are thought to be important for the therapeutic efficacy of cancer immunotherapies3,4,5. Accumulating evidence suggests that neo-antigens may be commonly recognized by intratumoral CD8+ T cells3,4,5,6,7, but it is unclear whether neo-antigen–specific CD4+ T cells also frequently reside within human tumors. In view of the accepted role of tumor-specific CD4+ T-cell responses in tumor control8,9,10, we addressed whether neo-antigen–specific CD4+ T-cell reactivity is a common property in human melanoma.

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

Access options

Subscribe to this journal

Receive 12 print issues and online access

$209.00 per year

only $17.42 per issue

Buy this article

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

Additional access options:

Similar content being viewed by others

Change history

In the version of this article initially published, the article did not mention some restrictions on the availability of reagents. The following text has been added to the HTML and PDF versions of the paper: “The retroviral vectors containing BCL-6 and BCL-xL have been generated by a for-profit company, AIMM Therapeutics, which makes the plasmids available. Obtaining the plasmids requires an MTA (http://www.aimmtherapeutics.com/partnering/academic-collaboration/) that includes financial obligations.” The error has been corrected in the HTML and PDF versions of the article.

References

  1. Alexandrov, L.B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).
    Article CAS PubMed PubMed Central Google Scholar
  2. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013).
    Article CAS PubMed PubMed Central Google Scholar
  3. 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 PubMed PubMed Central Google Scholar
  4. 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 PubMed Google Scholar
  5. Lu, Y.C. et al. Mutated PPP1R3B is recognized by T cells used to treat a melanoma patient who experienced a durable complete tumor regression. J. Immunol. 190, 6034–6042 (2013).
    Article CAS PubMed Google Scholar
  6. Wick, D.A. et al. Surveillance of the tumor mutanome by T cells during progression from primary to recurrent ovarian cancer. Clin. Cancer Res. 20, 1125–1134 (2014).
    Article CAS PubMed Google Scholar
  7. Rajasagi, M. et al. Systematic identification of personal tumor-specific neoantigens in chronic lymphocytic leukemia. Blood 124, 453–462 (2014).
    Article CAS PubMed PubMed Central Google Scholar
  8. Kenter, G.G. et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N. Engl. J. Med. 361, 1838–1847 (2009).
    Article CAS PubMed Google Scholar
  9. Quezada, S.A. et al. Tumor-reactive CD4+ T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts. J. Exp. Med. 207, 637–650 (2010).
    Article CAS PubMed PubMed Central Google Scholar
  10. Ossendorp, F., Mengede, E., Camps, M., Filius, R. & Melief, C.J. Specific T helper cell requirement for optimal induction of cytotoxic T lymphocytes against major histocompatibility complex class II negative tumors. J. Exp. Med. 187, 693–702 (1998).
    Article CAS PubMed PubMed Central Google Scholar
  11. Friedman, K.M. et al. Tumor-specific CD4+ melanoma tumor-infiltrating lymphocytes. J. Immunother. 35, 400–408 (2012).
    Article CAS PubMed PubMed Central Google Scholar
  12. Kitano, S. et al. Enhancement of tumor-reactive cytotoxic CD4+ T cell responses after ipilimumab treatment in four advanced melanoma patients. Cancer Immunol. Res. 1, 235–244 (2013).
    Article CAS PubMed Google Scholar
  13. 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 CAS PubMed PubMed Central Google Scholar
  14. Champiat, S., Ferte, C., Lebel-Binay, S., Eggermont, A. & Soria, J.C. Exomics and immunogenics: bridging mutational load and immune checkpoints efficacy. OncoImmunology 3, e27817 (2014).
    Article PubMed PubMed Central Google Scholar
  15. Britten, C.M. et al. The regulatory landscape for actively personalized cancer immunotherapies. Nat. Biotechnol. 31, 880–882 (2013).
    Article CAS PubMed Google Scholar
  16. Overwijk, W.W., Wang, E., Marincola, F.M., Rammensee, H.G. & Restifo, N.P. Mining the mutanome: developing highly personalized Immunotherapies based on mutational analysis of tumors. J. Immunother. Cancer 1, 11 (2013).
    Article PubMed PubMed Central Google Scholar
  17. 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 PubMed PubMed Central Google Scholar
  18. Kwakkenbos, M.J. et al. Genetic manipulation of B cells for the isolation of rare therapeutic antibodies from the human repertoire. Methods 65, 38–43 (2014).
    Article CAS PubMed Google Scholar
  19. 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 PubMed Google Scholar
  20. Mason, D. A very high level of crossreactivity is an essential feature of the T-cell receptor. Immunol. Today 19, 395–404 (1998).
    Article CAS PubMed Google Scholar
  21. Deffrennes, V. et al. Constitutive expression of MHC class II genes in melanoma cell lines results from the transcription of class II transactivator abnormally initiated from its B cell-specific promoter. J. Immunol. 167, 98–106 (2001).
    Article CAS PubMed Google Scholar
  22. Kvistborg, P. et al. TIL therapy broadens the tumor-reactive CD8(+) T cell compartment in melanoma patients. OncoImmunology 1, 409–418 (2012).
    Article PubMed PubMed Central Google Scholar
  23. Kvistborg, P. et al. Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci. Transl. Med. 6, 254ra128 (2014).
    Article PubMed Google Scholar
  24. 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 PubMed PubMed Central Google Scholar
  25. Jandus, C. et al. Tumor antigen-specific FOXP3+ CD4 T cells identified in human metastatic melanoma: peptide vaccination results in selective expansion of Th1-like counterparts. Cancer Res. 69, 8085–8093 (2009).
    Article CAS PubMed Google Scholar
  26. Mautner, J., Jaffee, E.M. & Pardoll, D.M. Tumor-specific CD4+ T cells from a patient with renal cell carcinoma recognize diverse shared antigens. Int. J. Cancer 115, 752–759 (2005).
    Article CAS PubMed Google Scholar
  27. Wang, R.F., Wang, X., Atwood, A.C., Topalian, S.L. & Rosenberg, S.A. Cloning genes encoding MHC class II-restricted antigens: mutated CDC27 as a tumor antigen. Science 284, 1351–1354 (1999).
    Article CAS PubMed Google Scholar
  28. Hunder, N.N. et al. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N. Engl. J. Med. 358, 2698–2703 (2008).
    Article CAS PubMed PubMed Central Google Scholar
  29. Varela, I. et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 469, 539–542 (2011).
    Article CAS PubMed PubMed Central Google Scholar
  30. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics 26, 589–595 (2010).
    Article PubMed PubMed Central Google Scholar
  31. Trapnell, C., Pachter, L. & Salzberg, S.L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).
    Article CAS PubMed PubMed Central Google Scholar
  32. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).
    Article CAS PubMed PubMed Central Google Scholar
  33. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).
    Article CAS PubMed PubMed Central Google Scholar
  34. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).
    PubMed PubMed Central Google Scholar
  35. Bolotin, D.A. et al. MiTCR: software for T-cell receptor sequencing data analysis. Nat. Methods 10, 813–814 (2013).
    Article CAS PubMed Google Scholar

Download references

Acknowledgements

We are grateful to A. Pfauth, F. van Diepen and C. Bachas for flow cytometric support; W. van de Kasteele and T. de Jong for technical assistance; N. van Rooij and B. Heemskerk for handling of patient material; and R. Kluin, M. Nieuwland and R. van Kerkhoven for support with next generation sequencing. We thank G. Bendle and P. Kvistborg for critical reading of the manuscript, and members from the Schumacher and Haanen laboratories for useful discussions. This work was supported by Worldwide Cancer Research grant 14-0321 (to C.L. and T.N.M.S.), the PhD Fellowship Program of Boehringer Ingelheim Fonds–Foundation for Basic Research in Biomedicine (to C.L.), Dutch Cancer Society grants: UVA 2010-4822 (to H.S.), NKI 2012-5463 (to J.B.A.G.H., S.H.v.d.B. and T.N.M.S.) and UL 2012-5544 (to E.M.E.V.), The Wellcome Trust Research Training Fellowship for Clinicians (to S.B.), The Wellcome Trust Grant WT098051 (to M.R.S.), the Anticancer Fund (to E.M.E.V., M.V.) the K.G. Jebsen Foundation (to T.N.M.S.), EU FP7 SUPERSIST, and the Stand Up To Cancer–Cancer Research Institute Cancer Immunology Translational Cancer Research Grant (to T.N.M.S.). SU2C is a program of the Entertainment Industry Foundation administered by the American Association for Cancer Research.

Author information

Author notes

  1. Carsten Linnemann, Marit M van Buuren and Laura Bies: These authors contributed equally to this work.
  2. Hergen Spits and Sjoerd H van der Burg: These authors jointly supervised this work.

Authors and Affiliations

  1. Division of Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
    Carsten Linnemann, Marit M van Buuren, Laura Bies, Jorg J A Calis, John B A G Haanen & Ton N M Schumacher
  2. Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
    Els M E Verdegaal, Marten Visser & Sjoerd H van der Burg
  3. AIMM Therapeutics B.V., Amsterdam, the Netherlands
    Remko Schotte & Hergen Spits
  4. Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
    Sam Behjati & Michael R Stratton
  5. Central Genomics Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
    Arno Velds
  6. Peptide Synthesis Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
    Henk Hilkmann & Dris el Atmioui
  7. Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
    John B A G Haanen

Authors

  1. Carsten Linnemann
    You can also search for this author inPubMed Google Scholar
  2. Marit M van Buuren
    You can also search for this author inPubMed Google Scholar
  3. Laura Bies
    You can also search for this author inPubMed Google Scholar
  4. Els M E Verdegaal
    You can also search for this author inPubMed Google Scholar
  5. Remko Schotte
    You can also search for this author inPubMed Google Scholar
  6. Jorg J A Calis
    You can also search for this author inPubMed Google Scholar
  7. Sam Behjati
    You can also search for this author inPubMed Google Scholar
  8. Arno Velds
    You can also search for this author inPubMed Google Scholar
  9. Henk Hilkmann
    You can also search for this author inPubMed Google Scholar
  10. Dris el Atmioui
    You can also search for this author inPubMed Google Scholar
  11. Marten Visser
    You can also search for this author inPubMed Google Scholar
  12. Michael R Stratton
    You can also search for this author inPubMed Google Scholar
  13. John B A G Haanen
    You can also search for this author inPubMed Google Scholar
  14. Hergen Spits
    You can also search for this author inPubMed Google Scholar
  15. Sjoerd H van der Burg
    You can also search for this author inPubMed Google Scholar
  16. Ton N M Schumacher
    You can also search for this author inPubMed Google Scholar

Contributions

C.L., M.M.v.B. and L.B. designed, performed, analyzed and interpreted experiments and wrote the paper. E.M.E.V. and M.V. designed, performed, analyzed, and interpreted the analysis of subject BO. R.S. generated BCL-6/BCL-XL–immortalized B-cell lines and provided essential culture reagents and advice for the development of the B-cell screening platform. J.J.A.C. developed bioinformatics scripts for TCR sequence identification in NGS data. H.H. and D.e.A. synthesized peptide libraries. S.B., M.R.S. and A.V. performed and analyzed exome and RNA sequencing and developed the pipeline for identification of somatic mutations. J.B.A.G.H. supervised the clinical treatment of NKI TIL patients, supplied patient material and provided clinical interpretation of results. H.S. developed the BCL-6/BCL-XL immortalization technology and provided essential culture reagents and advice for the development of the B-cell screening platform. S.H.v.d.B. supervised, analyzed and interpreted the analysis of subject BO. T.N.M.S. supervised the project, designed and interpreted all experiments, and wrote the paper.

Corresponding authors

Correspondence toCarsten Linnemann or Ton N M Schumacher.

Ethics declarations

Competing interests

The technology described in this manuscript is the subject of an EU patent application co-owned by NKI-AVL and AIMM Therapeutics B.V. Based on NKI-AVL policy on management of intellectual property, C.L., M.M.v.B., L.B. and T.N.M.S would be entitled to a portion of received royalty income. H.S. and R.S. own certificates of AIMM Therapeutics B.V. (minority stake). T.N.M.S. is a scientific advisor of AIMM Therapeutics B.V. and indirectly owns stock of AIMM Therapeutics B.V. (minority stake).

Supplementary information

Source data

Rights and permissions

About this article

Cite this article

Linnemann, C., van Buuren, M., Bies, L. 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). https://doi.org/10.1038/nm.3773

Download citation

Associated content