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:

Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients

Abstract

Detection of lymphocytes that target tumor-specific mutant neoantigens—derived from products encoded by mutated genes in the tumor—is mostly limited to tumor-resident lymphocytes1,2, but whether these lymphocytes often occur in the circulation is unclear. We recently reported that intratumoral expression of the programmed cell death 1 (PD-1) receptor can guide the identification of the patient-specific repertoire of tumor-reactive CD8+ lymphocytes that reside in the tumor3. In view of these findings, we investigated whether PD-1 expression on peripheral blood lymphocytes could be used as a biomarker to detect T cells that target neoantigens. By using a high-throughput personalized screening approach, we identified neoantigen-specific lymphocytes in the peripheral blood of three of four melanoma patients. Despite their low frequency in the circulation, we found that CD8+PD-1+, but not CD8+PD-1, cell populations had lymphocytes that targeted 3, 3 and 1 unique, patient-specific neoantigens, respectively. We show that neoantigen-specific T cells and gene-engineered lymphocytes expressing neoantigen-specific T cell receptors (TCRs) isolated from peripheral blood recognized autologous tumors. Notably, the tumor-antigen specificities and TCR repertoires of the circulating and tumor-infiltrating CD8+PD-1+ cells appeared similar, implying that the circulating CD8+PD-1+ lymphocytes could provide a window into the tumor-resident antitumor lymphocytes. Thus, expression of PD-1 identifies a diverse and patient-specific antitumor T cell response in peripheral blood, providing a novel noninvasive strategy to develop personalized therapies using neoantigen-reactive lymphocytes or TCRs to treat cancer.

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: Frequency of PD-1 expression on circulating and tumor-resident CD8+ lymphocytes and prospective identification of circulating neoantigen-reactive cells in melanoma patients.
Figure 2: Characterization of neoantigen-specific lymphocytes isolated from the circulating CD8+PD1+ subset of subject NCI-3998.
Figure 3: Identification of neoantigens targeted by circulating CD8+PD-1+ cells isolated from subjects NCI-3784, NCI-3903 and NCI-3713.
Figure 4: Recognition of tumors and self-antigens by TCRs or CD8+ T cells isolated from peripheral blood, and comparison of the specificity and TCR repertoire between circulating and tumor-infiltrating CD8+ T cell subsets.

Similar content being viewed by others

Accession codes

Primary accessions

BioProject

References

  1. 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 

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

    Article  CAS  Google Scholar 

  3. 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 

  4. Rosenberg, S.A. et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. J. Am. Med. Assoc. 271, 907–913 (1994).

    Article  CAS  Google Scholar 

  5. Rosenberg, S.A. et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T cell–transfer immunotherapy. Clin. Cancer Res. 17, 4550–4557 (2011).

    Article  CAS  Google Scholar 

  6. Hamid, O. et al. Safety and tumor responses with lambrolizumab (anti–PD-1) in melanoma. N. Engl. J. Med. 369, 134–144 (2013).

    Article  CAS  Google Scholar 

  7. Larkin, J. et al. Combined nivolumab and ipilimumab, or monotherapy, in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).

    Article  Google Scholar 

  8. Postow, M.A. et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N. Engl. J. Med. 372, 2006–2017 (2015).

    Article  Google Scholar 

  9. Topalian, S.L. et al. Survival, durable tumor remission and long-term safety in patients with advanced melanoma receiving nivolumab. J. Clin. Oncol. 32, 1020–1030 (2014).

    Article  CAS  Google Scholar 

  10. Alexandrov, L.B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  CAS  Google Scholar 

  11. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013).

    Article  CAS  Google Scholar 

  12. 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  CAS  Google Scholar 

  13. Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).

    Article  Google Scholar 

  14. 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 

  15. 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 

  16. 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  Google Scholar 

  17. 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  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  19. Rosenberg, S.A. & Restifo, N.P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62–68 (2015).

    Article  CAS  Google Scholar 

  20. 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 

  21. Cohen, C.J. et al. Isolation of neoantigen-specific T cells from tumor and peripheral lymphocytes. J. Clin. Invest. 125, 3981–3991 (2015).

    Article  Google Scholar 

  22. Altman, J.D. et al. Phenotypic analysis of antigen-specific T lymphocytes. Science 274, 94–96 (1996).

    Article  CAS  Google Scholar 

  23. Lennerz, V. et al. The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proc. Natl. Acad. Sci. USA 102, 16013–16018 (2005).

    Article  CAS  Google Scholar 

  24. Ahmadzadeh, M. et al. Tumor antigen–specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114, 1537–1544 (2009).

    Article  CAS  Google Scholar 

  25. Baitsch, L. et al. Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J. Clin. Invest. 121, 2350–2360 (2011).

    Article  CAS  Google Scholar 

  26. Inozume, T. et al. Selection of CD8+PD-1+ lymphocytes in fresh human melanomas enriches for tumor-reactive T cells. J. Immunother. 33, 956–964 (2010).

    Article  CAS  Google Scholar 

  27. Linnemann, C. et al. High-throughput identification of antigen-specific TCRs by TCR gene capture. Nat. Med. 19, 1534–1541 (2013).

    Article  CAS  Google Scholar 

  28. Van Allen, E.M. et al. Whole-exome sequencing and clinical interpretation of formalin-fixed, paraffin-embedded tumor samples to guide precision cancer medicine. Nat. Med. 20, 682–688 (2014).

    Article  CAS  Google Scholar 

  29. Lee, H.B. et al. The use of FNA samples for whole-exome sequencing and detection of somatic mutations in breast cancer surgical specimens. Cancer Cytopathol. 123, 669–677 (2015).

    Article  CAS  Google Scholar 

  30. Murtaza, M. et al. Noninvasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 497, 108–112 (2013).

    Article  CAS  Google Scholar 

  31. Lohr, J.G. et al. Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer. Nat. Biotechnol. 32, 479–484 (2014).

    Article  CAS  Google Scholar 

  32. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  Google Scholar 

  33. 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  Google Scholar 

  34. Wu, T.C. et al. Engineering an intracellular pathway for major histocompatibility complex class II presentation of antigens. Proc. Natl. Acad. Sci. USA 92, 11671–11675 (1995).

    Article  CAS  Google Scholar 

  35. Bai, Y., Ni, M., Cooper, B., Wei, Y. & Fury, W. Inference of high-resolution HLA types using genome-wide RNA- or DNA-sequencing reads. BMC Genomics 15, 325 (2014).

    Article  Google Scholar 

  36. Vita, R. et al. The immune epitope database (IEDB) 3.0. Nucleic Acids Res. 43, D405–D412 (2015).

    Article  CAS  Google Scholar 

  37. Nielsen, J.S., Wick, D.A., Tran, E., Nelson, B.H. & Webb, J.R. An in vitro–transcribed mRNA polyepitope construct encoding 32 distinct HLA class I–restricted epitopes from CMV, EBV and influenza for use as a functional control in human immune-monitoring studies. J. Immunol. Methods 360, 149–156 (2010).

    Article  CAS  Google Scholar 

  38. Cohen, C.J., Zhao, Y., Zheng, Z., Rosenberg, S.A. & Morgan, R.A. Enhanced antitumor activity of murine-human hybrid T cell receptor (TCR) in human lymphocytes is associated with improved pairing and TCR/CD3 stability. Cancer Res. 66, 8878–8886 (2006).

    Article  CAS  Google Scholar 

  39. Cohen, C.J. et al. Enhanced antitumor activity of T cells engineered to express T cell receptors with a second disulfide bond. Cancer Res. 67, 3898–3903 (2007).

    Article  CAS  Google Scholar 

  40. Haga-Friedman, A., Horovitz-Fried, M. & Cohen, C.J. Incorporation of transmembrane hydrophobic mutations in the TCR enhance its surface expression and T cell functional avidity. J. Immunol. 188, 5538–5546 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Center for Cancer Research intramural research program of the NCI, US National Institutes of Health (NIH). We thank the members of the Surgery branch for helpful discussions, R. Somerville and members of the tumor-infiltrating lymphocytes (TIL) lab for technical support, and L. Liu for kindly providing the PD-1–specific AMP-514 antibody. This work used the computational resources of the NIH High-Performance Computing (HPC) Biowulf cluster (http://hpc.nih.gov).

Author information

Authors and Affiliations

Authors

Contributions

A.G. designed, performed, analyzed and interpreted experiments; M.R.P. performed and analyzed experiments; E.T. and A.P. analyzed and interpreted the data; P.F.R. helped in selecting the mutant antigens screened and interpreted data; S.I. performed experiments; T.D.P. and J.S.C. provided valuable advice and reagents for subjects NCI-3713 and NCI-3903; J.J.G. performed the tumor-exome and transcriptome bioinformatics analysis; I.M.R. provided technical support; K.T.-M. provided valuable advice and reagents for subject NCI-3998; J.R.W. established tumor cell lines; J.C.Y. supervised the clinical treatment of the patients included in the study and interpreted the data; and S.A.R. supervised the clinical treatment of the patients included in the study, supervised the project, designed experiments and interpreted the data. A.G. and S.A.R. wrote the manuscript.

Corresponding author

Correspondence to Steven A Rosenberg.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Tables 1–9 (PDF 2964 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gros, A., Parkhurst, M., Tran, E. et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat Med 22, 433–438 (2016). https://doi.org/10.1038/nm.4051

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4051

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