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.

Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens

Abstract

The immune system influences the fate of developing cancers by not only functioning as a tumour promoter that facilitates cellular transformation, promotes tumour growth and sculpts tumour cell immunogenicity1,2,3,4,5,6, but also as an extrinsic tumour suppressor that either destroys developing tumours or restrains their expansion1,2,7. Yet, clinically apparent cancers still arise in immunocompetent individuals in part as a consequence of cancer-induced immunosuppression. In many individuals, immunosuppression is mediated by cytotoxic T-lymphocyte associated antigen-4 (CTLA-4) and programmed death-1 (PD-1), two immunomodulatory receptors expressed on T cells8,9. Monoclonal-antibody-based therapies targeting CTLA-4 and/or PD-1 (checkpoint blockade) have yielded significant clinical benefits—including durable responses—to patients with different malignancies10,11,12,13. However, little is known about the identity of the tumour antigens that function as the targets of T cells activated by checkpoint blockade immunotherapy and whether these antigens can be used to generate vaccines that are highly tumour-specific. Here we use genomics and bioinformatics approaches to identify tumour-specific mutant proteins as a major class of T-cell rejection antigens following anti-PD-1 and/or anti-CTLA-4 therapy of mice bearing progressively growing sarcomas, and we show that therapeutic synthetic long-peptide vaccines incorporating these mutant epitopes induce tumour rejection comparably to checkpoint blockade immunotherapy. Although mutant tumour-antigen-specific T cells are present in progressively growing tumours, they are reactivated following treatment with anti-PD-1 and/or anti-CTLA-4 and display some overlapping but mostly treatment-specific transcriptional profiles, rendering them capable of mediating tumour rejection. These results reveal that tumour-specific mutant antigens are not only important targets of checkpoint blockade therapy, but they can also be used to develop personalized cancer-specific vaccines and to probe the mechanistic underpinnings of different checkpoint blockade treatments.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Mutations in Lama4 and Alg8 form top predicted d42m1-T3 epitopes.
Figure 2: Mutant Lama4 and mAlg8 are therapeutically relevant d42m1-T3 TSMA.
Figure 3: Differential effects of checkpoint blockade therapy on tumour-antigen-specific CD8+ T cells.
Figure 4: Checkpoint blockade therapy alters the functional phenotypes of tumour-antigen-specific CD8+ T cells.

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

RNA-sequencing data are available at Gene Expression Omnibus (GEO) repository at http://www.ncbi.nlm.nih.gov/geo/ (accession number GSE62771).

References

  1. Shankaran, V. et al. IFNγ and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 410, 1107–1111 (2001)

    ADS  CAS  PubMed  Google Scholar 

  2. Dunn, G. P., Bruce, A. T., Ikeda, H., Old, L. J. & Schreiber, R. D. Cancer immunoediting: from immunosurveillance to tumor escape. Nature Immunol. 3, 991–998 (2002)

    CAS  Google Scholar 

  3. Mantovani, A., Allavena, P., Sica, A. & Balkwill, F. Cancer-related inflammation. Nature 454, 436–444 (2008)

    ADS  CAS  PubMed  Google Scholar 

  4. Grivennikov, S. I., Greten, F. R. & Karin, M. Immunity, inflammation, and cancer. Cell 140, 883–899 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Trinchieri, G. Cancer and inflammation: an old intuition with rapidly evolving new concepts. Annu. Rev. Immunol. 30, 677–706 (2012)

    CAS  PubMed  Google Scholar 

  6. Coussens, L. M., Zitvogel, L. & Palucka, A. K. Neutralizing tumor-promoting chronic inflammation: a magic bullet? Science 339, 286–291 (2013)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Koebel, C. M. et al. Adaptive immunity maintains occult cancer in an equilibrium state. Nature 450, 903–907 (2007)

    ADS  CAS  PubMed  Google Scholar 

  8. Quezada, S. A., Peggs, K. S., Simpson, T. R. & Allison, J. P. Shifting the equilibrium in cancer immunoediting: from tumor tolerance to eradication. Immunol. Rev. 241, 104–118 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Pardoll, D. M. The blockade of immune checkpoints in cancer immunotherapy. Nature Rev. Cancer 12, 252–264 (2012)

    CAS  Google Scholar 

  10. Wolchok, J. D. et al. Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 369, 122–133 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Hodi, F. S. et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363, 711–723 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Paul, S. et al. HLA class I alleles are associated with peptide-binding repertoires of different size, affinity, and immunogenicity. J. Immunol. 191, 5831–5839 (2013)

    CAS  PubMed  Google Scholar 

  16. West, E. E. et al. Tight regulation of memory CD8+ T cells limits their effectiveness during sustained high viral load. Immunity 35, 285–298 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Wherry, E. J. T cell exhaustion. Nature Immunol. 12, 492–499 (2011)

    CAS  Google Scholar 

  18. Castle, J. C. et al. Exploiting the mutanome for tumor vaccination. Cancer Res. 72, 1081–1091 (2012)

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  20. Fritsch, E. F. et al. HLA-binding properties of tumor neoepitopes in humans. Cancer Immunol. Res. 2, 522–529 (2014)

    CAS  PubMed  PubMed Central  Google Scholar 

  21. 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)

    PubMed  Google Scholar 

  22. Curran, M. A., Montalvo, W., Yagita, H. & Allison, J. P. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl Acad. Sci. USA 107, 4275–4280 (2010)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Brunner, M. C. et al. CTLA-4-mediated inhibition of early events of T cell proliferation. J. Immunol. 162, 5813–5820 (1999)

    CAS  PubMed  Google Scholar 

  24. Keir, M. E., Butte, M. J., Freeman, G. J. & Sharpe, A. H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 26, 677–704 (2008)

    CAS  PubMed  Google Scholar 

  25. Okazaki, T., Chikuma, S., Iwai, Y., Fagarasan, S. & Honjo, T. A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application. Nature Immunol. 14, 1212–1218 (2013)

    CAS  Google Scholar 

  26. Duraiswamy, J., Kaluza, K. M., Freeman, G. J. & Coukos, G. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res. 73, 3591–3603 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Spiotto, M. T., Rowley, D. A. & Schreiber, H. Bystander elimination of antigen loss variants in established tumors. Nature Med. 10, 294–298 (2004)

    CAS  PubMed  Google Scholar 

  28. Wolkers, M. C., Brouwenstijn, N., Bakker, A. H., Toebes, M. & Schumacher, T. N. Antigen bias in T cell cross-priming. Science 304, 1314–1317 (2004)

    ADS  CAS  PubMed  Google Scholar 

  29. Corbière, V. et al. Antigen spreading contributes to MAGE vaccination-induced regression of melanoma metastases. Cancer Res. 71, 1253–1262 (2011)

    PubMed  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8α+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100 (2008)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Peters, B. & Sette, A. Generating quantitative models describing the sequence specificity of biological processes with the stabilized matrix method. BMC Bioinformatics 6, 132 (2005)

    PubMed  PubMed Central  Google Scholar 

  33. Lundegaard, C. et al. NetMHC-3.0: accurate web accessible predictions of human, mouse and monkey MHC class I affinities for peptides of length 8-11. Nucleic Acids Res. 36, W509–W512 (2008)

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Hoof, I. et al. NetMHCpan, a method for MHC class I binding prediction beyond humans. Immunogenetics 61, 1–13 (2009)

    CAS  PubMed  Google Scholar 

  35. Nielsen, M., Lundegaard, C., Lund, O. & Kesmir, C. The role of the proteasome in generating cytotoxic T-cell epitopes: insights obtained from improved predictions of proteasomal cleavage. Immunogenetics 57, 33–41 (2005)

    CAS  PubMed  Google Scholar 

  36. Esquivel, F., Yewdell, J. & Bennink, J. RMA/S cells present endogenously synthesized cytosolic proteins to class I-restricted cytotoxic T lymphocytes. J. Exp. Med. 175, 163–168 (1992)

    CAS  PubMed  Google Scholar 

  37. Toebes, M. et al. Design and use of conditional MHC class I ligands. Nature Med. 12, 246–251 (2006)

    CAS  PubMed  Google Scholar 

  38. Andersen, R. S. et al. Parallel detection of antigen-specific T cell responses by combinatorial encoding of MHC multimers. Nature Protocols 7, 891–902 (2012)

    CAS  PubMed  Google Scholar 

  39. Kvistborg, P. et al. TIL therapy broadens the tumor-reactive CD8+ T cell compartment in melanoma patients. OncoImmunology 1, 409–418 (2012)

    PubMed  PubMed Central  Google Scholar 

  40. Kowalewski, D. J. & Stevanovic, S. Biochemical large-scale identification of MHC class I ligands. Methods Mol. Biol. 960, 145–157 (2013)

    CAS  PubMed  Google Scholar 

  41. Thommen, D. S. et al. Two preferentially expressed proteins protect vascular endothelial cells from an attack by peptide-specific CTL. J. Immunol. 188, 5283–5292 (2012)

    CAS  PubMed  Google Scholar 

  42. Domon, B. & Aebersold, R. Options and considerations when selecting a quantitative proteomics strategy. Nature Biotechnol. 28, 710–721 (2010)

    CAS  Google Scholar 

  43. MacLean, B. et al. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 26, 966–968 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Lange, V., Picotti, P., Domon, B. & Aebersold, R. Selected reaction monitoring for quantitative proteomics: a tutorial. Mol. Syst. Biol. 4, 222 (2008)

    PubMed  PubMed Central  Google Scholar 

  45. Escher, C. et al. Using iRT, a normalized retention time for more targeted measurement of peptides. Proteomics 12, 1111–1121 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to K. Murphy for the Batf3−/− mice, T. Hansen for providing MHC class I antibodies and the H-2Kb construct, D. Fremont for the human β2m construct, and the National Institutes of Health (NIH) Tetramer Core Facility for producing MHC class I tetramers. We also thank R. Ahmed and M. Hashimoto for the multiplex staining strategy used to define functional and dysfunctional T cells. We thank A. Bensimon, O. Schubert and P. Kouvonen for instrument maintenance and for technical support with the mass spectrometry measurements and R. Vanganipuram, M. Selby and J. Valle for generating and supplying anti-PD-1 and anti-CTLA-4 in endotoxin-free sterile form. We also thank K. Sheehan, P. Allen, G. Dunn and R. Chan for constructive criticisms and comments, all members of the Schreiber laboratory for discussions, and the many members of The Genome Institute at Washington University School of Medicine. We would also like to thank W. Song for his assistance with the bioinformatics approaches, P. Kvistborg for assistance with tetramer combinatorial coding, and Christopher Nelson for advice with peptide-MHC monomer purification. This work was supported by grants to R.D.S. from the National Cancer Institute (RO1 CA043059, U01 CA141541), the Cancer Research Institute and the WWW.W Foundation; to R.D.S. and W.E.G. from The Siteman Cancer Center/Barnes-Jewish Hospital (Cancer Frontier Fund); to W.E.G. from Susan G. Komen for the Cure (Promise grant); to E.R.M. from the National Human Genome Research Institute; to G.J.F. from the National Institute of Health (P50 CA101942, P01 AI054456, P50 CA101942); to A.H.S. from the National Institute of Health (P50 CA101942); and to T.N.S. from the Dutch Cancer Society (Queen Wilhelmina Research Award). E.C. is supported by a Marie Curie Intra-European Fellowship within the Seventh Framework Programme of the European Community for Research. M.M.G. was supported by a postdoctoral training grant (T32 CA00954729) from the National Cancer Institute and is currently supported by a postdoctoral training grant (Irvington Postdoctoral Fellowship) from the Cancer Research Institute. Aspects of studies at Washington University were performed with assistance by the Immunomonitoring Laboratory of the Center for Human Immunology and Immunotherapy Programs and the Siteman Comprehensive Cancer Center.

Author information

Authors and Affiliations

Authors

Contributions

M.M.G. and R.D.S. were involved in all aspects of this study including planning and performing experiments, analysing and interpreting data, and writing the manuscript. X.Z. performed peptide binding experiments, helped design and perform the vaccine experiments. H.S., E.C., R.A. and H.-G.R. planned and performed the mass spectrometry analyses, interpreted the data and were involved in writing the manuscript. T.N., J.P.W., C.D.A., M.D.V., S.S.K.L. and E.L.P., participated in assessing the phenotypes of the tumour-specific T-cell lines, interpreting the data and in writing the manuscript. M.T. helped generate MHC class I multimers. A.J.K., J.P.A., G.J.F. and A.H.S. provided mAbs, helped plan the checkpoint blockade therapy experiments, and contributed to writing the manuscript. T.N.S. helped generate MHC class I multimers, analysed data and was involved in writing the manuscript. W.-J.K., G.E.M. and C.J.M.M. produced and purified the synthetic long peptides, participated in the planning of the vaccine experiments, analysed data and were involved in writing the manuscript. J.H. and E.R.M. were responsible for genomic analyses and epitope prediction and participated in writing the manuscript. W.E.G. contributed to the design and analysis of peptide binding and vaccine experiments and in writing the manuscript. Y.I. and M.N.A were responsible for optimizing the epitope prediction method, performing the RNA-sequencing analyses, analysing data and writing the manuscript. R.D.S. oversaw all the work performed.

Corresponding author

Correspondence to Robert D. Schreiber.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Innate and adaptive immune components are required for rejection of d42m1-T3 after checkpoint blockade therapy.

a, Cohorts of Rag2−/−, Batf3−/− or wild-type mice were treated with control mAb, anti-CD4, anti-CD8α or anti-IFN-γ mAbs and then were injected with 1 × 106 d42m1-T3 tumour cells subcutaneously and subsequently treated with anti-CTLA-4 on days 3, 6 and 9 post-transplant. b, c, d42m1-T3 (b) or F244 (c) tumour cells were injected subcutaneously into wild-type mice (n = 5) that were subsequently treated with anti-PD-1 on days 3, 6, and 9. Fifty days after tumours were rejected, mice were rechallenged with d42m1-T3 or F244 tumour cells. Data are presented as average tumour diameter ± s.e.m. of 5 mice per group and are representative of at least two independent experiments.

Extended Data Figure 2 H-2Db mutant epitopes of d42m1-T3 tumours.

a, Missense mutations in d42m1-T3 were subjected to in silico analysis for the potential to form H-2Db-binding epitopes using three epitope prediction algorithms. The median predicted epitope-binding affinity for each peptide was calculated and expressed as ‘median affinity value’ where affinity value = 1/IC50 × 100. Predicted epitopes are arrayed along the x-axis in alphabetical order based on their protein of origin. b, Unfiltered median affinity values for the four predicted H-2Db epitopes. c, Median affinity values of remaining two H-2Db epitopes after filtering. d, Tetramer staining of CD8+ TIL from tumour-bearing mice treated with anti-PD-1 using H-2Db tetramers loaded with top 4 H-2Db synthetic peptides. e, IFN-γ and TNF-α intracellular cytokine staining of CD8+ TIL from tumour-bearing mice treated with anti-PD-1 immunotherapy following co-culture with naive irradiated splenocytes pulsed with the top four H-2Db synthetic peptides added at 1 μM final concentration. Data are presented as per cent of CD8+ TIL positive for IFN-γ, TNF-α or both cytokines. Data are representative of two independent experiments.

Extended Data Figure 3 mLama4 and mAlg8 stimulate CD8+ T cell lines generated against d42m1-T3 following anti-PD-1 immunotherapy.

a, CD8+ T cell lines generated from splenocytes of individual d42m1-T3-tumour-bearing mice that rejected their tumours after anti-PD-1 therapy were incubated with irradiated d42m1-T3 tumour cells (or F244 tumour cells) treated with blocking mAb specific for H-2Kb, and/or H-2Db and IFN-γ production was quantitated. Data are presented as means ± s.e.m. and are representative of two independent experiments. Samples were compared using an unpaired, two-tailed Student’s t test (***P < 0.001). b, IFN-γ release by the CTL 74 T cell line following co-culture with naive irradiated splenocytes pulsed with the top 62 H-2Kb synthetic peptides added at 1 μM final concentration.

Extended Data Figure 4 mLama4 and mAlg8 bind H-2Kb and stimulate CD8+ T cell lines generated against d42m1-T3 following anti-PD-1 immunotherapy.

a, IFN-γ release by CTL 62, CTL 73 or CTL 74 T cell lines following stimulation with naive irradiated splenocytes pulsed with wild-type or mutant forms of Lama4 or Alg8 peptides. b, RMA-S cells were incubated with 8 amino acid peptides of mLama4 or mAlg8 and surface expression of H-2Kb or H-2Db was assessed by flow cytometry. Mean fluorescent intensity of H-2Kb and H-2Db was expressed as peptide binding score. Data presented are representative of at least two independent experiments.

Extended Data Figure 5 Identification of a peptide bound to H-2Kb on d42m1-T3 tumour cells corresponding to mutant Lama4.

a, Identification of the peptide, VGFNFRTL, corresponding to mLama4 by discovery mass spectrometry. b, Validation of the mLama4 peptide using an isotope-labelled synthetic peptide (VGFNFRTL (13C6, 15N1)).

Extended Data Figure 6 Generation of SRM assay library for the detection of mutant H-2Kb peptides on d42m1-T3.

a, SRM transitions were optimized for 51 of the 62 top predicted H-2Kb peptides. The 51 peptides chosen were selected based on having physiochemical properties that would allow their detection by mass spectrometry if present. Only Lama4 and Alg8 are shown here for simplicity. The 51 peptides were synthesized and LC-tandem mass spectrometry acquisition was performed on each peptide to determine the best collision energy and to obtain the full fragment ion spectrum (left panel); three to seven of the highest intensity peaks were selected to be built into SRM transitions. Optimal SRM transitions displayed as extracted ion chromatograms are shown (right panel). Q1–Q3 transitions are indicated in parenthesis. The mutated amino acid in the peptide sequence is marked in red. b, F244 tumour cells, which lack the mLama4 and mAlg8 d42m1-T3 epitope, lack detectable mLama4 or mAlg8 in complex with H-2Kb as assessed by SRM.

Extended Data Figure 7 CD8+ T cells specific for mutant forms of Lama4 and Alg8 infiltrate d42m1-T3, but not F244, tumours.

a, Detection of tumour-infiltrating mLama4- or mAlg8-specific T cells infiltrating d42m1-T3 or F244 tumours of mice treated with anti-PD-1. Tumours were harvested on day 12 post-transplant. Cells were gated on live CD45+ and CD8α+ tumour-infiltrating lymphocytes. Detection of mLama4- or mAlg8-specific T cells was achieved by staining with peptide-MHC H-2Kb PE-labelled tetramers. Data are representative of at least five independent experiments. b, Detection of mLama4-specific tumour-infiltrating T cells from tumour-bearing mice treated with anti-PD-1, anti-CTLA-4, both anti-PD-1 plus anti-CTLA-4 or control mAb. Detection of mLama4-specific T cells was achieved by staining with mLama4-MHC H-2Kb PE-labelled tetramers. Data presented are plotted as the mean mLama4 tetramer-positive as a percent of CD8α+ tumour-infiltrating cells and are representative of at least three independent experiments.

Extended Data Figure 8 mAlg8 and mLama4 SLP vaccine control d42m1-T3 tumour outgrowth when administered therapeutically or prophylactically.

a, Tumour growth of d42m1-T3 tumours from mice therapeutically vaccinated with mLama4 and mAlg8 SLP plus poly(I:C), HPV control SLP plus poly(I:C) or poly(I:C) alone. Data shown are mean ± s.e.m. Mutant Lama4 and mAlg8 SLP vaccine group was compared to HPV control SLP vaccine group using an unpaired, two-tailed Student’s t test (*P < 0.05 and **P < 0.01). b, Kaplan–Meier survival curves of d42m1-T3-tumour-bearing mice (7 per group) prophylactically vaccinated with SLP vaccines plus poly(I:C). mLama4 plus mAlg8 compared to HPV control: P = 0.0003 (log-rank (Mantel–Cox) test). Representative of two independent experiments. c, Cumulative number of mice (7–10 per group) from at least two independent experiments rejecting d42m1-T3 or F244 tumours as a consequence of SLP or minimal epitope peptide prophylactic vaccination.

Extended Data Figure 9 Detection of TIM-3, LAG-3, IFN-γ and TNF-α expression in tumour-infiltrating CD8+ T cells.

a, Representative histogram of TIM-3 or LAG-3 expression on mLama4-specific CD8+ tumour-infiltrating T cells from tumour-bearing mice treated with anti-PD-1, anti-CTLA-4, both anti-PD-1 and anti-CTLA-4 or control mAbs. b, TIM-3 and LAG-3 are reduced in mAlg8-specific CD8+ TIL from tumour-bearing mice treated with anti-PD-1, anti-CTLA-4, or both anti-PD-1 and anti-CTLA-4 compared to mice treated with control mAb. n = 5 mice per group pooled. Data are presented as mean ± s.e.m. of at least three independent experiments. Samples were compared using an unpaired, two-tailed Student’s t test (*P < 0.05, **P < 0.01). c, Representative dot plots of IFN-γ and TNF-α stained CD8+ tumour-infiltrating T cells from tumour-bearing mice following treatment with anti-PD-1, anti-CTLA-4, both anti-PD-1 and anti-CTLA-4 or control mAbs. Data presented are representative of at least three independent experiments.

Extended Data Table 1 Commonly differentially regulated genes and GSEA pathways in mLama4+ TIL during checkpoint blockade therapy

Supplementary information

Supplementary Data 1

This file contains MS traces for each of the mutant H-2Kb d42m1-T3 epitopes tested by targeted MS. (PDF 1891 kb)

Supplementary Information

This file contains Supplementary Tables 1-3. Supplementary Table 1 contains a complete list of d42m1-T3 H-2Kb-bound peptides identified by discovery MS. Supplementary Table 2 shows all differentially expressed genes in mLama4-tetramer-positive CD8+ TILs from mice treated with checkpoint blockade therapy compared to mLama4-tetramer-positive CD8+ TILs from control mice. Supplementary Table 3 shows differentially regulated pathways (GSEA pathway analysis) in mLama4-tetramer-positive CD8+ TILs from mice treated with checkpoint blockade therapy compared to mLama4-tetramer-positive CD8+ TILs from control mice. (PDF 3029 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gubin, M., Zhang, X., Schuster, H. et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515, 577–581 (2014). https://doi.org/10.1038/nature13988

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13988

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