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:

Neoantigen-targeted CD8+ T cell responses with PD-1 blockade therapy

Abstract

Neoantigens are peptides derived from non-synonymous mutations presented by human leukocyte antigens (HLAs), which are recognized by antitumour T cells1,2,3,4,5,6,7,8,9,10,11,12,13,14. The large HLA allele diversity and limiting clinical samples have restricted the study of the landscape of neoantigen-targeted T cell responses in patients over their treatment course. Here we applied recently developed technologies15,16,17 to capture neoantigen-specific T cells from blood and tumours from patients with metastatic melanoma with or without response to anti-programmed death receptor 1 (PD-1) immunotherapy. We generated personalized libraries of neoantigen–HLA capture reagents to single-cell isolate the T cells and clone their T cell receptors (neoTCRs). Multiple T cells with different neoTCR sequences (T cell clonotypes) recognized a limited number of mutations in samples from seven patients with long-lasting clinical responses. These neoTCR clonotypes were recurrently detected over time in the blood and tumour. Samples from four patients with no response to anti-PD-1 also demonstrated neoantigen-specific T cell responses in the blood and tumour to a restricted number of mutations with lower TCR polyclonality and were not recurrently detected in sequential samples. Reconstitution of the neoTCRs in donor T cells using non-viral CRISPR–Cas9 gene editing demonstrated specific recognition and cytotoxicity to patient-matched melanoma cell lines. Thus, effective anti-PD-1 immunotherapy is associated with the presence of polyclonal CD8+ T cells in the tumour and blood specific for a limited number of immunodominant mutations, which are recurrently recognized over time.

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

Fig. 1: Neoantigen-specific T cell screening and TCR clonotype identification.
Fig. 2: Neoantigen-specific T cell isolation from TILs and PBMCs in patients with a response to anti-PD-1 therapy.
Fig. 3: Antitumour activity of the neoantigen-specific TCRs isolated in patients with a response to anti-PD-1.
Fig. 4: Neoantigen-specific T cell isolation from TILs and PBMCs, and neoTCR antitumour activity in patients without a response to anti-PD-1.

Similar content being viewed by others

Data availability

Raw sequencing data derived from cell line and normal PBMC control samples collected under UCLA Institutional Review Board approvals 11-003254 have been deposited to dbGAP under accession number phs003153.v1.p1.

Code availability

The software used to analyse the data include: WA-MEM, Sentieon (v.201911.01), VarScan2 (v.2.3.9 and v.2.4.2), MuTect (v.1.1.7 and v.3.6), HISAT2 (v.2.0.4), STAR, RSEM, StringTie (v.1.2.2), Optitype (v.1.3.1), netMHCpan (v.3.4 and v.4.1), CWL (v.1.1), Strelka (v.2.9.9), Pindel (v.0.2.4b8), pVACtools (v.1.1.4; supported by NetMHCpan (v.4.0), NetMHC (v.4.0), NetMHCcons (v.1.1), PickPocket (v.1.1), SMM (v.1.0), SMMPMBEC (v.1.0), MHCflurry (v.1.2.2) and MHCnuggets (v.2.2)), Kallisto and MiXCR (v.2.1.3).

References

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

    Article  CAS  PubMed  ADS  Google Scholar 

  2. Tran, E., Robbins, P. F. & Rosenberg, S. A. Final common pathway’ of human cancer immunotherapy: targeting random somatic mutations. Nat. Immunol. 18, 255–262 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gros, A. et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med. 22, 433–438 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  5. 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  PubMed  Google Scholar 

  6. 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 

  7. 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  ADS  Google Scholar 

  8. McGranahan, N. et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351, 1463–1469 (2016).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  9. Verdegaal, E. M. et al. Neoantigen landscape dynamics during human melanoma-T cell interactions. Nature 536, 91–95 (2016).

    Article  CAS  PubMed  ADS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. Fehlings, M. et al. Late-differentiated effector neoantigen-specific CD8+ T cells are enriched in peripheral blood of non-small cell lung carcinoma patients responding to atezolizumab treatment. J. Immunother. Cancer 7, 249 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Caushi, J. X. et al. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature 596, 126–132 (2021).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  13. Oliveira, G. et al. Phenotype, specificity and avidity of antitumour CD8+ T cells in melanoma. Nature 596, 119–125 (2021).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  14. Parkhurst, M. R. et al. Unique neoantigens arise from somatic mutations in patients with gastrointestinal cancers. Cancer Discov. 9, 1022–1035 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Foy, S. P. et al. Non-viral precision T cell receptor replacement for personalized cell therapy. Nature https://doi.org/10.1038/s41586-022-05531-1 (2022).

  16. Chour, W. et al. Shared antigen-specific CD8+ T cell responses against the SARS-COV-2 spike protein in HLAA*02:01 COVID-19 Participants. Preprint at https://doi.org/10.1101/2020.05.04.20085779 (2020).

  17. Peng, S. et al. Sensitive detection and analysis of neoantigen-specific T cell populations from tumors and blood. Cell Rep. 28, 2728–2738 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Roth, T. L. et al. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature 559, 405–409 (2018).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  19. Oh, S. A. et al. High-efficiency nonviral CRISPR/Cas9-mediated gene editing of human T cells using plasmid donor DNA. J. Exp. Med. 219, e20211530 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Richters, M. M. et al. Best practices for bioinformatic characterization of neoantigens for clinical utility. Genome Med. 11, 56 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Hundal, J. et al. pVACtools: a computational toolkit to identify and visualize cancer neoantigens. Cancer Immunol. Res. 8, 409–420 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gee, M. H. et al. Antigen identification for orphan T cell receptors expressed on tumor-infiltrating lymphocytes. Cell 172, 549–563 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Yu, Y. Y., Netuschil, N., Lybarger, L., Connolly, J. M. & Hansen, T. H. Cutting edge: single-chain trimers of MHC class I molecules form stable structures that potently stimulate antigen-specific T cells and B cells. J. Immunol. 168, 3145–3149 (2002).

    Article  CAS  PubMed  Google Scholar 

  24. Krishna, S. et al. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science 370, 1328–1334 (2020).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  25. Duhen, T. et al. Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat. Commun. 9, 2724 (2018).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  26. Simoni, Y. et al. Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 557, 575–579 (2018).

    Article  CAS  PubMed  ADS  Google Scholar 

  27. Ribas, A. et al. SD-101 in combination with pembrolizumab in advanced melanoma: results of a phase Ib, multicenter study. Cancer Discov. 8, 1250–1257 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hugo, W. et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 168, 542 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  30. 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  PubMed  PubMed Central  ADS  Google Scholar 

  31. Boon, T., Coulie, P. G., Van den Eynde, B. J. & van der Bruggen, P. Human T cell responses against melanoma. Annu. Rev. Immunol. 24, 175–208 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Yewdell, J. W. Confronting complexity: real-world immunodominance in antiviral CD8+ T cell responses. Immunity 25, 533–543 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Lang, F., Schrörs, B., Löwer, M., Türeci, Ö. & Sahin, U. Identification of neoantigens for individualized therapeutic cancer vaccines. Nat. Rev. Drug Discov. 21, 261–282 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lowery, F. J. et al. Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science 375, 877–884 (2022).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  35. Prickett, T. D. et al. Durable complete response from metastatic melanoma after transfer of autologous T cells recognizing 10 mutated tumor antigens. Cancer Immunol. Res. 4, 669–678 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Scheper, W. et al. Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat. Med. 25, 89–94 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Wu, T. D. et al. Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature 579, 274–278 (2020).

    Article  CAS  PubMed  ADS  Google Scholar 

  38. Voabil, P. et al. An ex vivo tumor fragment platform to dissect response to PD-1 blockade in cancer. Nat. Med. 27, 1250–1261 (2021).

    Article  CAS  PubMed  Google Scholar 

  39. Yost, K. E. et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat. Med. 25, 1251–1259 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gros, A. et al. Recognition of human gastrointestinal cancer neoantigens by circulating PD-1+ lymphocytes. J. Clin. Invest. 129, 4992–5004 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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  PubMed  Google Scholar 

  42. Dudley, M. E., Wunderlich, J. R., Shelton, T. E., Even, J. & Rosenberg, S. A. Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J. Immunother. 26, 332–342 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. Preprint at https://arxiv.org/abs/1303.3997 (2013).

  44. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Koboldt, D. C. et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22, 568–576 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lai, Z. et al. VarDict: a novel and versatile variant caller for next-generation sequencing in cancer research. Nucleic Acids Res. 44, e108 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Szolek, A. et al. OptiType: precision HLA typing from next-generation sequencing data. Bioinformatics 30, 3310–3316 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Reynisson, B., Alvarez, B., Paul, S., Peters, B. & Nielsen, M. NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand data. Nucleic Acids Res. 48, W449–W454 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lybarger, L. et al. Enhanced immune presentation of a single-chain major histocompatibility complex class I molecule engineered to optimize linkage of a C-terminally extended peptide. J. Biol. Chem. 278, 27105–27111 (2003).

    Article  CAS  PubMed  Google Scholar 

  50. Bethune, M. T. et al. Peptide-MHC comPACTs. PACT Pharma. WO/2019/195310, PCT/US2019/025415 (2019).

  51. Peng, S. et al. Compositions and methods for identification of antigen specific T cells. PACT Pharma. WO2020167918A1, PCT/US2020/17887 (2020).

  52. Lefranc, M. P. et al. IMGT®, the international ImMunoGeneTics information system® 25 years on. Nucleic Acids Res. 43, D413–D422 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Bolotin, D. A. et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods 12, 380–381 (2015).

    Article  CAS  PubMed  Google Scholar 

  54. Bethune, M. T., Comin-Anduix, B., Hwang Fu, Y. H., Ribas, A. & Baltimore, D. Preparation of peptide-MHC and T-cell receptor dextramers by biotinylated dextran doping. Biotechniques 62, 123–130 (2017).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank B. Berent-Maoz and J. Pang for help in sample collections; J. M. Chen and J. Trent for administrative and logistics support; and Y. Qi, Y. Murtanu, L. Guo, M. Dhar, E. Huang and M. Wallace for their help in contributing suggestions and processing the samples. This study was funded in part by PACT Pharma, the Parker Institute for Cancer Immunotherapy (PICI) (to A.R. and to C.P.-S.), the Melanoma Research Alliance—Young Investigator Award (to C.P.-S.), NIH grants R35 CA197633 and P01 CA244118 (to A.R.), the Ressler Family Fund and contributions from K. and D. Schultz, T. and D. Jones, and T. Stutz (to A.R.). C.P.-S. is a Parker Senior Fellow supported by PICI. G.A.-R. was supported by the Isabel and Harvey Kibel Fellowship award and the Alan Ghitis Fellowship Award for Melanoma Research, and currently by a Parker Scholar award; and J.D.S. by the UCLA Tumour Immunology Training Grant (USHHS Ruth L. Kirschstein Institutional National Research Service Award T32 CA009120). Flow Cytometry was performed at the UCLA Jonsson Comprehensive Cancer Center (JCCC) Flow Cytometry Shared Resource that is supported by the National Institutes of Health award P30CA016042 and by the JCCC and the David Geffen School of Medicine at UCLA. Cell sorting was performed at the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research University of California, Los Angeles Flow Cytometry Core Resource. Sequencing studies were conducted at the UCLA Technology Center for Genomics & Bioinformatics.

Author information

Authors and Affiliations

Authors

Contributions

C.P.-S., A.F., S.J.M. and A.R. designed the study and provided overall guidance. C.P.-S. B.S., S.P., Z.P., K.J., O.D., C.L.W., A.F., S.J.M. and A.R. wrote the first version of the manuscript. All of the authors contributed to the final manuscript. C.P.-S., B.S., S.P., Z.P., K.J., O.D., C.L.W., A.F., S.J.M. and A.R. analysed the results. G.C., D.J.W., B.C. and A.R. contributed to patient treatment and care. C.P.-S., S.J., J.D.S., I.P.-G., A.V.-C., I.B.-C., G.C. and A.R. contributed to patient sample collection and processing. Z.P., H.X., Y.M., S.Z., E.M., J. Hundal., O.L.G. and M.G. contributed to bioinformatics analysis. M.Y., C.S., K.H., O.D. and M.T.B. performed the protein library synthesis. S.P., C.L.W., M.T.B., D.A., B.B.Q., B.Y., D.N., S. Said., K.S., J.G., J. Hoover., X.R.B., R.T., C.M., P.P., S.N., J.R.H., A.F. and S.J.M. contributed to the design and/or performance of the neoantigen-specific isolation experiments. K.J., W.L., D.N., S.C., M.J.P., R.M., S.P.F., T.H., A.F. and S.J.M. contributed to the design and/or performance of the gene-editing experiments. C.P.-S., B.S., B.P., A.C., M.T., S.J., J.D.S., A.F., S.J.M. and A.R. contributed to the design and/or performance of the T cell functional studies. G.A.-R., D.E.S. and A.G. provided guidance, samples and materials throughout the different stages of these studies.

Corresponding authors

Correspondence to Cristina Puig-Saus or Antoni Ribas.

Ethics declarations

Competing interests

B.S., S.P., C.L.W., Z.P., B.P., A.C., D.A., B.B.Q., B.Y., K.J., O.D., D.N., K.S., J.G., J. Hoover., S. Said., W.L., C.S., K.H., Y.M., S.C., M.J.P., M.T., R.T., C.M., P.P., S.N., S.P.F., T.H., M.Y., S. Sandoval., R.M., X.R.B., M.T.B., A.F. and S.J.M. are current or former employees of PACT Pharma, and hold stock in the company. S.P., M.T.B., J.R.H. and A.R. are co-founders of PACT Pharma, and hold founder stock. J.R.H. and A.R. are members of the scientific advisory board of PACT Pharma. J.R.H. and A.R. are members of the board of directors of PACT Pharma. A.R. has received honoraria from consulting with Amgen, Bristol-Myers Squibb, Chugai, Dynavax, Genentech, Merck, Nektar, Novartis, Roche and Sanofi; is or has been a member of the scientific advisory board and holds stock in Advaxis, Arcus Biosciences, Bioncotech Therapeutics, Compugen, CytomX, Five Prime, RAPT, ImaginAb, Isoplexis, Kite-Gilead, Lutris Pharma, Merus, Rgenix and Tango Therapeutics. C.P.-S. and A.R. are listed as inventors on and receive licensing revenue from a patent application covering the use of non-viral gene editing of T cells that was licensed by The Regents of the University of California to Arsenal Bio (South San Francisco; WO2019084552A1, application filed by The Regents Of The University Of California); the methods and technology described therein were not used in the experiments performed herein. A.G. reports receiving funding from Novartis, VCN Biosciences and Merck KGaA, has received speaker honoraria from Roche, and has consulted for Achilles Therapeutics, Neon Therapeutics, PACT Pharma and Oxford Immunotherapy. Patent applications have been filed on aspects of the described work by PACT Pharma, entitled ‘Peptide-MHC comPACTs’, ‘Compositions and Methods For Identification of Antigen Specific T Cells’ and ‘Primary Cell Gene Editing’. The other authors declare no competing interests.

Peer review

Peer review information

Nature thanks James Gulley, Naiyer Rizvi and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Neoantigen-specific T-cell isolation and TCR clonotype identification. Summary of key parameters from the longitudinal landscape analysis of neoantigen-specific T cells in patients with and without response to therapy.

a, Total number of non-synonymous mutations. b, Total number of mutations screened. c, Total number of predicted neoantigen–HLA complexes screened. c, Total number of mutations targeted. e, Total number of neoantigen-specific TCR clonotypes isolated. f, Ratio of the number of neoantigen specific TCR clonotypes isolated per mutation targeted in each patient. Mean ± SD and individual values are plotted. (n) indicates the number of different patients, n = 7 for responders and n = 4 for non-responders. * p < 0.05, two-tailed unpaired t test, using the Two-stage linear step-up procedure of Benjamini, Krieger and Yekutieli, with Q = 1%. p = 0.0190 for TCR clonotypes and p = 0.0434 for TCR clonotype/mutation targeted.

Extended Data Fig. 2 Neoantigen-specific T-cell isolation from PBMCs in patients with response to anti-PD-1 therapy.

a, Landscape analysis of the neoantigen-specific T cells over time in patient 3. Bottom panel shows mRNA expression and predicted HLA binding affinity of the putative neoantigens screened. Neoantigens targeted by T cells are highlighted in different colours. The same colour code is used in the top panels to show the neoantigen specificity of the isolated T cells. The top panels show the evolution over time of the neoantigen-specific T cells in PBMCs. Each box represents one isolated T cell, each cross is equivalent to ten isolated T cells, and each circle is equivalent to 100 isolated T cells. Each colour represents a different neoantigen-specific T-cell clonotype. The TCR ID is also plotted. The number of isolated T cells is normalized to 100,000 CD8+ T cells using a round up method to plot the data. The mutated gene name, the point mutation, the sequence of the neoantigen, and the HLA are indicated on top of the figure. The T cell clonotypes shown have not been validated by expression in healthy donor T cells and binding to neoantigen–HLA complexes. b, Same as a for patient 4. c, Same as a for patient 5. d, Same as a for patient 7.

Extended Data Fig. 3 Captured neoTCR specific neoantigen–HLA binding validation.

After capture of the neoantigen-specific T cells, the cognate TCR is sequenced, and the sequence used to gene edit healthy donor T cells replacing the endogenous TCR by the neoTCR. The neoTCR specificity and stability are validated by multimer staining of the gene-edited T cells. Only validated TCRs are shown. a-f. Multimer staining of the gene edited T-cell products gated on live cells from patient 1 or Live/CD45+ cells from patients 2, 6, 9, 10, 11 (n = 1 for patient 1, n = 2 for patient 6, 9 and 11, and n = 3 for patients 2 and 10). Mean ± SD and individual values are plotted. (n) indicates the number of technical replicates.

Extended Data Fig. 4 Cytotoxicity induced by neoantigen-specific TCRs from patient 1 upon co-culture with the autologous cell line.

Healthy donor T cells genetically engineered to express the captured neoTCRs from patient 1 were co-cultured with the autologous (M495) or a mismatched cell line (M202). a-b, Specific target-cell killing by neoTCR gene-edited T cells of the autologous cell line and the mismatched control (P:T ratio 5:1, n = 4)). Melanoma cell lines were pre-treated with media with IFNγ 24 h prior co-culture with T cells. The plots are divided between TCRs without killing activity (a) and TCRs with intermediate killing (b). * p < 0.05 vs Neo12, two-tailed unpaired t test with Holm-Sidak adjustment for multiple comparison (n) indicates the number of biological replicates. Exact p-values provided in Supplementary Information. Mean and individual values are shown. All T-cell products contain CD8+ and CD4+ gene-edited T cells.

Extended Data Fig. 5 Titration of cytokine secretion by neoantigen-specific TCRs from patient 1 upon binding to specific neoantigen–HLA multimer.

Healthy donor T cells genetically engineered to express the captured neoTCRs from patient 1 were incubated with increasing amounts of plate-bound neoantigen–HLA multimer specific for each TCR and, 24 h after incubation, the IFNγ, IL-2, and TNF secreted were measured. a, NeoTCRs targeting UBE2J1 mutation presented by HLA-A*24:02. b, NeoTCRs targeting NUP188 mutation presented by HLA-A*24:02. c, NeoTCRs targeting WDR1 mutation presented by HLA-A*03:01. d, NeoTCRs targeting NRP1 mutation presented by HLA-A*03:01. e, NeoTCRs targeting TRAPPC10 mutation presented by HLA-C*04:01. f, NeoTCRs targeting IFNLR1 mutation presented by HLA-A*03:01. g, NeoTCRs targeting PLA2G4A mutation presented by HLA-A*03:01. h, NeoTCRs targeting SLC6A3 mutation presented by HLA-A*03:01. i, NeoTCRs targeting GSDMB mutation presented by HLA-C*12:02. j, Summary EC50 calculated for each TCR and each cytokine. (n = 2), (n) indicates the number of biological replicates. Mean and individual values are shown. All T-cell products contain CD8+ and CD4+ gene-edited T cells.

Extended Data Fig. 6 Activation, cytotoxicity, cytokine secretion, and proliferation induced by neoantigen-specific TCRs from patient 2 upon co-culture with the autologous cell line.

Healthy donor T cells genetically engineered to express the captured neoTCRs from patient 2 were co-cultured with the autologous (M489) or a mismatched cell line (M202). a, 4-1BB, OX-40, and CD107a upregulation in the CD8+ neoTCR+ T cells after co-culture. Melanoma cell lines were pre-treated with regular media or media with IFNγ 24 h prior co-culture with T cells (n = 3). b, percentage of tumour growth inhibition in M489 autologous cell line compared to the cell growth in media alone at 24, 48, 72 and 96 h (n = 4). c, Cytokine release at 24 h after co-culture (n = 3). d, Proliferation of CD8+ neoTCR+ T cells measured by Ki67 mean fluorescence intensity upon 24, 48 and 72 h co-culture with autologous melanoma cell line (M489, top panel) or a mismatched cell line (M202, bottom panel) (n = 3). * p < 0.05, ** p < 0.005, ***p < 0.0005, ****p < 0.0001 vs Neo12, two-tailed unpaired t test with Holm-Sidak adjustment for multiple comparisons in figure a, b and c. * p < 0.05, ** p < 0.005, ***p < 0.0005, ****p < 0.0001 vs M202, two-tailed unpaired t test with Holm-Sidak adjustment for multiple comparisons in figure d. Exact p-values provided in Supplementary Information. (n) indicates the number of biological replicates. Mean ± SD and individual values are shown. All T-cell products contain CD8+ and CD4+ gene-edited T cells.

Extended Data Fig. 7 Function of the CD8-independent TCRs in gene-edited CD4+ T cells.

a,c,e,g,i. Percentage of CD8+ and CD4+ in the neoTCR+ T cell population. n = 2 for patients 6, 9 and 11, and n = 3 for patients 2 and 10). CD8-dependent TCRs, described as TCRs that require CD8 co-receptor engagement to bind to the MHC-peptide, are marked with a red arrow. b,d,f,h, Percentage of OX-40+ cells in the NeoTCR+CD4+ T cells. (n = 3) *p < 0.05, **p < 0.005, ***p < 0.0005,****p < 0.0001 vs Neo12, two-tailed unpaired t test with Holm-Sidak adjustment for multiple comparison in figures b, d, and h. The same test without adjustment for multiple comparisons was used in figure f. Exact p-values provided in Supplementary Information. (n) indicates the number of biological replicates. Mean ± SD are shown. All T-cell products contain CD8+ and CD4+ gene-edited T cells.

Extended Data Fig. 8 Activation, cytotoxicity, cytokine secretion, and proliferation induced by neoantigen-specific TCRs from patient 6 upon co-culture with the autologous cell line.

Healthy donor T cells genetically engineered to express the captured neoTCRs from patient 6 were co-cultured with the autologous (M490) or a mismatched cell line (M202). a, 4-1BB and OX-40 upregulation in the CD8+ neoTCR+ T cells after co-culture. Melanoma cell lines were pre-treated with regular media or media with IFNγ 24 h prior co-culture with T cells (n = 3). b, Specific target-cell killing in the autologous cell line (top panel) or a mismatched cell line (bottom panel), (P:T ratio 10:1, n = 4). c, Cytokine release at 24 h after co-culture (n = 3). Melanoma cell lines were pre-treated with IFNγ for 24 h before co-culture with T cells. d, Proliferation of CD8+ neoTCR+ T cells measured by Ki67 mean fluorescence intensity upon 24, 48 and 72 h co-culture with autologous melanoma cell line (M490, top panel) or a mismatched cell line (M202, bottom panel). Melanoma cell lines were pre-treated with IFNγ 24 h prior co-culture with T cells (n = 3). *p < 0.05, **p < 0.005, ***p < 0.0005, ****p < 0.0001 vs Neo12, two-tailed unpaired t test with Holm-Sidak adjustment for multiple comparisons in figure a, b and c. *p < 0.05, **p < 0.005, ***p < 0.0005, ****p < 0.0001 vs M202, two-tailed unpaired t test with Holm-Sidak adjustment for multiple comparisons in figure d. (n) indicates the number of biological replicates. Exact p-values provided in Supplementary Information. Mean ± SD and individual values are shown in a, c and d. Mean and individual values are shown in b. All T-cell products contain CD8+ and CD4+ gene-edited T cells.

Extended Data Fig. 9 Neoantigen-specific T-cell isolation from PBMCs in patients without response to anti-PD-1 therapy.

a, Landscape analysis of the neoantigen-specific T cells over time in patient 8. Bottom panel shows mRNA expression and predicted HLA binding affinity of the predicted neoantigens screened. Neoantigens targeted by T cells are highlighted in different colours. The same colour code is used in the top panels to show the neoantigen specificity of the isolated T cells. The top panels show the evolution over time of the neoantigen-specific T cells in PBMCs. Each box represents one isolated T cell, each cross is equivalent to ten isolated T cells, and each circle is equivalent to 100 isolated T cells. Each colour represents a different neoantigen-specific T-cell clonotype. The number of isolated T cells is normalized to 100,000 CD8+ T cells using a round up method to plot the data. The mutated gene name, the point mutation, the sequence of the neoantigen, and the HLA are indicated on top of the figure. The T cell clonotypes shown have not been validated by expression in healthy donor T cells and binding to neoantigen–HLA complexes.

Extended Data Fig. 10 Activation, cytokine secretion, and proliferation induced by neoantigen-specific TCRs from patients without response to anti-PD-1 upon co-culture with the autologous cell lines.

Healthy donor T cells genetically engineered to express the captured neoTCRs from patient9 (a), 10 (b-d) and 11 (e-f) were co-cultured with the autologous (M488, M485 and M486 respectively) or a mismatched cell line (M202). a, 4-1BB upregulation in the CD8+ neoTCR+ T cells from patient 9 after co-culture. Melanoma cell lines were pre-treated with regular media or media with IFNγ 24 h prior co-culture with T cells (n = 3). b, 4-1BB, OX-40, and CD107a upregulation in CD8+ neoTCR+ T cells from patient 10 after co-culture. Melanoma cell lines were pre-treated with regular media or media with IFNγ 24 h prior co-culture with T cells (n = 3). c, Cytokine release at 24 h after co-culture (n = 3). d, Proliferation of CD8+ neoTCR+ T cells from patient 10 measured by Ki67 mean fluorescence intensity upon 24, 48 and 72 h co-culture with autologous melanoma cell line (M485, top panel) or a mismatched cell line (M202, bottom panel) (n = 3). e, 4-1BB and OX-40 upregulation in CD8+ neoTCR+ T cells from patient 11 after co-culture. Melanoma cell lines were pre-treated with regular media or media with IFNγ 24 h prior co-culture with T cells (n = 3). f, Cytokine release at 24 h after co-culture (n = 3). * p < 0.05, ** p < 0.005, ***p < 0.0005, ****p < 0.0001 vs Neo12, two-tailed unpaired t test with Holm-Sidak adjustment for multiple comparisons in figure a, b, c, e, and f. * p < 0.05, ** p < 0.005, ***p < 0.0005, ****p < 0.0001 vs M202, two-tailed unpaired t test with Holm-Sidak adjustment for multiple comparisons in figure d. Exact p-values provided in Supplementary Information. (n) indicates the number of biological replicates. Mean ± SD and individual values are shown. All T-cell products contain CD8+ and CD4+ gene-edited T cells.

Extended Data Table 1 Patient Characteristics

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2, Supplementary Tables 1 and 2 (Supplementary Data) and statistical data tables (P values).

Reporting Summary

Peer Review File

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Puig-Saus, C., Sennino, B., Peng, S. et al. Neoantigen-targeted CD8+ T cell responses with PD-1 blockade therapy. Nature 615, 697–704 (2023). https://doi.org/10.1038/s41586-023-05787-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-05787-1

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: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer