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.

  • Analysis
  • Published:

Tissue-specific thresholds of mutation burden associated with anti-PD-1/L1 therapy benefit and prognosis in microsatellite-stable cancers

Abstract

Immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 or its ligand (PD-1/L1) have expanded the treatment landscape against cancers but are effective in only a subset of patients. Tumor mutation burden (TMB) is postulated to be a generic determinant of ICI-dependent tumor rejection. Here we describe the association between TMB and survival outcomes among microsatellite-stable cancers in a real-world clinicogenomic cohort consisting of 70,698 patients distributed across 27 histologies. TMB was associated with survival benefit or detriment depending on tissue and treatment context, with eight cancer types demonstrating a specific association between TMB and improved outcomes upon treatment with anti-PD-1/L1 therapies. Survival benefits were noted over a broad range of TMB cutoffs across cancer types, and a dose-dependent relationship between TMB and outcomes was observed in a subset of cancers. These results have implications for the use of cancer-agnostic and universal TMB cutoffs to guide the use of anti-PD-1/L1 therapies, and they underline the importance of tissue context in the development of ICI biomarkers.

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: Associations between TMB cutoffs and OS among ICI-treated and non-ICI-treated cohorts for different cancer types.
Fig. 2: Time on treatment and OS HRs of ICI-treated patients.
Fig. 3: Lowest absolute and percentile TMB thresholds with detectable survival benefit after treatment with ICIs.
Fig. 4: TME characteristics among TMB-H cancers across cancer types.

Similar content being viewed by others

Data availability

Data will be made available upon reasonable request with the permission of Caris Life Sciences. Raw sequencing data are owned by Caris Life Sciences and cannot be shared due to patient privacy and protected proprietary information. Access to aggregated data can be requested by contacting the corresponding author, including a brief description of the requirements and intended use. Requests will be discussed with the Caris data access team and a response given within 4 weeks. External datasets used in this study are available from the following public resources: gnomAD, gnomad.broadinstitute.org; International Genome Sample Resource (1000 Genomes Project), www.internationalgenome.org; dbSNP, www.ncbi.nlm.nih.gov/snp. Source data are provided with this paper.

References

  1. Bagchi, S., Yuan, R. & Engleman, E. G. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu. Rev. Pathol. 16, 223–249 (2021).

    Article  CAS  PubMed  Google Scholar 

  2. Robert, C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat. Commun. 11, 3801 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Vaddepally, R. K., Kharel, P., Pandey, R., Garje, R. & Chandra, A. B. Review of indications of FDA-approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers 12, 738 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lei, Y., Li, X., Huang, Q., Zheng, X. & Liu, M. Progress and challenges of predictive biomarkers for immune checkpoint blockade. Front. Oncol. 11, 617335 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wang, D. R., Wu, X. L. & Sun, Y. L. Therapeutic targets and biomarkers of tumor immunotherapy: response versus non-response. Signal Transduct. Target. Ther. 7, 331 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gunjur, A. et al. ‘Know thyself’—host factors influencing cancer response to immune checkpoint inhibitors. J. Pathol. 257, 513–525 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lybaert, L. et al. Challenges in neoantigen-directed therapeutics. Cancer Cell 41, 15–40 (2023).

    Article  CAS  PubMed  Google Scholar 

  8. Ganesan, S. & Mehnert, J. Biomarkers for response to immune checkpoint blockade. Annu. Rev. Cancer Biol. 4, 331–351 (2020).

    Article  Google Scholar 

  9. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  15. Segal, N. H. et al. Epitope landscape in breast and colorectal cancer. Cancer Res. 68, 889–892 (2008).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hugo, W. et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 165, 35–44 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Marabelle, A. et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 21, 1353–1365 (2020).

    Article  CAS  PubMed  Google Scholar 

  21. Samstein, R. M. et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 51, 202–206 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Valero, C. et al. The association between tumor mutational burden and prognosis is dependent on treatment context. Nat. Genet. 53, 11–15 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Valero, C. et al. Response rates to anti-PD-1 immunotherapy in microsatellite-stable solid tumors with 10 or more mutations per megabase. JAMA Oncol. 7, 739–743 (2021).

    Article  PubMed  Google Scholar 

  24. McGrail, D. J. et al. High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types. Ann. Oncol. 32, 661–672 (2021).

    Article  CAS  PubMed  Google Scholar 

  25. Merino, D. M. et al. Establishing guidelines to harmonize tumor mutational burden (TMB): in silico assessment of variation in TMB quantification across diagnostic platforms: phase I of the Friends of Cancer Research TMB Harmonization Project. J. Immunother. Cancer 8, e000147 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Offin, M. et al. Tumor mutation burden and efficacy of EGFR-tyrosine kinase inhibitors in patients with EGFR-mutant lung cancers. Clin. Cancer Res. 25, 1063–1069 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. McGranahan, N. & Swanton, C. Neoantigen quality, not quantity. Sci. Transl. Med. 11, eaax7918 (2019).

    Article  PubMed  Google Scholar 

  28. Sha, D. et al. Tumor mutational burden as a predictive biomarker in solid tumors. Cancer Discov. 10, 1808–1825 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Chowell, D. et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 359, 582–587 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  30. Havel, J. J., Chowell, D. & Chan, T. A. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat. Rev. Cancer 19, 133–150 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Doroshow, D. B. et al. PD-L1 as a biomarker of response to immune-checkpoint inhibitors. Nat. Rev. Clin. Oncol. 18, 345–362 (2021).

    Article  CAS  PubMed  Google Scholar 

  32. Davis, A. A. & Patel, V. G. The role of PD-L1 expression as a predictive biomarker: an analysis of all US Food and Drug Administration (FDA) approvals of immune checkpoint inhibitors. J. Immunother. Cancer 7, 278 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Li, F. et al. The association between CD8+ tumor-infiltrating lymphocytes and the clinical outcome of cancer immunotherapy: a systematic review and meta-analysis. EClinicalMedicine 41, 101134 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Ott, P. A. et al. T-cell-inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J. Clin. Oncol. 37, 318–327 (2019).

    Article  PubMed  Google Scholar 

  35. Finotello, F. et al. Molecular and pharmacological modulators of the tumor immune contexture revealed by deconvolution of RNA-seq data. Genome Med. 11, 34 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Bao, R., Stapor, D. & Luke, J. J. Molecular correlates and therapeutic targets in T cell-inflamed versus non-T cell-inflamed tumors across cancer types. Genome Med. 12, 90 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Marcus, L. et al. FDA approval summary: pembrolizumab for the treatment of tumor mutational burden-high solid tumors. Clin. Cancer Res. 27, 4685–4689 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Subbiah, V., Solit, D. B., Chan, T. A. & Kurzrock, R. The FDA approval of pembrolizumab for adult and pediatric patients with tumor mutational burden (TMB) ≥10: a decision centered on empowering patients and their physicians. Ann. Oncol. 31, 1115–1118 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. Prasad, V. & Addeo, A. The FDA approval of pembrolizumab for patients with TMB > 10 mut/Mb: was it a wise decision? No. Ann. Oncol. 31, 1112–1114 (2020).

    Article  CAS  PubMed  Google Scholar 

  40. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Jardim, D. L., Goodman, A., de Melo Gagliato, D. & Kurzrock, R. The challenges of tumor mutational burden as an immunotherapy biomarker. Cancer Cell 39, 154–173 (2021).

    Article  CAS  PubMed  Google Scholar 

  42. Gupta, R., Mehta, A. & Wajapeyee, N. Transcriptional determinants of cancer immunotherapy response and resistance. Trends Cancer 8, 404–415 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Nowicki, T. S., Hu-Lieskovan, S. & Ribas, A. Mechanisms of resistance to PD-1 and PD-L1 blockade. Cancer J. 24, 47–53 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Richman, L. P., Vonderheide, R. H. & Rech, A. J. Neoantigen dissimilarity to the self-proteome predicts immunogenicity and response to immune checkpoint blockade. Cell Syst. 9, 375–382 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Luksza, M. et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 551, 517–520 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wolf, Y. & Sameuls, Y. Neoantigens in cancer immunotherapy: quantity vs. quality. Mol. Oncol. 17, 1457–1459 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Kieffer, Y. et al. Single-cell analysis reveals fibroblast clusters linked to immunotherapy resistance in cancer. Cancer Discov. 10, 1330–1351 (2020).

    Article  CAS  PubMed  Google Scholar 

  48. Niknafs, N. et al. Persistent mutation burden drives sustained anti-tumor immune responses. Nat. Med. 29, 440–449 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

D.H. is supported by a Cancer Prevention and Research Institute of Texas Early Clinical Investigator Award (RP200549) and the Josephine Hughes Sterling Foundation. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the paper. D.S.B.H. is supported by the Dr. Miriam and Sheldon Adelson Medical Research Foundation. The remaining authors received no specific funding for this work.

Author information

Authors and Affiliations

Authors

Contributions

D.H. and H.Z. conceptualized the study. M.M., M.E., A.E., J.X. and D.H. performed data analyses. A.S., W.E.-D., E.S.A., S.L.G., M.J.H., H.B., D.S.B.H., S.V.L., P.C.M., R.R.M., T.W.-D., J.M., G.W.S., D.S., H.Z. and D.H. contributed to the assembly of the CARIS cohort. M.M. and D.H. drafted the paper, and all authors participated in the review and editing of the paper.

Corresponding author

Correspondence to David Hsiehchen.

Ethics declarations

Competing interests

A.E., J.X., G.W.S. and D.S. are employees of Caris Life Sciences. S.L.G. serves a paid consultant and advisor to Pfizer, Daiichi Sankyo, Eli Lilly, AstraZeneca, Genentech, SeaGen, Novartis and Menarini and has stock ownership in HCA Healthcare. E.S.A. serves as a paid consultant and advisor to Janssen, Astellas, Sanofi, Dendreon, Bayer, BMS, Amgen, Constellation, Blue Earth, Exact Sciences, Invitae, Curium, Pfizer, Merck, AstraZeneca, Clovis and Eli Lilly; has received research support (to his institution) from Janssen, J&J, Sanofi, BMS, Pfizer, AstraZeneca, Novartis, Curium, Constellation, Celgene, Merck, Bayer and Clovis; and is the co-inventor of a patented AR-V7 biomarker technology that has been licensed to Qiagen. The other authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Samra Turajlic and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 TMB cutoffs associated with ICI benefit retain predictive value across demographics.

Subset analyses of patients stratified by age groups and self-reported sex show that the association between TMB cutoffs and outcomes are similar across demographics. Forest plots depict hazard ratios (squares) and error bars indicate 95% confidence intervals.

Source data

Extended Data Fig. 2 Associations between overall survival and TMB at the 75th percentile for individual cancer types are independent of the sequencing platform used.

Hazard ratios of overall survival in the ICI cohort using a TMB threshold at the 75th percentile for individual cancer types were separately calculated for cases analyzed by the 592-gene panel or exome sequencing. Forest plots depict hazard ratios (squares) and error bars indicate 95% confidence intervals.

Source data

Extended Data Fig. 3 Immune correlates between TMB-high and TMB-low cancers using the earliest cutoff at which ICIs are predictive.

PD-L1 positive cell frequency, T-cell inflammatory score, and CD8 + T cell frequency are compared between TMB-high cancers and TMB-low cancers using the earliest cutoff at which ICIs are associated with OS benefit. Biomarkers enriched in TMB-high and TMB-low cancers using a false discovery rate of 0.05 are highlighted.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2.

Reporting Summary

Source data

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

Muquith, M., Espinoza, M., Elliott, A. et al. Tissue-specific thresholds of mutation burden associated with anti-PD-1/L1 therapy benefit and prognosis in microsatellite-stable cancers. Nat Cancer (2024). https://doi.org/10.1038/s43018-024-00752-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s43018-024-00752-x

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