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:

Improved prediction of immune checkpoint blockade efficacy across multiple cancer types

Abstract

Only a fraction of patients with cancer respond to immune checkpoint blockade (ICB) treatment, but current decision-making procedures have limited accuracy. In this study, we developed a machine learning model to predict ICB response by integrating genomic, molecular, demographic and clinical data from a comprehensively curated cohort (MSK-IMPACT) with 1,479 patients treated with ICB across 16 different cancer types. In a retrospective analysis, the model achieved high sensitivity and specificity in predicting clinical response to immunotherapy and predicted both overall survival and progression-free survival in the test data across different cancer types. Our model significantly outperformed predictions based on tumor mutational burden, which was recently approved by the U.S. Food and Drug Administration for this purpose1. Additionally, the model provides quantitative assessments of the model features that are most salient for the predictions. We anticipate that this approach will substantially improve clinical decision-making in immunotherapy and inform future interventions.

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: Overview of development of the model for integrated clinical-genetic prediction of ICB response.
Fig. 2: Model performance across multiple cancer types in the test set.
Fig. 3: Model predicts OS and PFS across multiple cancer types in the test set.

Similar content being viewed by others

Data availability

All de-identified data needed to replicate all analyses are in Supplementary Table 3 and are available online at https://www.ioexplorer.org.

Code availability

The code used in this study is deposited at https://github.com/CCF-ChanLab/MSK-IMPACT-IO.

References

  1. 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 

  2. Topalian, S. L., Taube, J. M., Anders, R. A. & Pardoll, D. M. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat. Rev. Cancer 16, 275–287 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bendell, J. et al. Efficacy and safety results from IMblaze370, a randomised phase III study comparing atezolizumab plus cobimetinib and atezolizumab monotherapy vs regorafenib in chemotherapy-refractory metastatic colorectal cancer. Ann. Oncol. 29, 123–123 (2018).

    Article  Google Scholar 

  4. Carbone, D. P. et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N. Engl. J. Med. 376, 2415–2426 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cohen, E. E. et al. Pembrolizumab (pembro) vs standard of care (SOC) for recurrent or metastatic head and neck squamous cell carcinoma (R/M HNSCC): phase 3 KEYNOTE-040 trial. Ann. Oncol. 28 https://oncologypro.esmo.org/meeting-resources/esmo-2017-congress/Pembrolizumab-pembro-vs-standard-of-care-SOC-for-recurrent-or-metastatic-head-and-neck-squamous-cell-carcinoma-R-M-HNSCC-Phase-3-KEYNOTE-040-trial (2017).

  6. Powles, T. et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet 391, 748–757 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. 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 

  8. Keenan, T. E., Burke, K. P. & Van Allen, E. M. Genomic correlates of response to immune checkpoint blockade. Nat. Med. 25, 389–402 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Anagnostou, V. et al. Multimodal genomic features predict outcome of immune checkpoint blockade in non-small-cell lung cancer. Nat. Cancer 1, 99–111 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Topol, E. J. High-performance medicine: the convergence of human and artificial intelligence. Nat. Med. 25, 44–56 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. Rajkomar, A., Dean, J. & Kohane, I. Machine learning in medicine. N. Engl. J. Med. 380, 1347–1358 (2019).

    Article  PubMed  Google Scholar 

  12. Eisenhauer, E. A. et al. New response evaluation criteria in solid tumors: RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Zehir, A. et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 23, 703–713 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Breiman, L. Random forests. Mach. Learn. 45, 5–32 (2001).

    Article  Google Scholar 

  15. 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  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  CAS  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  CAS  PubMed  PubMed Central  Google Scholar 

  18. Riaz, N. et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell 171, 934–949 (2017).

  19. Goodman, A. M. et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol. Cancer Ther. 16, 2598–2608 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Samstein, R. M. et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 51, 202–20 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    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. Davoli, T., Uno, H., Wooten, E. C. & Elledge, S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 355, eaaf8399 (2017).

  24. Chowell, D. et al. Evolutionary divergence of HLA class I genotype impacts efficacy of cancer immunotherapy. Nat. Med. 25, 1715–1720 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  26. Mandal, R. et al. Genetic diversity of tumors with mismatch repair deficiency influences anti-PD-1 immunotherapy response. Science 364, 485–491 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

  28. Wang, Z. et al. Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade. Nat. Med. 25, 141–151 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Sanchez, A. et al. Transcriptomic signatures related to the obesity paradox in patients with clear cell renal cell carcinoma: a cohort study. Lancet Oncol. 21, 283–293 (2020).

    Article  CAS  PubMed  Google Scholar 

  30. Conforti, F. et al. Cancer immunotherapy efficacy and patients’ sex: a systematic review and meta-analysis. Lancet Oncol. 19, 737–746 (2018).

    Article  CAS  PubMed  Google Scholar 

  31. Jaillon, S. et al. Neutrophil diversity and plasticity in tumour progression and therapy. Nat. Rev. Cancer 20, 485–503 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Li, M. J. et al. Change in neutrophil to lymphocyte ratio during immunotherapy treatment is a non-linear predictor of patient outcomes in advanced cancers. J. Cancer Res. Clin. 145, 2541–2546 (2019).

    Article  CAS  Google Scholar 

  33. Valero, C. et al. Pretreatment neutrophil-to-lymphocyte ratio and mutational burden as biomarkers of tumor response to immune checkpoint inhibitors. Nat. Commun. 12, 729 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kuai, J., Yang, F., Li, G. J., Fang, X. J. & Gao, B. Q. In vitro-activated tumor-specific T lymphocytes prolong the survival of patients with advanced gastric cancer: a retrospective cohort study. Onco Targets Ther. 9, 3763–3770 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Ikeguchi, A., Machiorlatti, M. & Vesely, S. K. Disparity in outcomes of melanoma adjuvant immunotherapy by demographic profile. Melanoma Manag 7, MMT43 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Jurasz, P., Alonso-Escolano, D. & Radomski, M. W. Platelet–cancer interactions: mechanisms and pharmacology of tumour cell-induced platelet aggregation. Br. J. Pharmacol. 143, 819–826 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gupta, D. & Lis, C. G. Pretreatment serum albumin as a predictor of cancer survival: a systematic review of the epidemiological literature. Nutr. J. 9, 69 (2010).

  38. Caro, J. J., Salas, M., Ward, A. & Goss, G. Anemia as an independent prognostic factor for survival in patients with cancer—a systematic, quantitative review. Cancer 91, 2214–2221 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Peng, D. et al. Prognostic significance of HALP (hemoglobin, albumin, lymphocyte and platelet) in patients with bladder cancer after radical cystectomy. Sci Rep. 8, 794 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bindea, G., Mlecnik, B., Fridman, W. H., Pages, F. & Galon, J. Natural immunity to cancer in humans. Curr. Opin. Immunol. 22, 215–222 (2010).

    Article  CAS  PubMed  Google Scholar 

  42. Harrell, F. E. Jr., Lee, K. L. & Mark, D. B. Multivariable prognostic models: issues in developing models, evaluating assumptions and adequacy, and measuring and reducing errors. Stat. Med. 15, 361–387 (1996).

    Article  PubMed  Google Scholar 

  43. Steyerberg, E. W. et al. Assessing the performance of prediction models: a framework for traditional and novel measures. Epidemiology 21, 128–138 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Gurjao, C., Tsukrov, D., Imakaev, M., Luquette, L. J. & Mirny, L. A. Limited evidence of tumour mutational burden as a biomarker of response to immunotherapy. Preprint at https://www.biorxiv.org/content/10.1101/2020.09.03.260265v2 (2020).

  45. Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 24, 541–550 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Krishna, C. et al. Single-cell sequencing links multiregional immune landscapes and tissue-resident T cells in ccRCC to tumor topology and therapy efficacy. Cancer Cell 39, 662–677 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. 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  Google Scholar 

  48. Maier, B. et al. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 580, 257–262 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97–103 (2018).

    Article  CAS  PubMed  Google Scholar 

  51. Krishna, C., Chowell, D., Gonen, M., Elhanati, Y. & Chan, T. A. Genetic and environmental determinants of human TCR repertoire diversity. Immun. Ageing 17, 26 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Braun, D. A. et al. Interplay of somatic alterations and immune infiltration modulates response to PD-1 blockade in advanced clear cell renal cell carcinoma. Nat. Med. 26, 909–918 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Miao, D. et al. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat. Genet. 50, 1271–1281 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Patel, S. J. et al. Identification of essential genes for cancer immunotherapy. Nature 548, 537–542 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zaretsky, J. M. et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 375, 819–829 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Skoulidis, F. et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 8, 822–835 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Samstein, R. M. et al. Mutations in BRCA1 and BRCA2 differentially affect the tumor microenvironment and response to checkpoint blockade immunotherapy. Nat. Cancer 1, 1188–1203 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wang, F. et al. Evaluation of POLE and POLD1 mutations as biomarkers for immunotherapy outcomes across multiple cancer types. JAMA Oncol. 5, 1504–1506 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Amin, M. B. et al. The Eighth Edition AJCC Cancer Staging Manual: continuing to build a bridge from a population-based to a more ‘personalized’ approach to cancer staging. CA Cancer J. Clin. 67, 93–99 (2017).

    Article  PubMed  Google Scholar 

  60. Zhou, J. et al. Analysis of tumor genomic pathway alterations using broad-panel next-generation sequencing in surgically resected lung adenocarcinoma. Clin. Cancer Res. 25, 7475–7484 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Shen, R. L. & Seshan, V. E. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 44, e131 (2016).

  62. Niu, B. F. et al. MSIsensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics 30, 1015–1016 (2014).

    Article  CAS  PubMed  Google Scholar 

  63. Shukla, S. A. et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat. Biotechnol. 33, 1152–1158 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Pierini, F. & Lenz, T. L. Divergent allele advantage at human MHC genes: signatures of past and ongoing selection. Mol. Biol. Evol. 35, 2145–2158 (2018).

  66. Robinson, J. et al. The IPD and IMGT/HLA database: allele variant databases. Nucleic Acids Res. 43, D423–D431 (2015).

    Article  CAS  PubMed  Google Scholar 

  67. Zerbino, D. R. et al. Ensembl 2018. Nucleic Acids Res. 46, D754–D761 (2018).

    Article  CAS  PubMed  Google Scholar 

  68. Grantham, R. Amino acid difference formula to help explain protein evolution. Science 185, 862–864 (1974).

    Article  CAS  PubMed  Google Scholar 

  69. Edgar, R. C. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 32, 1792–1797 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Pedregosa, F. et al. Scikit-learn: machine learning in Python. J. Mach. Learn. Res. 12, 2825–2830 (2011).

    Google Scholar 

  71. Saito, T. & Rehmsmeier, M. Precrec: fast and accurate precision-recall and ROC curve calculations in R. Bioinformatics 33, 145–147 (2017).

    Article  CAS  PubMed  Google Scholar 

  72. Robin, X. et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics.12, 77 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Schroder, M. S., Culhane, A. C., Quackenbush, J. & Haibe-Kains, B. survcomp: an R/Bioconductor package for performance assessment and comparison of survival models. Bioinformatics 27, 3206–3208 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Mogensen, U. B., Ishwaran, H. & Gerds, T. A. Evaluating random forests for survival analysis using prediction error curves. J. Stat. Softw. 50, 1–23 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Chan lab and members of the Immunogenomics and Precision Oncology Platform for advice and input. This work was supported, in part, by NIH R35 CA232097 (T.A.C.), NIH RO1 CA205426 (T.A.C.), the PaineWebber Chair (T.A.C.), NIH/NCI Cancer Center Support Grant (P30 CA008748), Fundación Alfonso Martín Escudero (C.V.), NIH K08 DE024774, NIH R01 DE027738, the Sebastian Nativo Fund and the Jayme and Peter Flowers Fund (to L.G.T.M.).

Author information

Authors and Affiliations

Authors

Contributions

D.C., S.-K.Y., C.V., A.P., L.G.T.M., N.W. and T.A.C conceived and designed the study. D.C. and S.-K.Y. developed the machine learning model. D.C., S.-K.Y., C.V., A.P., C.K., M.L., D.H., H.S., D.W.K., N.P., V.M., K.W., T.L., R.M.S., N.R., P.S.A., V.P.B., G.P., A.A.H., A.N.S., M.A.P., R.J.M, M.L., A.Z., M.F.B., L.G.T.M. and N.W. acquired, analyzed or interpreted the data. M.G. provided statistical advice. All authors critically revised the manuscript for important intellectual content. L.G.T.M., N.W. and T.A.C. supervised the study.

Corresponding authors

Correspondence to Luc G. T. Morris, Nils Weinhold or Timothy A. Chan.

Ethics declarations

Competing interests

T.A.C. is a co-founder of Gritstone Oncology and holds equity. T.A.C. holds equity in An2H. T.A.C. acknowledges grant funding from Bristol Myers Squibb, AstraZeneca, Illumina, Pfizer, An2H and Eisai. T.A.C. has served as an advisor for Bristol Myers, MedImmune, Squibb, Illumina, Eisai, AstraZeneca and An2H. T.A.C., L.G.T.M. and D.C. hold ownership of intellectual property on using tumor mutational burden to predict immunotherapy response, with a pending patent, which has been licensed to PGDx. M.A.P. reports consulting fees from Bristol Myers Squibb, Merck, Array BioPharma, Novartis, Incyte, NewLink Genetics, Aduro and Eisai; honoraria from Bristol Myers Squibb and Merck; and institutional support from RGenix, Infinity, Bristol Myers Squibb, Merck, Array BioPharma, Novartis and AstraZeneca. M.L. has received advisory board compensation from Merck and Bristol Myers Squibb. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Biotechnology thanks Victor Velculescu and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Supplementary information

Supplementary Information

Supplementary Figs. 1–18 and Supplementary Tables 1–3

Reporting Summary

Supplementary Table

Datasets

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chowell, D., Yoo, SK., Valero, C. et al. Improved prediction of immune checkpoint blockade efficacy across multiple cancer types. Nat Biotechnol 40, 499–506 (2022). https://doi.org/10.1038/s41587-021-01070-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-021-01070-8

This article is cited by

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