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:

Netie: inferring the evolution of neoantigen–T cell interactions in tumors

Abstract

Neoantigens are the key targets of antitumor immune responses from cytotoxic T cells and play a critical role in affecting tumor progressions and immunotherapy treatment responses. However, little is known about how the interaction between neoantigens and T cells ultimately affects the evolution of cancerous masses. Here, we develop a hierarchical Bayesian model, named neoantigen–T cell interaction estimation (netie) to infer the history of neoantigen-CD8+ T cell interactions in tumors. Netie was systematically validated and applied to examine the molecular patterns of 3,219 tumors, compiled from a panel of 18 cancer types. We showed that tumors with an increase in immune selection pressure over time are associated with T cells that have an activation-related expression signature. We also identified a subset of exhausted cytotoxic T cells postimmunotherapy associated with tumor clones that newly arise after treatment. These analyses demonstrate how netie enables the interrogation of the relationship between individual neoantigen repertoires and the tumor molecular profiles. We found that a T cell inflammation gene expression profile (TIGEP) is more predictive of patient outcomes in the tumors with an increase in immune pressure over time, which reveals a curious synergy between T cells and neoantigen distributions. Overall, we provide a new tool that is capable of revealing the imprints left by neoantigens during each tumor’s developmental process and of predicting how tumors will progress under further pressure of the host’s immune system.

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: The rationale for inferring the history of anti-neoantigen immune pressure during tumor development.
Fig. 2: Immune selection pressure variations correlate with the phenotypes of the tumor and tumor clones.
Fig. 3: Netie is capable of performing multi-sample joint analyses.
Fig. 4: History of immune selection pressure is predictive of tumor prognosis and response to immunotherapy.

Similar content being viewed by others

Data availability

The baseline patients come from TCGA and the patients with kidney cancer from our previous study21. The TCGA data (expression, mutation and survival) were downloaded from the TCGA firehose website (https://gdac.broadinstitute.org/,version of 2016012800.0.0). TCGA HLA typing data were provided by Thorsson et al.55. For the treatment cohorts, we gathered 32 patients with gastric cancer from Cristescu et al.42 and Kim et al.45; eight patients with NSCLC from Anagnostou et al.46; 105 patients with HNSCC from Cristescu et al.42; 352 patients with melanoma from Cristescu et al.42, Hugo et al.5, Liu et al.47, Riaz et al.6 and Van Allen et al.48; and 157 patients with RCC from IMmotion150 cohort49 and Miao et al.7. Information on access to data is provided in these original reports. The in-house MDACC patients’ raw genomic data have been uploaded into the European Genome-Phenome Archive (EGA) with the accession numbers EGAD00001008382 and EGAD00001008482. The accession codes and links for each dataset are listed in Supplementary Table 4. Source data are provided with this paper.

Code availability

The netie R package is available at https://github.com/tianshilu/Netie with Apache license version v.2.0.

References

  1. Lu, T. et al. Tumor neoantigenicity assessment with CSiN score incorporates clonality and immunogenicity to predict immunotherapy outcomes. Sci. Immunol. 21, eaaz3199 (2020).

  2. Hsiehchen, D. et al. DNA repair gene mutations as predictors of immune checkpoint inhibitor response beyond tumor mutation burden. Cell Rep. Med. 1, 100034 (2020).

  3. Chung, A. S. et al. Immune checkpoint inhibition is safe and effective for liver cancer prevention in a mouse model of hepatocellular carcinoma. Cancer Prev. Res. 13, 911–922 (2020).

    Article  CAS  Google Scholar 

  4. Lu, T. et al. Deep learning-based prediction of the T cell receptor-antigen binding specificity. Nat. Mach. Intell. 3, 864–875 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. Riaz, N. et al. Tumor and microenvironment evolution during immunotherapy with Nivolumab. Cell 171, 934–949.e16 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Miao, D. et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science 359, 801–806 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hellmann, M. D. et al. Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell. 33, 843–852.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Matsushita, H. et al. Neoantigen load, antigen presentation machinery, and immune signatures determine prognosis in clear cell renal cell carcinoma. Cancer Immunol. Res. 4, 463–471 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Miller, A. et al. High somatic mutation and neoantigen burden are correlated with decreased progression-free survival in multiple myeloma. Blood Cancer J. 7, e612 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Matsushita, H. et al. The frequency of neoantigens per somatic mutation rather than overall mutational load or number of predicted neoantigens per se is a prognostic factor in ovarian clear cell carcinoma. Oncoimmunology 6, e1338996 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Gettinger, S. et al. Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer. Cancer Discov. 7, 1420–1435 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sato, Y. et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat. Genet. 45, 860–867 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Jiang, T. et al. Tumor neoantigens: from basic research to clinical applications. J. Hematol. Oncol. 12, 93 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Blass, E. & Ott, P. A. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat. Rev. Clin. Oncol. 18, 215–229 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Li, S. et al. Characterization of neoantigen-specific T cells in cancer resistant to immune checkpoint therapies. Proc. Natl Acad. Sci. USA 27, 118 (2021).

  17. Roth, A. et al. PyClone: statistical inference of clonal population structure in cancer. Nat. Methods 11, 396–398 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Deshwar, A. G. et al. PhyloWGS: reconstructing subclonal composition and evolution from whole-genome sequencing of tumors. Genome Biol. 16, 35 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Miller, C. A. et al. SciClone: inferring clonal architecture and tracking the spatial and temporal patterns of tumor evolution. PLoS Comput. Biol. 10, e1003665 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  21. Wang, T. et al. An empirical approach leveraging tumorgrafts to dissect the tumor microenvironment in renal cell carcinoma identifies missing link to prognostic inflammatory factors. Cancer Discov. 8, 1142–1155 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yi, J. S., Cox, M. A. & Zajac, A. J. T-cell exhaustion: characteristics, causes and conversion. Immunology 129, 474–481 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Mehta, A. K., Gracias, D. T. & Croft, M. TNF activity and T cells. Cytokine 101, 14–18 (2018).

    Article  CAS  PubMed  Google Scholar 

  25. Anderson, A. C. Tim-3, a negative regulator of anti-tumor immunity. Curr. Opin. Immunol. 24, 213–216 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Yang, Z.-Z. et al. Expression of LAG-3 defines exhaustion of intratumoral PD-1+ T cells and correlates with poor outcome in follicular lymphoma. Oncotarget 8, 61425–61439 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Zhang, B. et al. Nidogen-1 expression is associated with overall survival and temozolomide sensitivity in low-grade glioma patients. Aging 13, 9085–9107 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Yang, C. et al. Transcriptomic analysis identified ARHGAP family as a novel biomarker associated with tumor-promoting immune infiltration and nanomechanical characteristics in bladder cancer. Front Cell Dev. Biol. 9, 657219 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Feng, Q., Li, L., Li, M. & Wang, X. Immunological classification of gliomas based on immunogenomic profiling. J. Neuroinflammation 17, 360 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Griffin, G. K. et al. Epigenetic silencing by SETDB1 suppresses tumour intrinsic immunogenicity. Nature 595, 309–314 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lyu, X., Li, G. & Qiao, Q. Identification of an immune classification for cervical cancer and integrative analysis of multiomics data. J. Transl. Med. 19, 200 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Chen, X., Su, C., Ren, S., Zhou, C. & Jiang, T. Pan-cancer analysis of KEAP1 mutations as biomarkers for immunotherapy outcomes. Ann. Transl. Med. 8, 141 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Biton, J. et al. TP53, STK11, and EGFR mutations predict tumor immune profile and the response to Anti-PD-1 in lung adenocarcinoma. Clin. Cancer Res. 24, 5710–5723 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Xie, Z. et al. Gene set knowledge discovery with Enrichr. Curr. Protoc. 1, e90 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Voronov, E. & Apte, R. N. Targeting the tumor microenvironment by intervention in interleukin-1 biology. Curr. Pharm. Des. 23, 4893–4905 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Dinarello, C. A. in Textbook of Autoinflammation (eds Hashkes, P. J. et al.) 711–725 (Springer, 2019).

  37. Dang, H. X. et al. ClonEvol: clonal ordering and visualization in cancer sequencing. Ann. Oncol. 28, 3076–3082 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liu, B. et al. Temporal single-cell tracing reveals clonal revival and expansion of precursor exhausted T cells during anti-PD-1 therapy in lung cancer. Nat. Cancer 3, 108–121 (2022).

    Article  CAS  PubMed  Google Scholar 

  39. Barnes, T. A. & Amir, E. HYPE or HOPE: the prognostic value of infiltrating immune cells in cancer. Br. J. Cancer 118, e5 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Melero, I., Rouzaut, A., Motz, G. T. & Coukos, G. T-cell and NK-cell infiltration into solid tumors: a key limiting factor for efficacious cancer immunotherapy. Cancer Discov. 4, 522–526 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zuo, S., Wei, M., Wang, S., Dong, J. & Wei, J. Pan-cancer analysis of immune cell infiltration identifies a prognostic Immune-Cell Characteristic Score (ICCS) in lung adenocarcinoma. Front Immunol. 11, 1218 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Cristescu, R. et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 12, 362 (2018).

  43. Şenbabaoğlu, Y. et al. Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures. Genome Biol. 17, 231 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  44. O’Donnell, T. J., Rubinsteyn, A. & Laserson, U. MHCflurry 2.0: improved pan-allele prediction of MHC class I-presented peptides by incorporating antigen processing. Cell Syst. 11, 418–419 (2020).

    Article  PubMed  Google Scholar 

  45. Kim, S. T. et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat. Med. 24, 1449–1458 (2018).

    Article  CAS  PubMed  Google Scholar 

  46. Anagnostou, V. et al. Evolution of neoantigen landscape during immune checkpoint blockade in non-small cell lung. Cancer Discov. 7, 264–276 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Liu, D. et al. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat. Med. 25, 1916–1927 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  49. McDermott, D. F. et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat. Med. 24, 749–757 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Cristescu, R. et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat. Med. 21, 449–456 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Mitchell, T. J. et al. Timing the landmark events in the evolution of clear cell renal cell cancer: tracerx renal. Cell 173, 611–623.e17 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Salichos, L., Meyerson, W., Warrell, J. & Gerstein, M. Estimating growth patterns and driver effects in tumor evolution from individual samples. Nat. Commun. 11, 732 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lu, T. et al. Overcoming expressional drop-outs in lineage reconstruction from single-cell RNA-sequencing data. Cell Rep. 34, 108589 (2021).

    Article  CAS  PubMed  Google Scholar 

  54. Eden, E., Navon, R., Steinfeld, I., Lipson, D. & Yakhini, Z. GOrilla: a tool for discovery and visualization of enriched GO terms in ranked gene lists. BMC Bioinf. 10, 48 (2009).

    Article  Google Scholar 

  55. Thorsson, V. et al. The immune landscape of cancer. Immunity 48, 812–830.e14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge A. Gouru and A. Passey for helping with proof-reading of the manuscript. This study was supported by the National Institutes of Health (NIH) (grant nos. CCSG 5P30CA142543, 1R01CA258584, 5P30CA142543 and U01AI156189 to T.W. and R01CA234629 to J.Z.), Cancer Prevention Research Institute of Texas (grant nos. CPRIT RP190208 to T.W., CPRIT RP160668 to J.Z., RP160668 to I.W.), University of Texas MDACC (Physician Scientist Program, J.Z. and Lung Cancer Moon Shot, A.R.), Cancer Foundation at the University of Texas MDACC (Institutional Research Grant, A.R.), the Waun Ki Hong Lung Cancer Research Fund (A.R.), Exon 20 Group (A.R.) and Rexanna’s Foundation for Fighting Lung Cancer (A.R.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

T.L. performed all analyses. T.W. supervised the study. S.M.H., J.V.H., P.A.F., I.W., A.R. and J.Z. provided the in-house sequencing data. Y.H. and Y.W. provided critical input in the science and also the writing. T.L., S.P. and T.W. wrote the paper.

Corresponding author

Correspondence to Tao Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Madhura Mukhopadhyay, in collaboration with the Nature Methods team.

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 Applying netie on the simulation data.

Two more simulation datasets (a) and (b,c). (a) Simulation setting of the second dataset and netie’s inference results. (b) Simulation setting of the third dataset. (c) Netie’s inference results for the third dataset. The same simulation and analysis procedures, as in Fig. 1e–g, were carried out.

Source data

Extended Data Fig. 2 Immune selection pressure variations correlate with the genotypes of the tumor and tumor clones.

(a) The top genes with smallest Wilcoxon test P values comparing the immune pressure variations in the tumor clones with and without mutations in each gene. (b) Boxplots of the immune pressure variation (ac) in the tumor clones with and without mutations in SETDB1 and FN1. (c) Enriched GO terms of the genes with Wilcoxon test P value<0.05. For boxplot in (b), box boundaries represent interquantile ranges, whiskers extend to the most extreme data point which is no more than 1.5 times the interquartile range, and the line in the middle of the box represents the median.

Source data

Extended Data Fig. 3 The immune selection pressure scores of the shared and the private tumor clones of the other MDACC lung cancer patients with multi-region sampling.

The center of the error bar represents the inferred immune pressure variation (ac), and the error bars represent 95% confidence intervals.

Source data

Extended Data Fig. 4 Further validating the implication of Netie classifications for prognosis of patients (a,b) Multivariate analysis testing the association between TIGEP and overall survival in INisp and DEisp patients.

(a) INisp patients, (b) DEisp patients. The association between TIGEP and overall survival is tested in a CoxPH model, with multi-variate adjustment for pathological stage, gender, age, and tumor types.

Source data

Extended Data Fig. 5 Analyses as conducted in Fig. 4a, but with the clonality inference conducted by PhyloWGS and SciClone (a), or with the neoantigens predicted by MHCflurry (b).

The TCGA LUAD cohort was employed as an example.

Source data

Supplementary information

Supplementary Information

Note 1: Detailed description of the netie model. Note 2: Additional analyses and discussions involving the netie model.

Reporting Summary

Supplementary Software

A zipped version of the netie software for documentation purpose.

Supplementary Tables

Table 1: Wilcoxon test P values of the genes investigated in Extended Data Fig. 2. Table 2: The effect of the tuning parameters on the performance of the model. Table 3: Detailed characteristics, including age, gender and smoking status, of the in-house MDACC patients with lung cancer cohort. Table 4: Accession methods of the data used in our study.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Rights and permissions

Springer Nature or its licensor 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

Lu, T., Park, S., Han, Y. et al. Netie: inferring the evolution of neoantigen–T cell interactions in tumors. Nat Methods 19, 1480–1489 (2022). https://doi.org/10.1038/s41592-022-01644-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-022-01644-7

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