Abstract
Despite the transformative impact of cancer immunotherapy, the need for improved patient stratification remains critical due to suboptimal response rates. While neoantigens are central to anti-tumor immunity, current metrics, such as tumor mutation burden (TMB), are limited by their neglect of immunogenicity and tumor heterogeneity. Here we present NeoPrecis, a computational framework designed to improve immunotherapy response prediction by refining neoantigen characterization across MHC-I and MHC-II pathways and by integrating tumor clonality information. NeoPrecis features an interpretable T-cell-recognition model that reveals the critical influence of MHC molecules on TCR recognition beyond mere antigen presentation. Benefit HLA alleles, identified through model-driven contribution analysis, exhibit significant predictive power for patient outcomes in immune checkpoint inhibitor treatment (melanoma: p-value = 0.04; NSCLC: p-value = 0.01). NeoPrecis, via its clonality-aware neoantigen landscape feature, improves immunotherapy response prediction in tumor types with varying prevalence of neoantigens, including heterogeneous NSCLC, which retains more subclonal neoantigens due to lower immunoediting pressure. We thus propose NeoPrecis as a comprehensive evaluative framework for neoantigen assessment by incorporating both immunogenicity and tumor clonality, offering insights into the link between the collective quality of neoantigen landscapes and immunotherapy response.
Similar content being viewed by others
Data availability
The processed data generated in this study are provided in the Supplementary Data files, including cross-reactive peptide triplets, CEDAR immunogenicity data, processed NCI gastrointestinal cancer cohort data, and the ICI cohort metadata with analysis results. Publicly available datasets analyzed in this study were obtained from the Immune Epitope Database (IEDB, https://www.iedb.org), VDJdb (https://vdjdb.cdr3.net), and the Cancer Epitope Database and Analysis Resource (CEDAR, https://cedar.iedb.org). The mutation-centric immunogenicity data (NCI dataset) were obtained from the supplementary materials of Parkhurst et al. (https://doi.org/10.1158/2159-8290.CD-18-1494). Clinical and genomic data from the reanalyzed ICI cohorts were obtained with the following accession numbers: Hugo et al. (SRP090294 [https://trace.ncbi.nlm.nih.gov/Traces/study/?acc=SRP090294], SRP067938), Van Allen et al. (SRP011540 [https://trace.ncbi.nlm.nih.gov/Traces/study/?acc=SRP011540]), Snyder et al. (SRP072934 [https://trace.ncbi.nlm.nih.gov/Traces/study/?acc=SRP072934]), Riaz et al. (SRP094781 [https://trace.ncbi.nlm.nih.gov/Traces/study/?acc=SRP094781]), Liu et al. (SRP011540 [https://trace.ncbi.nlm.nih.gov/Traces/study/?acc=SRP011540]), Ravi et al. (SRP413932 [https://trace.ncbi.nlm.nih.gov/Traces/study/?acc=SRP413932]), Anagnostou et al. (SRP238904 [https://trace.ncbi.nlm.nih.gov/Traces/study/?acc=SRP238904]), Rizvi et al. (SRP064805 [https://trace.ncbi.nlm.nih.gov/Traces/study/?acc=SRP064805]).
Code availability
Codes for conducting immunogenicity prediction and neoantigen landscape evaluation are deposited at both GitHub (https://github.com/cartercompbio/NeoPrecis) and Zenodo (https://doi.org/10.5281/zenodo.17959604).
References
Schumacher, T. N. & Schreiber, R. D. Neoantigens in cancer immunotherapy. Science 348, 69–74 (2015).
Saxena, M., Van Der Burg, S. H., Melief, C. J. M. & Bhardwaj, N. Therapeutic cancer vaccines. Nat. Rev. Cancer 21, 360–378 (2021).
Ott, P. A. et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 547, 217–221 (2017).
Sahin, U. et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 547, 222–226 (2017).
Weber, J. S. et al. Individualised neoantigen therapy mRNA-4157 (V940) plus pembrolizumab versus pembrolizumab monotherapy in resected melanoma (KEYNOTE-942): a randomised, phase 2b study. Lancet 403, 632–644 (2024).
Ott, P. A. et al. A Phase Ib Trial of Personalized Neoantigen Therapy Plus Anti-PD-1 in patients with advanced melanoma, non-small cell lung cancer, or bladder cancer. Cell 183, 347–362.e24 (2020).
Keskin, D. B. et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature 565, 234–239 (2019).
Hilf, N. et al. Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature 565, 240–245 (2019).
Rojas, L. A. et al. Personalized RNA neoantigen vaccines stimulate T cells in pancreatic cancer. Nature 618, 144–150 (2023).
Yarchoan, M. et al. Personalized neoantigen vaccine and pembrolizumab in advanced hepatocellular carcinoma: a phase 1/2 trial. Nat. Med. 30, 1044–1053 (2024).
Braun, D. A. et al. A neoantigen vaccine generates antitumour immunity in renal cell carcinoma. Nature 639, 474–482 (2025).
Hodi, F. S. et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N. Engl. J. Med. 363, 711–723 (2010).
Robert, C. et al. Ipilimumab plus Dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 364, 2517–2526 (2011).
Borghaei, H. et al. Nivolumab versus Docetaxel in advanced nonsquamous non–small-cell lung cancer. N. Engl. J. Med. 373, 1627–1639 (2015).
Brahmer, J. et al. Nivolumab versus Docetaxel in advanced squamous-cell non–small-cell lung cancer. N. Engl. J. Med. 373, 123–135 (2015).
Yarchoan, M., Hopkins, A. & Jaffee, E. M. Tumor mutational burden and response rate to PD-1 inhibition. N. Engl. J. Med. 377, 2500–2501 (2017).
Wu, Y. et al. The predictive value of tumor mutation burden on efficacy of immune checkpoint inhibitors in cancers: a systematic review and meta-analysis. Front. Oncol. 9, 1161 (2019).
Larkin, J. et al. Combined Nivolumab and Ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).
Robert, C. et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 372, 2521–2532 (2015).
Wang, D. Y. et al. Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis. JAMA Oncol. 4, 1721 (2018).
Chan, T. A. et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann. Oncol. 30, 44–56 (2019).
Zou, X. et al. Prognostic Value of Neoantigen load in immune checkpoint inhibitor therapy for cancer. Front. Immunol. 12, 689076 (2021).
Wells, D. K. et al. Key Parameters of tumor epitope immunogenicity revealed through a Consortium approach improve neoantigen prediction. Cell 183, 818–834.e13 (2020).
Castro, A., Zanetti, M. & Carter, H. Neoantigen controversies. Annu. Rev. Biomed. Data Sci. 4, 227–253 (2021).
Duan, F. et al. Genomic and bioinformatic profiling of mutational neoepitopes reveals new rules to predict anticancer immunogenicity. J. Exp. Med. 211, 2231–2248 (2014).
Łuksza, M. et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 551, 517–520 (2017).
Wan, Y. R., Koşaloğlu-Yalçın, Z., Peters, B. & Nielsen, M. A large-scale study of peptide features defining immunogenicity of cancer neo-epitopes. NAR Cancer 6, zcae002 (2024).
Kim, J. Y. et al. MHC II immunogenicity shapes the neoepitope landscape in human tumors. Nat. Genet. 55, 221–231 (2023).
Schmidt, J. et al. Prediction of neo-epitope immunogenicity reveals TCR recognition determinants and provides insight into immunoediting. Cell Rep. Med. 2, 100194 (2021).
Gfeller, D. et al. Improved predictions of antigen presentation and TCR recognition with MixMHCpred2.2 and PRIME2.0 reveal potent SARS-CoV-2 CD8+ T-cell epitopes. Cell Syst. 14, 72–83.e5 (2023).
Łuksza, M. et al. Neoantigen quality predicts immunoediting in survivors of pancreatic cancer. Nature 606, 389–395 (2022).
Jung, D. & Alt, F. W. Unraveling V(D)J recombination: insights into gene regulation. Cell 116, 299–311 (2004).
Klein, L., Kyewski, B., Allen, P. M. & Hogquist, K. A. Positive and negative selection of the T cell repertoire: what thymocytes see (and don’t see). Nat. Rev. Immunol. 14, 377–391 (2014).
McGranahan, N. et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351, 1463–1469 (2016).
Ravi, A. et al. Genomic and transcriptomic analysis of checkpoint blockade response in advanced non-small cell lung cancer. Nat. Genet. 55, 807–819 (2023).
McDonald, K.-A. et al. Tumor heterogeneity correlates with less immune response and worse survival in breast cancer patients. Ann. Surg. Oncol. 26, 2191–2199 (2019).
Wolf, Y. & Samuels, Y. Intratumor heterogeneity and antitumor immunity shape one another bidirectionally. Clin. Cancer Res. 28, 2994–3001 (2022).
Andor, N. et al. Pan-cancer analysis of the extent and consequences of intratumor heterogeneity. Nat. Med. 22, 105–113 (2016).
Jamal-Hanjani, M. et al. Tracking the evolution of non–small-cell lung cancer. N. Engl. J. Med. 376, 2109–2121 (2017).
Wolf, Y. et al. UVB-induced tumor heterogeneity diminishes immune response in melanoma. Cell 179, 219–235.e21 (2019).
Gupta, R. G., Li, F., Roszik, J. & Lizée, G. Exploiting tumor neoantigens to target cancer evolution: current challenges and promising therapeutic approaches. Cancer Discov. 11, 1024–1039 (2021).
Bawden, E. G. et al. CD4+ T cell immunity against cutaneous melanoma encompasses multifaceted MHC II–dependent responses. Sci. Immunol. 9, eadi9517 (2024).
Espinosa-Carrasco, G. et al. Intratumoral immune triads are required for immunotherapy-mediated elimination of solid tumors. Cancer Cell 42, 1202–1216.e8 (2024).
Pyke, R. M. et al. Evolutionary pressure against MHC Class II binding cancer mutations. Cell 175, 416–428.e13 (2018).
Alspach, E. et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature 574, 696–701 (2019).
Sears, T. J. et al. Integrated germline and somatic features reveal divergent immune pathways driving response to immune checkpoint blockade. Cancer Immunol. Res. 12, 1780–1795 (2024).
Sewell, A. K. Why must T cells be cross-reactive?. Nat. Rev. Immunol. 12, 669–677 (2012).
Vita, R. et al. The Immune Epitope Database (IEDB): 2018 update. Nucleic Acids Res. 47, D339–D343 (2019).
Goncharov, M. et al. VDJdb in the pandemic era: a compendium of T cell receptors specific for SARS-CoV-2. Nat. Methods 19, 1017–1019 (2022).
Koşaloğlu-Yalçın, Z. et al. The Cancer Epitope Database and Analysis Resource (CEDAR). Nucleic Acids Res. 51, D845–D852 (2023).
Kim, Y., Sidney, J., Pinilla, C., Sette, A. & Peters, B. Derivation of an amino acid similarity matrix for peptide:MHC binding and its application as a Bayesian prior. BMC Bioinforma. 10, 394 (2009).
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, 449–454 (2020).
Nilsson, J. B. et al. Accurate prediction of HLA class II antigen presentation across all loci using tailored data acquisition and refined machine learning. Sci. Adv. 9, eadj6367 (2023).
Parkhurst, M. R. et al. Unique Neoantigens Arise from Somatic Mutations in Patients with Gastrointestinal Cancers. Cancer Discov. 9, 1022–1035 (2019).
Alban, T. J. et al. Neoantigen immunogenicity landscapes and evolution of tumor ecosystems during immunotherapy with nivolumab. Nat. Med. 30, 3209–3222 (2024).
Chowell, D. et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 359, 582–587 (2018).
Cummings, A. L. et al. Mutational landscape influences immunotherapy outcomes among patients with non-small-cell lung cancer with human leukocyte antigen supertype B44. Nat. Cancer 1, 1167–1175 (2020).
Castro, A. et al. Elevated neoantigen levels in tumors with somatic mutations in the HLA-A, HLA-B, HLA-C and B2M genes. BMC Med. Genomics 12, 107 (2019).
Marty, R. et al. MHC-I genotype restricts the oncogenic mutational landscape. Cell 171, 1272–1283.e15 (2017).
Hugo, W. et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 165, 35–44 (2016).
Snyder, A. et al. Genetic basis for clinical response to CTLA-4 Blockade in Melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).
Van Allen, E. M. et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 350, 207–211 (2015).
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).
Riaz, N. et al. Tumor and microenvironment evolution during immunotherapy with Nivolumab. Cell 171, 934–949.e16 (2017).
Rizvi, N. A. et al. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science 348, 124–128 (2015).
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).
Roth, A. et al. PyClone: statistical inference of clonal population structure in cancer. Nat. Methods 11, 396–398 (2014).
Lu, T. et al. Tumor neoantigenicity assessment with CSiN score incorporates clonality and immunogenicity to predict immunotherapy outcomes. Sci. Immunol. 5, eaaz3199 (2020).
Su, X. et al. Construction and validation of an immunoediting-based optimized neoantigen load (ioTNL) model to predict the response and prognosis of immune checkpoint therapy in various cancers. Aging 14, 4586–4605 (2022).
Dai, L. et al. The effect of smoking status on efficacy of immune checkpoint inhibitors in metastatic non-small cell lung cancer: A systematic review and meta-analysis. eClinicalMedicine 38, 100990 (2021).
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).
Xie, N. et al. Neoantigens: promising targets for cancer therapy. Signal Transduct. Target. Ther. 8, 1–38 (2023).
Wu, P. et al. Mechano-regulation of Peptide-MHC Class I conformations determines TCR antigen recognition. Mol. Cell 73, 1015–1027.e7 (2019).
Cole, D. K. et al. Modification of MHC anchor residues generates heteroclitic peptides that alter TCR B inding and T cell recognition. J. Immunol. 185, 2600–2610 (2010).
Smith, A. R. et al. Structurally silent peptide anchor modifications allosterically modulate T cell recognition in a receptor-dependent manner. Proc. Natl. Acad. Sci. 118, e2018125118 (2021).
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.e4 (2019).
Magen, A. et al. Intratumoral dendritic cell–CD4+ T helper cell niches enable CD8+ T cell differentiation following PD-1 blockade in hepatocellular carcinoma. Nat. Med. 29, 1389–1399 (2023).
The TRACERx consortium et al. Neoantigen-directed immune escape in lung cancer evolution. Nature 567, 479–485 (2019).
Niknafs, N. et al. Persistent mutation burden drives sustained anti-tumor immune responses. Nat. Med. 29, 440–449 (2023).
Turajlic, S. et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol. 18, 1009–1021 (2017).
Carson, R. T., Vignali, K. M., Woodland, D. L. & Vignali, D. A. A. T cell receptor recognition of MHC Class II–bound peptide flanking residues enhances immunogenicity and results in altered TCR V region usage. Immunity 7, 387–399 (1997).
Holland, C. J., Cole, D. K. & Godkin, A. Re-Directing CD4+ T cell responses with the flanking residues of MHC Class II-bound peptides: the core is not enough. Front. Immunol. 4, 172 (2013).
Sayaman, R. W. et al. Germline genetic contribution to the immune landscape of cancer. Immunity 54, 367–386.e8 (2021).
Pagadala, M. et al. Germline modifiers of the tumor immune microenvironment implicate drivers of cancer risk and immunotherapy response. Nat. Commun. 14, 2744 (2023).
Loshchilov, I. & Hutter, F. Decoupled Weight Decay Regularization. in Proc. Int. Conf. Learn. Repr. (ICLR) (2019).
Khanna, A. et al. Bam-readcount - rapid generation of basepair-resolution sequence metrics. J. Open Source Softw. 7, 3722 (2022).
Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinforma. 12, 323 (2011).
Garcia, M. et al. Sarek: A portable workflow for whole-genome sequencing analysis of germline and somatic variants. F1000Research 9, 63 (2020).
Hanssen, F. et al. Scalable and efficient DNA sequencing analysis on different compute infrastructures aiding variant discovery. NAR Genomics Bioinforma. 6, lqae031 (2024).
Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
Kawaguchi, S., Higasa, K., Shimizu, M., Yamada, R. & Matsuda, F. HLA-HD: An accurate HLA typing algorithm for next-generation sequencing data. Hum. Mutat. 38, 788–797 (2017).
Acknowledgements
This work was funded by the Mark Foundation Emerging Leader Award #18-022-ELA, NCI grant R01CA269919, and support from NCI grant U24CA248138 to H. Carter. Computational resources were supported by an infrastructure grant 2P41GM103504-11. The ICI cohorts were collected from several published studies. The melanoma cohort from Hugo and colleagues was obtained from the Sequence Read Archive (SRA) under accessions SRP090294 and SRP067938. The melanoma cohorts from Van Allen et al. and Liu et al. were obtained from dbGaP under accession phs000452, supported by the National Human Genome Research Institute (NHGRI) Large Scale Sequencing Program, Grant U54 HG003067 to the Broad Institute (PI, Lander). The melanoma cohort from Riaz and colleagues was obtained from SRA under accession SRP094781. The melanoma cohort from Snyder and colleagues was obtained from dbGaP under accession phs001041; we thank Martin Miller at Memorial Sloan Kettering Cancer Center (MSKCC) for his assistance with the NetMHC server, Agnes Viale and Kety Huberman at the MSKCC Genomics Core, Annamalai Selvakumar and Alice Yeh at the MSKCC HLA typing laboratory for their technical assistance, and John Khoury for assistance in chart review. The NSCLC cohort from Rizvi and colleagues was obtained from dbGaP under accession phs000980. We thank the members of the Thoracic Oncology Service and the Chan and Wolchok labs at MSKCC for helpful discussions. We thank the Immune Monitoring Core at MSKCC, including L. Caro, R. Ramsawak, and Z. Mu, for exceptional support with processing and banking peripheral blood lymphocytes. We thank P. Worrell and E. Brzostowski for help in identifying tumor specimens for analysis. We thank A. Viale for superb technical assistance. We thank D. Philips, M. van Buuren, and M. Toebes for help performing the combinatorial coding screens. The data presented in this paper are tabulated in the main paper and in the supplementary materials. This work was supported by the Geoffrey Beene Cancer Research Center (MDH, NAR, TAC, JDW, AS), the Society for Memorial Sloan Kettering Cancer Center (MDH), Lung Cancer Research Foundation (WL), Frederick Adler Chair Fund (TAC), The One Ball Matt Memorial Golf Tournament (EBG), Queen Wilhelmina Cancer Research Award (TNS), The STARR Foundation (TAC, JDW), the Ludwig Trust (JDW), and a Stand Up To Cancer-Cancer Research Institute Cancer Immunology Translational Cancer Research Grant (JDW, TNS, TAC). Stand Up To Cancer is a program of the Entertainment Industry Foundation administered by the American Association for Cancer Research. The NSCLC cohort from Anagnostou and colleagues was obtained from dbGaP under accession phs001940, supported in part by US National Institutes of Health grant CA121113. The NSCLC cohort from Ravi and colleagues was obtained from dbGaP under accession phs002822. We express our deep gratitude to the patients and families whose participation enabled this study. We further thank the respective sequencing centers at Yale University, Johns Hopkins University, and the Broad Institute of MIT and Harvard for processing the whole exome and RNA-seq data presented here. Funding for this study was provided by a Stand Up To Cancer - American Cancer Society Lung Cancer Dream Team Translational Research Grant (Grant Number: SU2C-AACR-DT17-15). Stand Up to Cancer is a program of the Entertainment Industry Foundation. Research grants are administered by the American Association for Cancer Research, the scientific partner of SU2C. This work was additionally supported by The Mark Foundation for Cancer Research (Grant Number: 19-029-MIA) Expanding Therapeutic Options for Lung Cancer (EXTOL) project.
Author information
Authors and Affiliations
Contributions
Conceptualization: K.H.L., T.J.S., M.Z., H.C. Methodology: K.H.L. Investigation: K.H.L., T.J.S. Visualization: K.H.L. Funding acquisition: H.C. Project administration: H.C. Supervision: M.Z., H.C. Writing – original draft: K.H.L. Writing – review & editing: T.J.S., M.Z., H.C.
Corresponding author
Ethics declarations
Competing interests
MZ is an advisor to the board of Invectys Inc. All other authors declare no competing interests.
Peer review
Peer review information
Nature Communications thanks the anonymous reviewer(s) for their contribution to the peer review of this work. A peer review file is available.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Lee, KH., Sears, T.J., Zanetti, M. et al. NeoPrecis: enhancing immunotherapy response prediction through integration of qualified immunogenicity and clonality-aware neoantigen landscapes. Nat Commun (2026). https://doi.org/10.1038/s41467-026-68651-6
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41467-026-68651-6


